NV

Novo Nordisk

- NYSE:NVO
Last Updated 2024-04-23

Patent Grants Data

Patents granted to organizations.
Ticker Symbol Entity Name Publication Date Filing Date Patent ID Invention Title Abstract Patent Number Claims Number of Claims Description Application Number Assignee Country Kind Code Kind Code Description url Classification Code Length of Grant Date Added Date Updated Company Name Sector Industry
nyse:nvo Novo Nordisk Apr 26th, 2022 12:00AM Nov 20th, 2014 12:00AM https://www.uspto.gov?id=US11311678-20220426 Rotary sensor assembly with space efficient design A sensor assembly comprising a first rotary sensor part having a plurality of individual electrically conducting code segments arranged in a circumferential pattern, and a plurality of electrically conducting reference segments between the code segments, and a second rotary sensor part arranged rotationally relative to the first part a plurality of contact structures, each contact structure being arranged to be in contact with either a code segment or a reference segment depending on the rotational position between the first and second rotary sensor part. The contact structures are configured to engage and connect to different sensor segments as the first and second rotary sensor part rotate relative to each, the created connections being indicative of a rotational position between the first and second rotary sensor part. For a given rotational position, at least one contact structure engages a code segment and at least one contact structure engages a reference segment. 11311678 1. A sensor assembly comprising: a first rotary sensor part comprising a surface having: a circumferentially arranged reference track, a plurality of individual electrically conducting code segments arranged in a pattern and aligned circumferentially with the reference track but electrically isolated therefrom, a plurality of electrically conducting reference segments, the reference segments being connected to electrically form a single combined reference segment to thereby form a single conductive structure, a second rotary sensor part arranged rotationally relative to the first rotary sensor part, comprising: a plurality of contact structures, each contact structure being arranged to be in contact with either a code segment or a reference segment depending on a rotational position between the first and second rotary sensor part, wherein the contact structures are configured to engage and connect to different sensor segments as the first and second rotary sensor part rotate relative to each other, the created connections being indicative of the rotational position between the first and second rotary sensor part, and for a given rotational position, at least one contact structure engages a code segment and at least one contact structure engages a reference segment, and wherein, the single conductive structure comprises: narrow strips of plating surrounding radial sides of the code segments, ground segments connected on only one side of the code segments, or connections formed on opposed sides of the second rotary sensor part in the form of a metallic disc member. 2. The sensor assembly as in claim 1, wherein the metallic disc member further comprises a plurality of integrally formed flexible arms forming the contact structures. 3. The sensor assembly as in claim 1, wherein the reference segments and the code segments are formed on the surface of the first rotary sensor part by a plating process. 4. The sensor assembly as in claim 1, wherein the reference segments, the electrical connections there between, and the code segments are formed on the surface of the first rotary sensor part by a plating process. 4 CROSS-REFERENCE TO RELATED APPLICATIONS This application is a 35 U.S.C. § 371 National Stage application of International Application PCT/EP2014/075179 (published as WO2015/075134), filed Nov. 20, 2014, which claims priority to European Patent Application 13193882.1, filed Nov. 21, 2013; this application claims priority under 35 U.S.C. § 119 to U.S. Provisional Application 61/907,488; filed Nov. 22, 2013. The present invention relates to devices, assemblies and systems adapted for capturing information in respect of rotational movement. In a specific aspect the invention addresses issues relating to electronic dose data capturing in and for a drug delivery device. BACKGROUND OF THE INVENTION In the disclosure of the present invention reference is mostly made to the treatment of diabetes by delivery of insulin using a drug delivery device, however, this is only an exemplary use of the present invention. Drug delivery devices have greatly improved the lives of patients who must self-administer drugs and biological agents. Drug delivery devices may take many forms, including simple disposable devices that are little more than an ampoule with an injection means or they may be durable devices adapted to be used with pre-filled cartridges. Regardless of their form and type, they have proven to be great aids in assisting patients to self-administer injectable drugs and biological agents. They also greatly assist care givers in administering injectable medicines to those incapable of performing self-injections. Performing the necessary insulin injection at the right time and in the right size is essential for managing diabetes, i.e. compliance with the specified insulin regimen is important. In order to make it possible for medical personnel to determine the effectiveness of a prescribed dosage pattern, diabetes patients are encouraged to keep a log of the size and time of each injection. However, such logs are normally kept in handwritten notebooks, from the logged information may not be easily uploaded to a computer for data processing. Furthermore, as only events, which are noted by the patient, are logged, the note book system requires that the patient remembers to log each injection, if the logged information is to have any value in the treatment of the patient's disease. A missing or erroneous record in the log results in a misleading picture of the injection history and thus a misleading basis for the medical personnel's decision making with respect to future medication. Accordingly, it may be desirable to automate the logging of ejection information from medication delivery systems. Correspondingly, a number of drug delivery devices with a dose monitoring/acquisition feature has been provided or suggested, see e.g. in US 2009/0318865, WO 2010/052275 and U.S. Pat. No. 7,008,399. However, most devices of today are without it. When providing a drug delivery with a monitoring feature, a rotary sensor may be incorporated to detect relative movement between components of the drug delivery mechanism, the movement being indicative of a set and/or expelled dose of drug. A traditional rotary sensor is discloses in e.g. WO 96/19872 comprising a code disc with code segments and a reference track arranged in two ring-shaped structures as well as a contact structure for each ring structure. Having regard to the above, it is an object of the present invention to provide a drug delivery device as well as components and assemblies therefore which in a safe, user-friendly, cost-effective and reliable way allows detection and storage of dose data related to use of a drug delivery device. It is a further object to provide such components and assemblies which could be used also in other applications having the same types of input. DISCLOSURE OF THE INVENTION In the disclosure of the present invention, embodiments and aspects will be described which will address one or more of the above objects or which will address objects apparent from the below disclosure as well as from the description of exemplary embodiments. Thus, in a first aspect of the invention a sensor assembly is provided comprising a first rotary sensor part and a second rotary sensor part. The first rotary sensor part comprises a surface having a circumferentially arranged reference track, and a plurality of individual electrically conducting code segments arranged in a pattern. Each code segment is arranged in the reference track but electrically isolated therefrom, whereby a plurality of electrically conducting reference segments are formed between the code segments. The second rotary sensor part is arranged rotationally relative to the first part and comprises a plurality of contact structures, each contact structure being arranged to be in contact with either a code segment or a reference segment depending on the rotational position between the first and second rotary sensor part. The contact structures are configured to engage and connect to different sensor segments as the first and second rotary sensor part rotate relative to each, the created connections being indicative of a rotational position between the first and second rotary sensor part. For a given rotational position, at least one contact structure engages a code segment and at least one contact structure engages a reference segment. The term “given rotational position” is meant to cover normal operational states in which a given switch contact is positioned in contact with a single sensor or reference segment, i.e. outside the gaps formed between two neighbouring sensor segments. By such an arrangement one or more of the following can be achieved: Minimizing physical volume of a positional sensor by having both reference (ground) connections and code information on the same track, and by using the same contact structures for both code and reference connection. Simplification of sensor segments part by removal of dedicated reference connection and using contact structures dynamically for both code and reference connections. By the above features a sensor design is provided which in itself allows a smaller and simpler sensor to be manufactured in a cost-effective way, but also provides a higher degree of freedom of design when incorporating the sensor system in a given mechanical construction such as a drug delivery device. The second rotary sensor part may be in the form of a metallic disc member comprising a plurality of integrally formed flexible arms forming the contact structures. The reference segments may be electrically connected to each other to thereby form a single conductive structure, this allowing a simple design with only a single electric connection between the interconnected reference segments on the first rotary sensor part and the associated electronic circuitry of the sensor assembly. For example, the code segments may be arranged in the reference track as “islands” surrounded by conducting portions of the reference track on both side, or alternatively with conducting portions on only one side. As a further alternative the reference segments may be connected with conductors arranged on the opposed surface. The reference segments and the code segments may be formed on the first rotary sensor part surface by a plating process, especially, the reference segments, the electrical connections there between, and the code segments may be formed on the first rotary sensor part surface by a plating process. The first rotary sensor part may further be provided with one or more circumferentially arranged switch segments, the second rotary sensor part comprising one or more contact structures providing an axial switch contact having a connected position in which the switch contact is in contact with a sensor switch segment and a dis-connected position in which the switch contact is not in contact with a sensor switch segment. The sensor assembly may further comprise electronic circuitry adapted to determine a rotational position between the first and second rotary sensor part based on a given pattern of created connections. In an exemplary application the rotary sensor assembly can be incorporated cost-effectively and reliably in a drug delivery device comprising a rotational member which rotates corresponding to a set and/or expelled dose. Correspondingly, in an exemplary embodiment a drug delivery device is provided comprising a sensor assembly as described above, the drug delivery device further comprising a housing, a drug-filled cartridge or means for receiving a drug-filled cartridge, and drug expelling means, the cartridge comprising an axially displaceable piston and a distal outlet portion. The drug expelling means comprises dose setting means allowing a user to set a dose of drug to be expelled, an axially displaceable piston rod adapted to move the piston of a cartridge in a distal direction to thereby expel drug from the cartridge, a rotational member adapted to rotate corresponding to a set and/or expelled dose, and an axially moveable actuation member adapted to actuate the drug expelling means to thereby expel the set dose of drug. In such an arrangement the first and second rotary sensor parts rotate relative to each other during setting and/or expelling of a dose of drug. The first portion of the sensor assembly may be mounted to and rotate with the rotational member, the first portion comprising electronic circuitry adapted to estimate an amount of expelled drug based on detection of rotational movement between the first and second portions corresponding to a set and/or expelled dose. The rotational member may be adapted to move axially between an initial and an actuated position, the first portion of the sensor assembly being mounted to move axially with the rotational member. Also the second portion may be mounted to move axially with the rotational member. For such a design the inherent movements of the rotational member can be detected simple and effectively by the sensor assembly when provided with axial switch means. The electronic circuitry may be provided with logging means adapted to create a log for dose amounts of drug expelled from a cartridge by the drug expelling means, the dose amounts being calculated based on relative rotation between the first and second rotary sensor parts during setting and/or expelling of a dose of drug. The electronic circuitry may be provided with transmitter means adapted to transmit stored data to an external receiver. Alternatively or in addition, the first portion may comprise a display which may be controlled to be turned off during rotation of the first portion. In a specific embodiment of the invention a drug delivery device is provided comprising a housing, a drug-filled cartridge or means for receiving a drug-filled cartridge, and drug expelling means, the cartridge comprising an axially displaceable piston and a distal outlet portion. The drug expelling means comprises dose setting means allowing a user to set a dose of drug to be expelled, an axially displaceable piston rod adapted to move the piston of a cartridge in a distal direction to thereby expel drug from the cartridge, a rotational member adapted to rotate corresponding to a set and/or expelled dose, and an axially moveable actuation member adapted to actuate the drug expelling means to thereby expel the set dose of drug. The drug delivery device further comprises sensor means adapted to detect a set and/or an expelled dose, comprising (i) a first portion comprising a first rotary sensor part, the first rotary sensor part comprising a surface with a plurality of sensor and reference segments forming a combined code and reference track as described above, and (ii) a second portion comprising a second rotary sensor part arranged rotationally relative to the first portion, the second rotary sensor part comprising a plurality of combined code and reference contact structures as described above. One of the contact structures may comprise an axial switch contact having a connected position in which the switch contact is in contact with a sensor area and a dis-connected position in which the switch contact is not in contact with a sensor area. The first and second rotary sensor parts rotate relative to each other during setting and/or expelling of a dose of drug, and the axial switch is actuated between its two positions when the actuation member is moved axially. The electronic circuitry may be adapted to determine a rotational position between the first and second portions based on a given pattern of created connections, the electronic circuitry comprising logging means adapted to create a log for dose amounts of drug expelled from a cartridge by the drug expelling means, wherein the dose amounts are calculated based on relative rotation between the first and second rotary sensor parts during setting and/or expelling of a dose of drug. The drug delivery device may comprise additional features as described above. In a second aspect of the invention a sensor assembly, e.g. a rotary encoder disc assembly, is provided comprising an electrical connector structure comprising at least one terminal, a non-conducting moulded matrix member, and at least one conducting structure having a contact surface. A portion of the electrical connector structure comprising at least one terminal is in-moulded in the matrix member, at least one conducting structure is in-moulded in the matrix member with at least a portion of the contact surface being free, and at least one in-moulded conducting structure is connected to an in-moulded terminal. A sensor member such as a rotary encoder disc comprises a disc with a code pattern as well as a connector structure allowing the conductive areas of the code pattern to be connected to a PCB, e.g. using a traditional male-female connector. By in-moulding the terminals of an electrical connector structure, e.g. a flex-connector, in a non-conductive disc matrix and creating the conductive code areas by in-moulding, a compact and reliable sensor assembly is provided. In a most simple embodiment a single conductive structure providing a single contact surface is in-moulded in direct contact with a single terminal. The at least one conducting structure may be formed from a conducting polymer, the matrix member and the at least one conducting structure being formed by two-shot moulding. For a more complex sensor assembly it may further comprise at least one conducting connector structure connecting a conducting structure and a terminal, wherein at least one conducting connector structure is in-moulded in the matrix member, e.g. during the two-shot moulding. In an exemplary embodiment the matrix member comprises a first surface with at least one cavity with an in-moulded conducting structure, and a second opposed surface with at least one cavity with an in-moulded conducting connector structure, the conducting structure and the conducting connector structure being connected to each other through an opening formed in the matrix member, the conducting connector structure being connected to a terminal. In a third aspect of the invention a rotary sensor member adapted to rotate corresponding to an axis of rotation is provided, comprising at least two surfaces, wherein the first surface comprises a plurality of individual electrically conducting encoder sensor segments arranged in a circumferential pattern, and the second surface comprises a further circumferential track in the form of a ground track or a second code track having a second plurality of individual electrically conducting encoder sensor segments arranged in a second circumferential pattern. By arranging the two circumferential structures on two different surfaces of a rotary sensor member, e.g. opposed sides of a disc, the desired functionality can be achieved with a sensor member having a small diameter. Further, by separating the two circumferential structures on two surfaces the risk of short circuit is reduced. In an exemplary embodiment the rotary sensor member may comprise at least two surfaces arranged axially offset and perpendicularly relative to the axis of rotation, e.g. in the form of the two opposed surfaces on a disc member. The surfaces will typically be generally planar. The rotary sensor member may comprise at least one generally cylindrical surface, each generally cylindrical surface, when more than one generally cylindrical surface is provided, being arranged radially offset relative to the axis of rotation. The rotary sensor member may comprise at least three surfaces and at least one further circumferential track in the form of a (further) ground track or a further code track. In an exemplary embodiment a sensor assembly is provided comprising a rotary sensor member as described above, further comprising a plurality of switch contact structures, each switch contact structure being arranged for sliding rotational engagement with a circumferential pattern or a circumferential track. As used herein, the term “drug” is meant to encompass any flowable medicine formulation capable of being passed through a delivery means such as a cannula or hollow needle in a controlled manner, such as a liquid, solution, gel or fine suspension, and containing one or more drug agents. The drug may be a single drug compound or a premixed or co-formulated multiple drug compounds drug agent from a single reservoir. Representative drugs include pharmaceuticals such as peptides (e.g. insulins, insulin containing drugs, GLP-1 containing drugs as well as derivatives thereof), proteins, and hormones, biologically derived or active agents, hormonal and gene based agents, nutritional formulas and other substances in both solid (dispensed) or liquid form. In the description of the exemplary embodiments reference will be made to the use of insulin and GLP-1 containing drugs, this including analogues thereof as well as combinations with one or more other drugs. BRIEF DESCRIPTION OF THE DRAWINGS In the following the invention will be further described with reference to the drawings, wherein FIGS. 1 and 2 show a front-loaded drug delivery device with respectively without a drug cartridge mounted, FIG. 3 shows in an exploded view a drug delivery device subassembly comprising a logging module, FIG. 4 shows an exploded view of the logging module of FIG. 3, FIGS. 5 and 6 show first respectively second rotary sensor parts of the module of FIG. 3, FIG. 7 shows the logging module of FIG. 4 in an assembled state, FIG. 8 shows a cross-sectional view of the subassembly of FIG. 3 in an assembled state, FIGS. 9A-9C show operation of an axial switch of the logging module in different operational states, FIG. 10 shows a schematic representation of how tracks and contacts of a rotary sensor can be arranged, FIG. 11A shows a further schematic representation of how tracks and contacts of a rotary sensor can be arranged, FIG. 11B shows a schematic representation of an alternative arrangement of tracks and contacts of a rotary sensor, FIGS. 12A and 12B show opposed sided of an end portion of a flexible ribbon connector, FIGS. 13A and 13B show opposed sides of a non-conducting moulded matrix member, FIGS. 14A and 14B show opposed sides of a finished ring-formed rotary sensor component, FIG. 15 shows combined conductive structures formed during a second moulding shot, FIG. 16 shows a disc-formed rotary sensor member, FIG. 17 shows a further rotary sensor member, and FIG. 18 shows a drug delivery pen provided with a logging module and in communication with a smartphone. In the figures like structures are mainly identified by like reference numerals. DESCRIPTION OF EXEMPLARY EMBODIMENTS When in the following terms such as “upper” and “lower”, “right” and “left”, “horizontal” and “vertical” or similar relative expressions are used, these only refer to the appended figures and not necessarily to an actual situation of use. The shown figures are schematic representations for which reason the configuration of the different structures as well as their relative dimensions are intended to serve illustrative purposes only. When the term member or element is used for a given component it generally indicates that in the described embodiment the component is a unitary component, however, the same member or element may alternatively comprise a number of sub-components just as two or more of the described components could be provided as unitary components, e.g. manufactured as a single injection moulded part. When it is defined that members are mounted axially free to each other it generally indicates that they can be moved relative to each other, typically between defined stop positions whereas when it is defined that members are mounted rotationally free to each other it generally indicates that they can be rotated relative to each other either freely or between defined stop positions. The terms “assembly” and “subassembly” do not imply that the described components necessary can be assembled to provide a unitary or functional assembly or subassembly during a given assembly procedure but is merely used to describe components grouped together as being functionally more closely related. Referring to FIG. 1 a pen-formed drug delivery device 100 will be described. The device represents a “generic” drug delivery device providing an example of a device in combination with which embodiments of the present invention is intended to be used, such a device comprising a rotational member adapted to rotate corresponding to a set and/or expelled dose of drug. More specifically, the pen device comprises a cap part (not shown) and a main part having a proximal body or drive assembly portion 120 with a housing 121 in which a drug expelling mechanism is arranged or integrated, and a distal cartridge holder portion in which a drug-filled transparent cartridge 180 with a distal needle-penetrable septum can be arranged and retained in place by a cartridge holder 110 attached to the proximal portion, the cartridge holder having openings allowing a portion of the cartridge to be inspected. The cartridge may for example contain an insulin, GLP-1 or growth hormone formulation. The device is designed to be loaded by the user with a new cartridge through a distal receiving opening in the cartridge holder, the cartridge being provided with a piston driven by a piston rod 128 forming part of the expelling mechanism. A proximal-most rotatable dose ring member 125 serves to manually set a desired dose of drug shown in display window 126 and which can then be expelled when the release button 127 is actuated. Depending on the type of expelling mechanism embodied in the drug delivery device, the expelling mechanism may comprise a spring which is strained during dose setting and then released to drive the piston rod when the release button is actuated. Alternatively the expelling mechanism may be fully manual in which case the dose ring member and the release button moves proximally during dose setting corresponding to the set dose size, and then moved distally by the user to expel the set dose. The cartridge is provided with distal coupling means in the form of a needle hub mount 182 having, in the shown example, an external thread 185 adapted to engage an inner thread of a corresponding hub of a needle assembly. In alternative embodiments the thread may be combined with or replaced by other connection means, e.g. a bayonet coupling. The cartridge holder comprises a distal opening adapted to receive a cartridge. More specifically, the cartridge holder comprises an outer rotatable tube member 170 operated by the user to control movement of gripping means to thereby open and close gripping shoulders 145 configured to grip and hold a cartridge. FIG. 2 shows the device with the cartridge removed and the gripping shoulders in their un-locked “open” position in which a cartridge can be removed and a new inserted. As appears, FIG. 1 shows a drug delivery device of the front-loaded type in which a cartridge is inserted through a distal opening in the cartridge holder which in non-removable attached to the main part of the device, however, the drug delivery device may alternatively comprise a cartridge holder adapted to be removed from the device main portion and in which a cartridge is received and removed through the proximal opening. With reference to FIG. 3 a subassembly 200 for a drug delivery device will be described, the subassembly comprising a logging module in combination with parts of the drug delivery device being directly functionally related to the incorporation and operation of logging unit. More specifically, the subassembly comprises an electronically controlled logging module 300, an inner tube member 210, a generally cylindrical inner housing member 220, a dial ring member 230 and a button assembly comprising a button ring 240, a button window 241 and a button spring 242. The inner housing member is configured to be arranged inside an outer housing member providing the exterior of the drug delivery device. The different components of the logging module 300 are shown in FIG. 4. More specifically, the logging module comprises a housing member 310 having a barrel-shaped proximal main portion 311 with a distally extending tube portion 312, a mounting foil member 313, a disc-formed first rotary sensor part 320 onto which a first connector 329 is to be mounted, a disc-formed second rotary sensor part 330, a rotary sensor holder 339 with a lateral projection 337, a flexible PCB 340 folded in a multi-layered stack and onto which a second connector 349 is to be mounted, a battery 345 and battery clip 346, a number of mounting rings 350, 351, 352, an antenna 360, an LCD 370 and an LCD frame 371. On the PCB electronic circuitry components are mounted, e.g. micro-controller, display driver, memory and wireless communication means. As will be described below in greater detail the first rotary sensor part 320 comprises a plurality of arc-formed discreet contact areas, and the second rotary sensor part 330 comprises a plurality of flexible contact arms of which the outer ones provide an axial switch having a laterally extending projection 334. FIG. 5 shows the first rotary sensor part 320 comprising a ring-formed disc formed from circuit board material and on which a number of contact areas (or segments) has been plated on forming three concentric rings, an inner, an intermediate and an outer ring. The disc comprises a central opening 327 with two opposed cut-outs 328 allowing the disc to be mounted non-rotationally on e.g. tube portion 312. In the shown embodiment the inner ring is a single contact area 321 used as ground (i.e. reference), the intermediate ring comprises four discrete arch-formed position contact segments 322 arranged with a certain circumferential distance there between, and the outer ring comprises three discrete arch-formed switch contact segments 323 arranged with only a small circumferential gap there between, the segments being individually connected to a given contact terminal of the multi-terminal connector 329 mounted on the rear (proximal) face of the disc. If a given segment is not connected to a terminal it can be considered a passive segment. The second rotary sensor part 330 shown in FIG. 6 is in the form of a metallic disc comprising a number of flexible arc-formed contact arms protruding proximally, the distal end of each contact arm comprising a dome-formed contact point 335 (facing downwards in the figure) adapted to create a galvanic connection with a given contact area. The contact arms are arranged corresponding to the three concentric rings of the first rotary sensor part. More specifically, the second rotary sensor part comprises two inner contact arms 331, three intermediate contact arms 332 and two outer contact arms 333. In this way a given pair of contact arms provides a combined contact structure adapted to create electric contact between two contact segments. In the shown embodiment the two inner ground contact arms 331 are provided to be in contact with the single ground contact area 321 of the inner concentric ring, the three position contacts arms 332 are provided to be in contact with the four position contact segments 322 of the intermediate concentric ring, and the two outer switch contact arms 333 are provided to be in contact with the three switch contact segments 323 of the outer concentric ring, the outer switch contact arms carrying a laterally extending projection 334. Indeed, for the intermediate and outer contact arms the rotational position between the two sensor parts will determine which contact segment is engaged with a given contact arm. In the shown embodiment the gaps between two neighbouring outer contact segments are dimensioned such that the dome-formed contact point will be in contact with both segments as it moves from one segment to the next, this being explained in greater detail below. The second rotary sensor part further comprises a gripping part 336 adapted to engage the projection 337 on the rotary sensor holder 339 to prevent rotational movement there between. In the shown embodiment the intermediate arms and contact segments provide the rotary sensor contacts whereas the outer arms and contact segments provide an axial switch as will be described in greater detail below. FIG. 7 shows the logging module 300 in an assembled state. The flexible PCB 340 with its mounted components and the antenna have been mounted in a sandwich configuration with the mounting rings 350, 351, 352 providing the required spacing and attachment via e.g. gluing or adhesives, the battery 345 being attached to the PCB via battery clip 346. The PCB sandwich is mounted with a “tongue” threaded through a distal opening in the housing 311 button portion and held in place with adhesive mounting foil member 325 (see FIG. 4) during assembly. The first rotary sensor part 320 is mounted non-rotationally on the tube portion 312 and connected to the PCB via the connectors 329, 349. The second rotary sensor part 330 is mounted non-rotationally and axially fixed on the rotary sensor holder 339 which is mounted rotationally free but axially fixed on the tube portion 312. By this arrangement the flexible rotary sensor arms are held in sliding contact with the contact surfaces. The LCD 370 is mounted on top of the PCB sandwich which together is held in place in the housing barrel by the display frame 371 which is permanently attached to the housing by e.g. welding. As appears, in this way an electronic logging module is provided comprising a distally arranged rotatable sensor part. As shown in FIG. 4 the housing main portion 311 comprises a circumferential distal flange 313 with a number of proximally projecting teeth 314 and a circumferential proximal groove 315. The tube portion 312 is provided with distal snap connectors 316 adapted to engage corresponding openings 211 in the inner tube member 210. FIG. 8 shows a cross-sectional view of the subassembly 200 in an assembled state. The term “subassembly” does not imply that the shown parts necessary are assembled to provide a subassembly as shown and which can be used in an assembly process for a given drug delivery device. In contrast, the shown logging module of FIG. 7 may be provided in the shown form as a “real” subassembly. Referring to the parts shown in FIGS. 3 and 4, the inner tube member 210 is connected rotationally and axially locked to the distal tube portion 312 of the logging module. This arrangement is mainly for the purpose of moulding and subsequent assembly. The dial ring member 230 is mounted on the proximal portion of the housing member 220 on which is allowed to freely rotate but not move axially. The dial ring member 230 comprises an inner circumferential coupling flange 231 with a plurality of distally facing teeth adapted to engage the proximally facing teeth 314 of the logging module to thereby rotationally lock the two components during engagement. The housing member 220 comprises first and second openings or cut-outs 221, 222 adapted to engage respectively the rotary sensor holder lateral projection 336 and the axial switch lateral projection 334, this ensuring non-rotational engagement between the second rotary sensor part and the housing yet allows axial movement. The button 240 with the window 241 attached is mounted on the module housing in gripping engagement with the circumferential groove 315, this allowing the button to rotate relative to the module housing. The axially compressed button assembly spring 242 is arranged in the circumferential gap between the module housing and the dial ring member and held in place between a distally facing ring portion of the button ring and the proximally facing portion of the coupling flange. In this way the spring provides an axial force biasing the module proximally into non-rotational engagement with the dial ring member 230 via the coupling flange, however, when a distally directed force is applied to the module via the button the module can be moved distally and thereby out of the rotational coupling with the dial ring member, this allowing the logging module main housing to rotate relative to the dial ring member. As indicated above, the shown rotary sensor comprises an axial switch, this switch serving to detect an axial position of the logging module relative to (here) the housing member 220. More specifically, FIG. 9A shows the logging module 300 biased into an initial proximal position by the button spring 242, FIG. 9B shows the logging module in an intermediate position in which it has been moved distally by the distance H1, and FIG. 9C shows the logging module in an actuated distal position in which it has been moved distally by the distance H2. In all three states the axial switch lateral projection 334 is positioned in the corresponding housing opening 221 and rotationally locked to the housing via the rotary sensor holder 339. As appears, in FIG. 9A the switch projection 334 engages a proximal edge of the opening and the flexible switch arm 333 with the contact point 335 is thereby held out of contact with the first rotary sensor part 320, in FIG. 9B the switch projection 334 still engages the proximal edge of the opening, however, the logging module has been moved distally and thereby the first rotary sensor part 320 has been moved into contact with the switch arm 333, this bringing the axial switch into an “on” state detectable by the logging module circuitry, and in FIG. 9C the logging module has been moved further distally to its actuated distal position. The switch projection 334 has been moved out of engagement with the proximal edge of the opening, the axial switch thus remaining in its “on” state. In an exemplary embodiment the axial movement between the different positions may be e.g. 1.5 mm, this ensuring that the expelling mode is safely registered by the axial switch before the dosing mechanism is actually released. The axial switch could also be used to control the functioning of the logging module when no dose has been set, see below. Returning to the first and second rotary sensor parts of FIGS. 5 and 6 the intermediate arms and contact segments provide the rotary sensor contacts whereas the outer arms and contact segments provide an axial switch as will be described in greater detail below. This is illustrated in FIG. 10 in which the intermediate segments provide a “Gray Code Track” with the segments denoted “GC”, the intermediate arms provide “Gray Code Contacts”, the outer segments provide a “Mode Switch Track” with segments denoted “MS” and the outer arms provide “Mode Switch Contacts”. As also illustrated in FIG. 10 the described rotary sensor has a resolution of 15 degrees, i.e. 24 steps for a full rotation with only steps 1-9 numbered in the figure, such that for each 15 degrees of rotation a pre-determined change in which of the individual position rotary contacts are on and off is created. As each of the shown contact segments is connected to the electronic circuitry 340 it is possible to determine the relative rotational position between the two rotary sensor parts (see below). In respect of the above-described axial switch, using only one switch there would be a single point of failure when the information is to be detected by electrical means. Correspondingly, as shown in FIG. 6, two axial switch contacts are provided, however, providing redundancy by merely adding a further contact would introduce a new single point of failure should one of the contacts fail. Accordingly, the axial switch of the described embodiment has been designed to allow detection of failure of one of the two axial switches, this allowing the system to take appropriate action, e.g. indicating an error condition, before the system will actually malfunction. More specifically, as shown in FIG. 5 the conductive outer ring has been split up in 3 segments 323 identified as MS1, MS2 and MS3 in FIG. 10. When the flexible outer switch arms 333 are moved into contact with the conductive ring, the arms and segments are arranged such that conductive contact will be established with at least two of the three segments. During a full rotation the arms will move over the three segments when the arms are pressed down, and thus give the following code pattern (24 steps for a full rotation), where the value “0” means that an arm is in contact with a segment: MS1 MS2 MS3 0 0 1 0 1 0 1 0 0 1 0 0 1 0 0 1 0 0 1 0 0 1 0 0 1 0 0 0 0 1 0 1 0 0 1 0 0 1 0 0 1 0 0 1 0 0 1 0 0 1 0 1 0 0 0 0 1 0 0 1 0 0 1 0 0 1 0 0 1 0 0 1 In case the arms are not pressed down, the values for the three segments are 1,1,1. If one of the switch contact segments or contact arms is faulty, the code pattern will be different from the above pattern. For example, if MS1 is faulty with the value “1” when the arms are pressed down then the first code would be 1,0,1. This fault is detectable since only one of the segments has the value “0” (at least two “0” is expected in a healthy system), this allowing a single contact failure to be detected. Theoretically, if one functioning contact arm was bridging the gap between two neighbouring sensor areas and the other contact arm was faulty, then this would represent a non-error condition with two “0” values. However, if error detection is performed during rotation this special condition could be detected and disregarded. If MS1 is faulty with the value “0” when the arms are not pressed down then this fault is detectable since the values for the three segments should be 1,1,1 when the arms are not pressed down. Although not implemented in the described embodiment, the outer contacts could also be used to provide additional rotational position information to the system. Returning to the first and second rotary sensor parts of FIGS. 5 and 6 the intermediate arms and contact segments provide the rotary sensor contacts whereas the inner arms and single circumferential contact segment provide a ground contact. This is illustrated in FIG. 11A in which the intermediate segments provide a “Code Track” with the segments denoted “GC” and the intermediate arms provide “Code Contacts” as in FIG. 10. The inner segment provides a “Ground reference track” denoted “GND” and the inner arms provide “Ground contacts”. In an alternative embodiment shown schematically in FIG. 11B the code track and the ground reference track GND has been combined to a single “Code and Ground Track” by “superimposing” the code segments 422 onto the ground track 421 just as the dedicated ground contacts have been removed, the former code contacts now serving as combined code and ground contacts 432. More specifically, the code segments have been arranged as isolated “islands” in the ground track, this providing a single combined code and ground track in which a given circumferential portion is represented by either a code segment or a ground segment, whereby a given combined code and ground contact will be in contact with either a code segment or one of the ground segments. As shown in FIG. 11B the ground segments are connected to electrically form a single combined ground segment. In the shown embodiment the individual ground segments are connected by narrow strips of plating surrounding the radial sides of the code segments, this giving the code segments an “island” appearance, however, the ground segments could be connected e.g. on only one side of the code segments or via connections formed on the opposed side of the disc. In the shown embodiment the code segments, the ground segments and the individual combined code and ground contact arms are arranged such that for a given rotational position at least one of the arms will be in contact with a ground segment, the remaining arms being in contact with a code segment to provide positional information. As appears, by this arrangement it is possible to maintain the same functionality as with two separate tracks and dedicated arms for each track, the design allowing a more compact and simpler sensor to be manufactured. Addressing the issue of providing a rotary sensor which is both compact and reliable, a further embodiment of a rotary sensor component as well as a method of manufacture using two-shot moulding will be described with reference to FIGS. 12-15. FIGS. 12A and 12B show opposed sided of an end portion 500 of a flexible ribbon connector having a plurality of conductors 505, each conductor having a connector pad 506 arranged corresponding to the free end region 501 of the ribbon connector. The remaining portion of the ribbon connector (not shown) may form part of a flexible PCB or may be provided with a connector for being connected to e.g. a PCB. FIGS. 13A and 13B show opposed sides of an non-conducting moulded matrix member forming an unfinished ring-formed rotary sensor component 510, the free end of the above-described flexible ribbon connector 500 being attached by in-moulding. A first surface 520 of the sensor component comprises a plurality of circumferentially arranged sensor cavities 521, and an opposed second surface 530 of the sensor component comprises a plurality of narrow circumferentially arranged connector cavities 531, each connector cavity having a proximal end 532 arranged corresponding to the in-moulded ribbon connector as well as one or more short leg portions 533 extending radially. The proximal end of each connector cavity is in communication with a corresponding connector pad and each distal leg portion is in communication with a corresponding sensor connector via an opening 522 formed in the matrix member. The central opening 527 is shown for illustrative purposes and is not intended for actual mounting on a specific structure. FIGS. 14A and 14B show opposed sides of a finished ring-formed rotary sensor component 560, the above-described cavities 521, 531 being filled with a conductive polymer during a second moulding shot, thereby forming a plurality of sensor areas 525 on the first surface 520 and a plurality of conductors 535 on the second surface 530, thereby electrically connecting one or more sensor areas to a given connector pad 506 (see FIG. 12B). In this way a both reliable and compact connection is established between the individual sensor areas and the corresponding ribbon conductors. FIG. 15 shows in a “virtual” representation the combined conductive structure formed during the second moulding shot, the combined structure comprising a plurality of sensor structures 525 each having a contact surface and being connected to one of a plurality of thread-like conductors 535. As appears, for illustrative purposes the sensor component 560 of FIG. 14A only comprises a single ring-formed sensor array, however, in alternative embodiments further conducting structures may be provided. Further, also the combined code and ground track of FIG. 11B may be realized using the above-described two-shot moulding process, the ground segments 421 being connected by moulded connectors on the opposed surface. Addressing the issue of providing a rotary sensor which is both compact and reliable, a yet further embodiment of a rotary sensor component will be described with reference to FIGS. 16 and 17. FIG. 16 shows a rotary sensor member 600 comprising a ring-formed disc formed from e.g. circuit board material, the disc having a first surface 601, an opposed second surface 602 and a circumferential edge surface 603. On the first surface a code track is formed and on the second surface a ground track is formed (not shown), e.g. by plating. The code track essentially corresponds to the code tracks shown in FIGS. 5 and 14A comprising a plurality of circumferentially arranged discrete arcuate contact areas 611, and the ground track essentially corresponds to the circumferential ground track shown in FIG. 5. By arranging the two tracks on two surfaces a code disc with a smaller diameter can be realized. Alternatively a ground track may be arranged on the edge surface 603. FIG. 17 shows a further embodiment of a rotary sensor member 650 comprising a ring-formed disc portion 651 and a central co-axially arranged cylinder portion 655, the disc portion having a first surface 661, an opposed second surface 662 and a first circumferential edge surface 663, the cylinder portion providing an upper third surface 664, an outer second circumferential cylindrical surface 665, and an inner third circumferential cylindrical surface 666. In the shown embodiment the rotary sensor member thus has three axially offset coplanar surfaces 661, 662, 664, and three radially offset co-axial cylinder surfaces. The rotary sensor member 650 may be formed from e.g. an injection moulded polymeric material. The three planar and two cylindrical surfaces provide a total of five surfaces which each may serve as a carrier for one or more circumferential sensor tracks. In the shown embodiment a first circumferential code track with a plurality of discrete contact areas 652 is formed on the first planar surface 661, a second circumferential code track with a plurality of discrete contact areas 656 is formed on the outer second cylindrical surface 665, a third circumferential code track with a plurality of discrete contact areas 657 is formed on the first cylindrical edge surface 663, a fourth circumferential code track with a plurality of discrete contact areas 659 is formed on the inner third cylindrical surface 666 and a circumferential ground track 658 is formed on the second planar surface 662. A further track may be formed on the not seen lower second planar surface. In respect of the embodiment of FIG. 17 one or more of the shown tracks may be omitted leaving at least two tracks on two of the described surfaces. By providing a rotary sensor member with a plurality of planar and cylindrical surfaces a larger number of tracks can be formed for a given diameter size of the sensor member. The parts of the subassembly 200, apart from module 300, as shown in FIG. 3 represent “generic” parts of a drug expelling mechanism having properties which are relevant for the implementation of embodiments of the present invention. More specifically, the shown module 300 is adapted to be implemented in a drug delivery device having a housing, dose setting means allowing a user to set a dose of drug to be expelled, and a rotational member adapted to rotate corresponding to a set and/or expelled dose. In the shown subassembly the inner tube member 210 represents a “generic” rotational member. Although not part of the present invention, in the following a short description of a drug expelling mechanism into which the shown inner tube member 210 could be integrated will be described. When setting a dose to be expelled the user rotates the dial ring member 230 and thereby the inner tube member 210 to a given rotational position representing a desired dose, this straining a torsional spring member arranged around the tube member and attached at its proximal end to a housing proximal portion and at its distal end to the tube member distal portion. A ratchet coupling arranged at the distal end of the inner tube member serves to hold the now rotationally biased tube member in the set position. A scale drum is coupled to and rotates with the tube member, the scale drum having a threaded connection with the housing (e.g. threads 226 in FIG. 3) whereby a spirally arranged series of numeric values is moved relative to a window in the housing (e.g. opening 225 in FIG. 3), the shown number indicating the presently set dose. To release the set and loaded mechanism the user pushes a proximal release button whereby the inner tube member is moved distally. By this action the ratchet coupling (serving as a release member) is released and the inner tube member is moved into engagement, directly or indirectly, with a rotational drive member, the drive member being arranged to rotate a piston rod which due to a threaded engagement with the housing is moved distally to thereby the set dose. As the tube member rotates backwards, thereby driving the piston rod distally, also the scale drum is rotated backwards and reaches its initial “zero” position together with the tube member. This kind of mechanism is known from e.g. the FlexTouch® drug delivery pen device marketed by Novo Nordisk for the injection of e.g. insulin formulations. As appears, in the described exemplary mechanism the inner tube member 210 (to which the main portion of the logging module 300 is rigidly mounted) rotates relative to the housing 220 during both setting and expelling of a given dose. As the second rotary sensor part 330 is rotationally locked to the housing, also the two rotary sensor parts 320, 330 rotate relative to each other during both setting and expelling of a given dose. As this is merely an exemplary mechanism, other mechanisms can be envisaged in which a given member rotates only during setting or expelling. This said, in the shown embodiment the logging module is adapted to detect rotation in both directions corresponding to a set dose and an expelled dose. In the shown embodiment the logging module is further provided with an axial switch allowing the module to detect whether the mechanism is in the setting or expelling mode, however, this is an optional feature. In the shown embodiment the code pattern has a step “resolution” of 15 degrees of rotations which for a given drug formulation and delivery device combination may correspond to 1 unit (IU) of insulin. Indeed, for a drug formulation having the double concentration a 7.5 degree rotary resolution would be necessary to register dose steps corresponding to 1 IU of insulin. The rotary sensor comprising the rotary contacts and the associated electronic circuitry could be designed to detect the amount of rotation using a number of designs, e.g. each 15 degrees increment may be counted, or a given position may be detected absolutely within sectors of e.g. 120 or 360 degrees, a counter registering the number of completed sectors. Such a counter could be implemented using the switch arms and outer contact areas described with reference to FIGS. 5 and 6. With a “counting” design it is important that the first increment is registered, however, modern electronics can be operated in a low-power “on” state avoiding the delay normally associated with a wake-up change of state from a “sleep” state to an “on” state. In an exemplary embodiment the rotary sensor is designed to count the number of steps during setting and to count down the number of steps during expelling, with the expelling steps being registered in the log as the dose being expelled. By counting in both directions proper registering and functioning of the logging module can be assured to a high degree. As a given dose of drug, especially if large, may be divided and injected with a given pause, the logging module may be programmed to log two dose amounts expelled within a given time window, e.g. 15 minutes, as one dose. The logging module may be configured to store and show data in different ways. To many users the time since last dose and the size of that dose are the most important values. To other users and/or a medical practitioner an overview of the entire log for a given period, e.g. a week or a month, may be of importance. To allow such an overview the logging module may be provided with output means allowing the dose log to be transferred, e.g. by NFC transfer, to an external display device, e.g. a smartphone or computer for better graphic overview, see below. To ensure that the full dose is expelled the logging module may be set up to display the last expelled dose only when the expelling mechanism has been returned to zero. Otherwise a given “half” dose will be stored in the log but not displayed. For example, if a dose of 40 IU is dialed and 20 IU are expelled immediately thereafter the display will not show data for that delivery. To have the dose shown in the display the user may expel the remaining dose and the combined dose of 40 IU together with a time stamp will be shown in the display. Alternatively the user may dial the expelling mechanism back to zero and the display will show 20 IU, or the user may dial the expelling mechanism back to 10 IU and expel the 10 IU and the display will show 30 IU. Indeed, for the expelled amounts to be combined the two (or more) doses will have to be expelled within the above-described time window, e.g. 15 minutes. Otherwise only the last portion of the dose will display, the first portion being stored merely as an entry in the log. The display can be configured to show data in different formats. For example, the display 711 of FIG. 18 is a two-line display in which time is shown using a HH:MM:SS stop watch design, this providing that the time since the last dose expelled from the device can be shown with a running second counter allowing a user to easily identify the shown information as a counting time value. After 24 hours the display may continue to display time in the HH:MM:SS format or change to a day and hour format. To save energy the display will turn off after a pre-determined amount of time, e.g. 30 seconds. To turn on the display again the user may e.g. press the button thereby using the axial switch to turn on the display, or the display may be turned on when the dose dial is turned away from and then back to zero. A user may desire to check the dose log directly on the module display. Toggling through the dose log could also be controlled by the axial switch, e.g. two fast pushes on the button 712 will bring the module into log display mode, each consecutive push on the button recalling the next log entry. The module may leave the log display mode automatically after a given amount of time, or the user may bring the module into normal display mode by e.g. dialling back and forth as described above. As an alternative, the electronic module may be provided with other types of input means, e.g. a motion sensor which would allow a user to turn on the display by shaking or tapping, or a touch sensor integrated in the display as is well known from e.g. smartphones which would allow a user to turn on the display by swiping a finger across the display. FIG. 18 shows a drug delivery pen 700 provided with a logging module 710 as described above and arranged next to a smartphone 730 configured to receive logging data from the logging module via wireless communication, e.g. NFC or Bluetooth®. As appears, the logging module is provided with a display configured to indicate the size of the last dose and the time since the last dose using the stopwatch display mode. In order to communicate with the logging module the smartphone has been provided with specific “insulin diary” software. When the software is activated to initiate data transfer the smartphone NFC transmitter will transmit specific code which will wake up any nearby logging module which will then retransmit a unique code identifying the specific module. If a specific code is received for the first time the user is asked to confirm pairing and is asked to select from a list the given drug that should be associated with the given logging module, e.g. “Mix 30” as shown. In this way the smartphone can create an insulin diary covering more than one drug. In the described simple “manual” set-up the user has to ensure that a correct cartridge, e.g. with Mix 30 insulin, is loaded in a drug delivery pen which has been associated with that type of drug. Indeed, other set-ups can be envisaged, e.g. a given pen may be (mechanically) coded to only accept a given type of cartridge with the designated type of drug, or the pen and logging module may be provided with the ability to identify different types of cartridges and thus types of drug. In the shown embodiment log data from a logging module associated with a Mix 30 insulin has been transferred. In the exemplary user interface the user can toggle back and forth between different day views, each day view showing the different amounts of drug delivered together with a real time value. In FIG. 18 on a given day 731 first and second amounts 732 of Mix 30 has been delivered with the time and amount shown for each delivery. In the above description of exemplary embodiments, the different structures and means providing the described functionality for the different components have been described to a degree to which the concept of the present invention will be apparent to the skilled reader. The detailed construction and specification for the different components are considered the object of a normal design procedure performed by the skilled person along the lines set out in the present specification. 15037256 novo nordisk a/s USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open Apr 27th, 2022 08:32AM Apr 27th, 2022 08:32AM Novo Nordisk Health Care Pharmaceuticals & Biotechnology
nyse:nvo Novo Nordisk Apr 26th, 2022 12:00AM Jun 25th, 2019 12:00AM https://www.uspto.gov?id=US11311679-20220426 Automatic injection device with a top release mechanism The present invention relates to a handheld mechanical injection device by which set doses of a liquid medicament can be injected from a medical reservoir. The medicament is expelled through an injection needle by release of a power reservoir in the device, the power reservoir being fully or partially released by actuation of a user operable release member being positioned at or near an upper end of the injection device, the upper end being that end of the injection device which is opposite the injection needle. 11311679 1. A handheld injection device by which set doses of a liquid medicament can be injected from a medical reservoir through an injection needle, comprising: a rotatable dose setting member, a power reservoir comprising a torsion spring for storing energy to expel the set doses of medication from the injection device, a user operable release member positioned at or near an upper end of the injection device (1), the upper end being that end of the injection device (1) which is opposite the injection needle, the injection device further comprising a multi-component driver having at least a part (38) adapted to drive a piston rod, and a further part (36) being axial movable into a position disconnected from the housing releasing the energy accumulated in the power reservoir, the further part (36) being axially movable by the user applying a force onto the release member, and wherein the injection device further comprises a display member adapted to display the dose to be ejected from the injection device in accordance with a setting of the dose setting member, the display member being rotatably mounted and rotatable over an angle corresponding to at least one revolution of the display member and which display member comprises a dose indicator barrel having numerals arranged along a substantially helical path on an outer surface thereof. 2. A handheld injection device according to claim 1, wherein the amount of power provided to the power reservoir (10) depends on the angle of rotation of the dose setting member (7). 3. A handheld injection device according to claim 1, wherein the release member is operatively connected to the dose setting member of the injection device. 4. A handheld injection device according to claim 3, wherein the release member engages the dose setting member via a key/keyway connection when the dose setting member is in a dose setting position. 5. A handheld injection device according to claim 4, wherein the release member is released from the key/keyway connection with the dose setting member when the dose setting member is in a dose injecting position. 6. A handheld injection device according to claim 1, wherein when energy from the torsion spring is released it is adapted to expel a set dose of medicine from a medicine containing reservoir through the injection needle. 6 CROSS-REFERENCE TO RELATED APPLICATIONS This application is a continuation of U.S. patent application Ser. No. 15/441,638, filed Feb. 24, 2017, which is a continuation of U.S. patent application Ser. No. 14/797,350, filed Jul. 13, 2015 (U.S. Pat. No. 9,616,180), which is a continuation of U.S. patent application Ser. No. 13/326,738, filed Dec. 15, 2011 (U.S. Pat. No. 9,108,002) which is a continuation of U.S. patent application Ser. No. 11/813,435 filed Jun. 2, 2008 (U.S. Pat. No. 8,096,978) which is a 35 U.S.C. § 371 national stage application of International Patent Application PCT/DK2006/000032 (published as WO 2006/076921), filed Jan. 20, 2006, which claimed priority of Danish Patent Application PA 2005 00113, filed Jan. 21, 2005; this application further claims priority under 35 U.S.C. § 119 of U.S. Provisional Application 60/647,320, filed Jan. 26, 2005; the contents of which are incorporated herein by reference. The present invention relates to an automatic and handheld mechanical injection device where an injection of a set dose of medicament is initiated by actuating a release member being arranged at or near the top of the injection device. BACKGROUND OF THE INVENTION Automatic injection devices have previously been disclosed in the patent literature. Automatic injection devices contain some sort of power reservoir where electrical or mechanical energy can be accumulated. The accumulated energy is easily released by actuating a release mechanism whereby the accumulated energy assists the user in injecting a set dose of medicine and/or assisting needle insertion. For example, EP 0 516 473 A1 discloses an injection device having a needle which, when the device is operated, is first caused to project, then liquid is forced out through it, and finally the needle is automatically retracted. The needle extends forwardly from a capsule that can slide longitudinally within a barrel-like body, a relatively weak spring normally maintaining the capsule and needle retracted. A more powerful spring acts oppositely on a plunger which, when released, shoots the capsule forward by acting on the liquid therein, and then forces the liquid out through the projecting needle. At the end of the forward stroke the plunger and capsule are decoupled and the weak spring returns the exhausted capsule and its needle to the retracted position. The spring acting on the plunger can be released by a release button positioned on the outer surface of the injection device. In WO 01/41838 discloses a handheld injection device by which set doses of a liquid medicament can be injected from a medical reservoir, such as cylinder ampoule, by release of a power reservoir in the device. The power reservoir can either be an electric battery by which a motor can be energized to press out a set dose of medicine, or a strained spring maintained in its strained position by a detent which spring when released can press out a set dose of medicine. When the power reservoir is released, the liquid medicine will be pressed out from the cylinder ampoule through an injection needle mounted on the cylinder ampoule or on the injection device carrying the cylinder ampoule. The power reservoir is released fully or partially by activating a release button, such as an electric switch, located on the housing of the injection device and in the distal half of the length of the injection device. By making at least a part of the distal third of the injection device of an ergonomic shaped cross section, the user can grip the injection device as a pencil is gripped by a thumb, an index finger and a long finger. In both EP 0 516 473 A1 and WO 01/41838 the release buttons are positioned on an outer surface of the injection devices. In EP 0 516 473 A1 the release button is position on the outer side of the cylindrical body, whereas in WO 01/41838 the release button is positioned close to the injection needle of the injection device. However, it may be advantageous to position the release button or mechanism so that the injection device can be activated by providing a force to the upper region of the injection device—preferably to a release button or mechanism arranged axially with the injection device. It is an object of the present invention to provide an automatic and handheld mechanical injection device having a combined release member and dose setting member It is a further object of the present invention to provide an automatic and handheld mechanical injection device where an injection of a set dose can be initiated using the thumb or the index finger of the hand handling the injection device by providing an axial force to an upper region of the injection device. It is a still further object of the present invention to provide an automatic and handheld mechanical injection device having an exterior design very similar to conventional manual injection devices. SUMMARY OF THE INVENTION The above-mentioned objects are complied with by providing, in a first aspect, a handheld injection device by which set doses of a liquid medicament can be injected from a medical reservoir through an injection needle by release of a power reservoir in the device, the power reservoir being adapted to be fully or partially released by actuation of a user operable release member positioned at or near an upper end of the injection device, the upper end being that end of the injection device which is opposite the injection needle, the power reservoir being adapted to be powered by rotation of a rotatably mounted dose setting member. The amount of power provided to the power reservoir may depend on the angle of rotation of the dose setting member. Thus, a rather limited rotation of the dose setting member provides a relatively small amount energy to the power reservoir, whereas a large rotation of the dose setting member provides a relatively large amount of energy to the power reservoir. The release member may be positioned less than one fifth or one sixth of the length of the injection device from the upper end. Alternatively, the release member may be axially arranged relative to the injection device so that the release member forms a push button like release member on the top of the injection device. The release member may be operatively connected to a dose setting member of the injection device in that the release member may engage the dose setting member via a key/keyway connection when the dose setting member is in a dose setting position. The release member may be released from the key/keyway connection with the dose setting member when the dose setting member is in a dose injecting position. With this arrangement, the handheld injection device has no rotating exterior parts or elements. The power reservoir may be a resilient member, such as a torsion spring or a linear spring, the resilient member being, when released, adapted to press out a set dose of medicine from the medical reservoir through the injection needle. The release member may be operatively connected to a release mechanism adapted to release the resilient member when said release member is actuated. The release member may have a shape which is ergonomic shaped to be activated by a thumb or an index finger of the user. The medical reservoir may be a cylindrical ampoule comprising a first and a second end of which the first end is closed by a pierceable membrane which may be pierced by a first end of the injection needle when this needle is mounted on the device. The other end of the injection needle may be sharp so as to be able to pierce the skin at the position where an injection is to be made. The second end of the ampoule may be closed by a piston which may be forced into the ampoule so as to expel medicament through the needle. The handheld injection device may further comprise a rotatably arranged drive member being adapted to at least partly engage with at least part of a drive track of an associated piston rod, the drive member being adapted to be positioned in a first axial position when the dose setting member is in a dose setting position, the drive member further being adapted to be positioned in a second axial position when the dose setting member is in a dose injection position, the drive member being adapted to release energy accumulated in the power reservoir when the drive member is in its second axial position. The drive member may be adapted to rotate the associated piston rod upon releasing the accumulated energy in the power reservoir. However, in its first axial position, the drive member is prevented from rotating because the drive member engages at least part of a housing of the injection device. The injection device may further comprise a resilient member, such as a linear spring, for biasing the drive member in a direction towards the dose setting member. The linear spring operatively connects the drive member and the housing. The dose setting member may be adapted to be moved a distance along an axial direction of the injection device so as to move the drive member between the first and second axial positions. The drive member may be adapted to be moved from the first to the second axial position by applying a force to the dose setting member, the force being applied along the axial direction of the injection device. The injection device may, as already mentioned, further comprise a push button axially arranged with the dose setting member, the push button being adapted to engage with the dose setting member when the dose setting member is in its dose setting position, and disengage from the dose setting member when the dose setting member is in its dose injection position. By disengage is meant that the push button and the dose setting member are mutually rotatable when this disengaged state is reached. The injection device may further comprise a resilient member, such as a linear spring, for axially biasing the push button in a direction away from the drive member. The handheld injection device may further comprise a rotatably mounted display member adapted to display the dose to be ejected from the injection device in accordance with a setting of the dose setting member, the rotatably mounted display member being rotatable over an angle corresponding to at least one revolution of the display member. The display member may comprise a dose indicator barrel having numerals arranged along a substantially helical path on an outer surface thereof. Alternatively or in addition, the display member may comprise a counting device having two or more display wheels having numerals arranged on an outer surface thereof. The handheld injection device may further comprise the associated the piston rod, the piston rod having a threaded outer surface with the drive track arranged in a longitudinal direction of the outer surface of the piston rod. The drive member may be operatively connected to the dose setting member via a ratchet. The power reservoir may be arranged between the housing and the dose setting member in such a way that when the dose setting member is rotated, energy is accumulated in the power reservoir. The power reservoir may comprise a torsion spring formed as a helical spring extending coaxially with the associated piston rod. It is to be noted that the interaction between the drive member, the piston rod and the housing may be implemented in various ways. Above, the piston rod has a threaded outer surface and a drive track arranged in the longitudinal direction of the rod. A key arranged on the drive member engages the drive track of the rod and the forward movement of the rod relative to the housing is caused by the threaded outer portion of the rod which meshes with a corresponding threaded portion of the housing. Alternatively, the threaded outer surface of the rod may mesh with a corresponding threaded portion of the drive member whereas the drive track arranged in the longitudinal direction of the rod engages with a key fixedly arranged relative to the housing. BRIEF DESCRIPTION OF THE INVENTION The present invention will now be explained in further details with reference to the accompanying figures wherein FIG. 1 shows an injection device according to the present invention where the release button arranged at the top of the device is activated by the thumb of the user, FIG. 2 shows an injection device according to the present invention where the release button arranged at the top of the device is activated by the index finger of the user, FIG. 3 shows an injection device according to the present invention where the release button is arranged on the top surface of the dose setting member, and where the drive member is in its locked position (dial position of dose setting member), FIG. 4 shows an injection device according to the present invention where the release button is arranged on the top surface of the dose setting member, and where the drive member is in its released position (dosing position of dose setting member), FIG. 5 shows an expanded view of the drive member in its released position, FIG. 6 shows an expanded view of the release member in its locked position with the dose setting member, FIG. 7 shows an expanded view of the release member in its released position with the dose setting member, FIG. 8 shows an expanded view of the release member in a further released position where the dose setting member is allowed to rotate, FIG. 9 shows one way of implementing the release mechanism for releasing the energized power reservoir, FIG. 10 shows another way of implementing the release mechanism for releasing the energized power reservoir, FIG. 11 shows a third way of implementing the release mechanism for releasing the energized power reservoir, FIG. 12 shows a fourth way of implementing the release mechanism for releasing the energized power reservoir, and FIG. 13 shows a fifth way of implementing the release mechanism for releasing the energized power reservoir. While the invention is susceptible to various modifications and alternative forms, specific embodiments have been shown by way of example in the drawings and will be described in detail herein. It should be understood, however, that the invention is not intended to be limited to the particular forms disclosed. Rather, the invention is to cover all modifications, equivalents, and alternatives falling within the spirit and scope of the invention as defined by the appended claims. DETAILED DESCRIPTION OF THE INVENTION FIGS. 1 and 2 show the present invention in its most general aspect. In FIG. 1 a handheld injection device 1 is shown. The injection device has an injection needle 2 fastened to one of its ends, whereas a release button 3 is arranged at the opposite end of the injection device. When the release button 3 is actuated by provided a force to it along the axial direction of the device energy is released from an internal power reservoir whereby a set dose of medicine is injected from the injection device. In FIG. 1 the release button is actuated by the thumb 4 of the user, whereas in FIG. 2 the release button is actuated by the index finger 5 of the user. The medicine to be injected is contained in a medical reservoir typically formed as a cylindrical ampoule. The energy released when the release button 3 is mechanical energy. The power reservoir can be a resilient member, such as a torsion spring, the resilient member being, when released, adapted to press out a set dose of medicine from the medical reservoir through the injection needle. The release button is operatively connected to some sort of release mechanism adapted to release the resilient member when the release button is actuated. FIG. 3 shows a cross-sectional view of one embodiment of the present invention. The injection device shown in FIG. 3 comprises a housing 6, a dose setting member 7, a drive member 8, a piston rod 9, a torsion spring 10, a biasing spring 11, a cylindrical ampoule 12 and a release member 13. FIG. 3 shows the injection device in a state where the dose setting member 7 is in its dose setting position. A dose is set by rotating the dose setting member 7 a certain angle or a certain number of turns. By rotating the dose setting member 7 the torsion spring 10 is strained because the two ends of the torsion spring 10 are fixed to the housing 6 and to the dose setting member 7, respectively. The dose setting member 7 is operatively connected to the drive member 8 via a ratchet (not shown). This ratchet prevents that the dose setting member 7 returns to its initial position upon straining the torsion spring 10. Since the drive member 8 engages the housing 6 via a key/keyway connection or a gear wheel, the drive member 8 is not allowed to rotate relative to the housing 6 as long as the dose setting member 7 is in its dose setting position as illustrated in FIG. 3. In order to keep the dose setting member 7 and the drive member 8 in the dose setting position, the drive member 8 and the dose setting member 7 is biased in a direction towards the top end of the injection device. This biasing is provided by a spring element, such as a linear spring 11, arranged between the drive member 8 and part of the housing 6. Thus, in order to release the drive member 8 from its engagement with the housing 6, a force needs to be provided in order move the dose setting member 7 and the drive member 8 towards the medicine ampoule 12. A miner cavity 14 ensures that this forward movement of the dose setting member 7 and the drive member 8 can be performed. Similarly, since the drive member 7 and the piston rod 9 engage via a key connection the drive member 8 is allowed to move axially relative to the piston rod 9. The drive member 8 has been released from its engagement with the housing 6 in FIG. 4. In order to achieve this releasing a force, indicated by arrow 15, has been provided to the release member 13 whereby the release member 13, the dose member 7 and the drive member 8 have all been moved a distance towards the medicine ampoule 12. The force indicated by arrow 15 would normally be provided by the thumb or the index finger of the user. As seen in FIG. 4 the engaging region 16 of the housing is now separated from the engaging region 17 of the drive member 8. This disengagement allows that the strained torsion spring 10 can release its energy to the dose setting member 7. The dose setting member 7 and the drive member 8 are fixedly related via the intermediate ratchet (not shown). Thus, when a disengagement between engaging regions 16 and 17 has been established, the dose setting member 7 and the drive member 9 will rotate until the torsion spring 10 reaches an unstrained state. Since the drive member 8 and the piston rod 9 is connected via a key connection the rotation of the dose setting member 7 and the drive member 8 will cause the piston rod 9 to rotate as well. The piston rod 9 has an outer threaded surface which engages with a corresponding threaded portion 18 of the housing whereby the piston rod 9, upon rotation thereof, will perform a translational movement along the axial direction of the injection device in the direction of the ampoule 12. Thus, the force provided to the release member 13 will release accumulated energy in the torsion spring. This energy is converted to a translational movement of the piston rod towards the ampoule whereby a set dose of medicine can be injected from the injection device. FIG. 5 shows a cut half illustration of the housing 6 of the injection device. As seen, the drive member 8 comprises an engagement region/part 17 formed as gear wheel. Similarly, the housing 6 comprises a corresponding engagement region/part 16 adapted to receive and engage with the teeth of the gear wheel 17. FIG. 6 shows another embodiment of the present invention. In contrast to the embodiment shown in FIGS. 3-5 the embodiment shown in FIG. 6 contains no rotating exterior parts or elements. All rotating parts or elements are positioned inside the housing 19. FIG. 6 shows a release member 20 (formed as a push button) which is mechanically biased towards the end of the injection device by spring element 22. The release member 20 and dose setting member 21 are forced into engagement as long as the dose setting member 21 is in its dose setting position. The dose setting member 21 is mechanically biased towards the same end of the injection device as the release member 20 due to a spring element (shown as spring element 11 in FIG. 3) acting on the drive member (shown as drive member 8 in FIG. 3) which again acts on dose setting member 21. As seen in FIG. 6 the dose setting member 21 is biased against a mechanical stop 24 where a shoulder formed in the dose setting member 21 abuts a part of the housing 19. In FIG. 7 an intermediate stage is illustrated. Here the release member 20 has been pushed an axial distance sufficient to release the release member 20 from the dose setting member 21. Note that the engagement region 25 and 26 are disengaged, but since the shoulder of the dose setting member still abuts the housing part no axial movement of the dose setting member 21 has been achieved at this stage. Thus, the dose setting member 21 is prevented from rotating since the drive member (not shown) is still engaging the housing. In FIG. 8 the dose setting member 21 has been moved an axial distance towards the ampoule (not shown) whereby the dose setting member is allowed to rotate freely causing the piston rod 27 push a set dose of medicine out of the ampoule (not shown). Note that the release member 20 and the dose setting member 21 are disengaged in FIG. 8. This means that the release member 20 is not rotating relative to the housing during injection of a set dose. Then the set dose has been injected the user removes his thumb or index finger from the release member whereby the release member and the dose setting member return to their respective positions as illustrated in FIG. 6, but now with the spring element 23 being in a relaxed state. In case the user wants to set a new dose, the user rotates the release member which engages the dose setting member whereby the new dose can be set. Injecting the set dose is achieved by following the steps illustrated in FIGS. 7 and 8. FIGS. 9-13 show various embodiments of release mechanisms for releasing the energized power reservoir. In FIG. 9 a torsion spring (not shown) is energized by rotating a ratchet 28 which is operatively connected to the housing 30 of the injection device when the dose to be injected is being set. In the dose setting position the ratchet 28 is operatively connected with housing part 31 via ratchet arm 32. Energy accumulated in the torsion spring is released by displacing the ratchet 28 axially whereby it is released from its connection with housing part 31 in that the ratchet arm 32 is moved into housing part 33 whereby the piston rod 34 is allowed to rotate thereby expelling a set dose of medicament. In the embodiment depicted in FIG. 9 a dose indicator barrel (not shown) moves in the direction away from the push-button (not shown) during setting of a dose. Obviously, the dose indicator barrel may be adapted to move in the opposite direction during setting of a dose, i.e. towards the push-button. In the embodiment depicted in FIG. 10 the ratchet 35 is only in indirect operation with the housing 39. The drive member of the embodiment depicted in FIG. 10 is constituted by three part—one part 36 being adapted to corporate with the housing 39, another part 38 being adapted to drive the piston rod 40 and a flexible member 37 connecting parts 36 and 38. The flexible member 37 is flexible in the axial direction but establishes a substantially stiff connection between parts 36 and 38 when these parts are rotated relative to each other. Thus, the flexible member 37 ensures that parts 36 and 38 are not rotatably arranged relative to each other. Thus, when the ratchet 35 is moved towards the needle end of the injection device the part 36 is disconnected from the housing 39 whereby parts 36, 37 and 38 are allowed to rotate thereby rotating the piston rod 40. The rotating piston rod 40 causes a set dose of medicament to be expelled from the injection device. The embodiment depicted in FIG. 11 is similar to the embodiment in FIG. 9 except that the piston rod is moved forward by having guiding tracks arranged in the housing (instead of in the drive member) and a threaded engagement between piston rod and the drive member (instead of a threaded engagement between piston rod and housing). FIGS. 12 and 13 show other release mechanisms between ratchet, drive member and housing. 16452049 novo nordisk a/s USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open Apr 27th, 2022 08:32AM Apr 27th, 2022 08:32AM Novo Nordisk Health Care Pharmaceuticals & Biotechnology
nyse:nvo Novo Nordisk Apr 12th, 2022 12:00AM Jun 8th, 2018 12:00AM https://www.uspto.gov?id=US11298461-20220412 Basal titration with adaptive target glucose level Systems and methods are provided for adjusting long acting insulin medicament dosages for a subject. A plurality of timestamped glucose measurements of the subject is obtained. A titration glucose level (246) is computed as a measure of central tendency (244, 268, 274) of a titration subset of small glucose measurements (240, 269, 273) identified within the timestamped glucose measurements, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window. A long acting insulin medicament dosage (216) is adjusted or maintained based upon the obtained titration glucose level (246). 11298461 1. A device for adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, wherein the device comprises one or more processors and a memory, the memory comprising: a first data structure that includes the prescribed insulin regimen including a basal insulin medicament dosage regimen, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage, and instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement in the plurality of glucose measurements, a corresponding timestamp representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level based on small glucose measurements and the state of the insulin state indicator by: (i) obtaining a primary time window within the time course defining the period of time comprising the glucose measurements in the first data set to be used for identifying the titration glucose level, (ii) identifying a titration subset of small glucose measurements, identified as a subset of small glucose measurements within the primary time window, (iii) obtaining the titration glucose level computed as a measure of central tendency of the titration subset of small glucose measurements, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, (iv) associating the titration glucose level with the measure of central tendency; (C) adjusting or maintaining the long acting insulin medicament dosage based upon the obtained titration glucose level, and wherein the first data structure further comprises an insulin state indicator, wherein the insulin state indicator can indicate a short-acting-insulin-influence state, wherein the glucose measurements within the primary time window may be influenced by a short acting insulin medicament, and an only-long-acting-insulin-influence state, wherein the glucose measurements within the primary time window can be influenced by a long acting insulin medicament, but the measurements cannot be influenced by a short acting insulin medicament. 2. The device of claim 1, wherein the obtaining the titration glucose level, in step B, further comprises: based on the status of the insulin state indicator, selecting one of the following evaluation modes: (B1) for the primary time window in a first evaluation mode, (i) obtaining an integer defining the number of glucose measurements to be selected for the titration subset of small glucose measurements, (ii) identifying and selecting the titration subset of small glucose measurements as a subset of smallest glucose measurements, by identifying and selecting the smallest glucose measurements within the primary time window, and ensuring that the number of measurements within the titration subset of small glucose measurements equals the obtained integer, (iii) obtaining the measure of central tendency as a first glucose measure of central tendency, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements, and associating the titration glucose level with the first glucose measure of central tendency, (B2) for the primary time window in a second evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows within the primary time window, wherein each secondary time window comprises a subset of overlapping glucose measurements being a subset of the glucose measurements in the primary time window, for each secondary time window within the plurality of secondary time windows, computing a corresponding second glucose measure of central tendency, and thereby obtaining a plurality of second glucose measures of central tendency, wherein each respective second glucose measure of central tendency is computed as a measure of central tendency of the glucose measurements within the corresponding secondary time window, for the plurality of second glucose measures of central tendency, identifying a smallest second glucose measure of central tendency as the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency, whereby the titration subset of small glucose measurements is identified as the subset comprising the glucose measurements within the secondary time window corresponding to the smallest second glucose measure of central tendency, and associating the titration glucose level with the smallest second glucose measure of central tendency, or (B3) for the primary time window in a third evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows within the primary time window, wherein each secondary time window comprises a subset of overlapping glucose measurements being a subset of the glucose measurements in the primary time window, for each secondary time window within the plurality of secondary time windows, computing a corresponding glucose measure of variability, and thereby obtaining a plurality of glucose measures of variability, wherein each respective glucose measure of variability is computed as a measure of variability of the glucose measurements within the corresponding secondary time window, for the plurality of glucose measures of variability, identifying a smallest glucose measure of variability as the smallest glucose measure of variability within the plurality of glucose measures of variability, whereby the titration subset of small glucose measurements is identified as the subset comprising the glucose measurements within the secondary time window corresponding to the smallest glucose measure of variability, and computing a smallest third glucose measure of central tendency as a measure of central tendency of the titration subset of small glucose measurements, and associating the titration glucose level with the smallest third glucose measure of central tendency. 3. The device of claim 2, wherein the secondary time window is 50 minutes to 70 minutes, 60 minutes to 120 minutes, 120 minutes to 180 minutes, or 180 minutes to 300 minutes. 4. The device of claim 2, wherein successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 4 minutes to 6 minutes, and wherein the secondary time window is 180 minutes to 300 minutes. 5. The device of claim 1, wherein the method further comprises: in response to identifying the state of the insulin state indicator, selecting the first or the second evaluation mode, upon the occurrence that the state of the insulin state indicator is identified as the only-long-acting-insulin-influence state, and thereby using a preferred method for obtaining the titration glucose level, which is preferred for titration with a long acting insulin medicament, when it is ensured that no short acting insulin medicament influences the glucose measurements. 6. The device of claim 1, wherein the method further comprises: in response to identifying the state of the insulin state indicator, selecting the third evaluation mode, upon the occurrence that the state of the insulin state indicator is identified as the a short-acting-insulin-influence state, and thereby using a preferred method for obtaining the titration glucose level, which is preferred for titration with a long acting insulin medicament, when it is identified that short acting insulin medicament may influence the glucose measurements. 7. The device of claim 1, wherein the method further comprises: obtaining a second data set from one or more insulin pens used by the subject to apply the prescribed insulin regimen, the second data set comprising a plurality of insulin medicament records over the time course, each insulin medicament record in the plurality of medicament records comprising: (i) a respective insulin medicament injection event representing an insulin medicament injection into the subject using a respective insulin pen in the one or more insulin pens, (ii) a corresponding electronic timestamp that is automatically generated by the respective insulin pen upon occurrence of the respective insulin medicament injection event, and (iii) the type of insulin medicament indicating whether the injected insulin medicament is a short acting insulin medicament type or a long acting insulin medicament type. 8. The device according to claim 7, wherein the prescribed insulin regimen further comprises a duration of action for the short acting insulin medicament, wherein the duration of action of a medicament specifies the duration that a measurable medicament effect persist, and thereby influences the glucose level. 9. The device according to claim 7, wherein the prescribed insulin regimen comprises a duration of action for the long acting insulin medicament, wherein the duration of action of a medicament specifies the duration that a measurable medicament effect persist, and thereby influences the glucose level. 10. The device of claim 7, wherein the insulin state indicator indicates the short-acting-insulin-influence state based on the second data set and the duration of action for the short acting insulin medicament. 11. The device of claim 7, wherein the insulin state indicator indicates the only-long-acting-insulin-influence state based on the second data set and the duration of action for the short acting insulin medicament. 12. The device of claim 1, wherein the prescribed insulin regimen further comprises a bolus insulin medicament dosage regimen, wherein the bolus insulin medicament dosage regimen specifies the short acting insulin dosage. 13. The device of claim 1, wherein the first data structure further comprises a hypoglycemic risk state indicator, wherein the hypoglycemic risk state indicator can indicate a high hypoglycemic risk state, wherein the subject may have a high hypoglycemic risk or wherein a high variability across the plurality of glucose measurements can be observed, and a non-high hypoglycemic risk state, wherein the subject may have a non-high hypoglycemic risk or wherein a low variability across the plurality of glucose measurements can be observed, and wherein the method further comprises: in response to identifying the state of the hypoglycemic risk state indicator, selecting the first evaluation mode, upon the occurrence that the state of the hypoglycemic risk state indicator is identified as the high hypoglycemic risk state, and thereby using a method for obtaining the titration glucose level which is more sensitive to low glucose values and noise. 14. A method for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, at a computer comprising one or more processors and a memory: the memory storing: a first data structure that includes the prescribed insulin regimen including a basal insulin medicament dosage regimen, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage, and the memory further storing instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement in the plurality of glucose measurements, a corresponding timestamp representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level based on small glucose measurements and the state of the insulin state indicator by: (i) obtaining a primary time window within the time course defining the period of time comprising the glucose measurements in the first data set to be used for identifying the titration glucose level, (ii) identifying a titration subset of small glucose measurements, identified as a subset of small glucose measurements within the primary time window, (iii) obtaining the titration glucose level computed as a measure of central tendency of the titration subset of small glucose measurements, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, (iv) associating the titration glucose level with the measure of central tendency; (C) adjusting or maintaining the long acting insulin medicament dosage based upon the obtained titration glucose level, and wherein an insulin state indicator, wherein the insulin state indicator can indicate a short-acting-insulin-influence state, wherein the glucose measurements within the primary time window may be influenced by a short acting insulin medicament, and an only-long-acting-insulin-influence state, wherein the glucose measurements within the primary time window can be influenced by a long acting insulin medicament, but the measurements cannot be influenced by a short acting insulin medicament. 15. A computer program comprising instructions that, when executed by a computer having one or more processors and a memory, perform the method of claim 14. 15 CROSS-REFERENCE TO RELATED APPLICATIONS This application is a 35 U.S.C. § 371 National Stage application of International Application PCT/EP2018/065126 (published as WO 2018/228932), filed Jun. 8, 2018, which claims priority to European Patent Applications 17178877.1, filed Jun. 29, 2017 and 17190146.5, filed Sep. 8, 2017, this application further claims priority under 35 U.S.C. § 119 of U.S. Provisional Applications 62/520,139, filed Jun. 15, 2017 and 62/522,811, filed Jun. 21, 2017, the contents of all above-named applications are incorporated herein by reference. TECHNICAL FIELD The present disclosure relates generally to systems, computer programs and computer-readable data carriers storing a computer program and methods for adjusting or maintaining a long acting insulin medicament dosage in a prescribed insulin regimen for a subject based on a titration subset of small glucose measurements identified as a subset of small glucose measurements within a plurality of glucose measurements of the subject taken over a time course. BACKGROUND Type 2 diabetes mellitus is characterized by progressive disruption of normal physiologic insulin secretion. In healthy individuals, basal insulin secretion by pancreatic β cells occurs continuously to maintain steady glucose levels for extended periods between meals. Also in healthy individuals, there is prandial secretion in which insulin is rapidly released in an initial first-phase spike in response to a meal, followed by prolonged insulin secretion that returns to basal levels after 2-3 hours. Insulin is a hormone that binds to insulin receptors to lower blood glucose by facilitating cellular uptake of glucose, amino acids, and fatty acids into skeletal muscle and fat and by inhibiting the output of glucose from the liver. In normal healthy individuals, physiologic basal and prandial insulin secretions maintain euglycemia, which affects fasting plasma glucose and postprandial plasma glucose concentrations. Basal and prandial insulin secretion is impaired in Type 2 diabetes and early post-meal response is absent. To address these adverse events, patients with Type 2 diabetes are provided with insulin medicament treatment regimens. Patients with Type 1 diabetes are also provided with insulin medicament treatment regimens. The goal of these insulin medicament treatment regimens is to maintain a desired fasting blood glucose target level that will minimize estimated risk of hypo- and hyperglycaemia. Smart titrators with adjustable step size and physiological parameter estimation and predefined fasting blood glucose target values have been developed to administer insulin medicament treatment regimens. A Continuous Glucose Monitor (CGM) is a small wearable device that tracks glucose levels during the day and night, and which can notify when glucose levels are too high or too low, enabling the patient to take action. The current standard of care is that patients self-monitor the blood glucose by repeated finger sticks and SMBG. However, due to the discrete nature of this practice, high or low blood glucose events may be missed, and the actual measurement time point may differ from the recommended (i.e. fasting blood glucose measured in a non-fasting state) and there may be barriers to adhere to the recommend number of measurements, see T. Walker, “The Rationale for Continuous Glucose Monitoring-based Diabetes Treatment Decisions and Non-adjunctive Continuous Glucose Monitoring Use”, European Endocrinology, 2016; 12(1): 24-30. In general, the point estimate accuracy of blood glucose meters is likely to be better compared to point estimates from CGM. Under normal circumstances, diabetes patients use SMBG to measure the fasting glucose level when they are still fasting before breakfast. However, sometimes the measurement is forgotten and either not done, or done after breakfast has been initiated, which reduces the precision of the fasting glucose level used for basal insulin titration. Automatic detection of the glucose level from CGM data to base basal insulin titration on is anticipated to reduce the risk of this use error, as CGM data are time stamped. In addition, basal insulin titration based on the morning fasting glucose level may lead to excessive insulin dosing due to the raising glucose levels in the morning due to the dawn phenomenon, see S. Wolfe, “Contribution of the dawn phenomenon to the fasting and postbreakfast hyperglycaemia in type 1 diabetes treated with once-nightly insulin glargine”, Endocr. Pract., year, 18, pp. 558-562, 2012. U.S. Pat. No. 8,370,077 B2 entitled “System for Optimizing A Patient's Inulin Dosage Regimen” to Hygieia, Inc. discloses a system for optimizing a patient's insulin dosage regimen over time in which inputs corresponding at least to one or more components in a patient's present insulin dosage regimen, and data inputs corresponding at least to the patient's blood-glucose-level measurements determined at a plurality of times. From the data inputs corresponding to the patient's blood-glucose-level measurements, determined at a plurality of times, a determination is made as to whether and by how much to vary at least one of the one or more components in the patient's present insulin dosage regimen in order to maintain the patient's future blood-glucose-level measurements within a predefined range. The blood-glucose-level measurements are tagged with an identifier reflective of when the reading was input; specifically, whether it is a morning measurement, a pre-lunch measurement, a pre-dinner measurement, a bedtime measurement, or a nighttime measurement. United States Publication No. 2011/313674 A entitled “Insulin Optimization Systems and Testing Methods with Adjusted Exit Criterion Accounting for System Noise Associated with Biomarkers” to Roche Diagnostics Operations, Inc., discloses a method for optimizing a therapy to a diabetic patient comprising collecting at least one sampling set of biomarker data. The diabetic patient may begin collection of one or more sampling sets of biomarker data, wherein each sampling set comprises one or more sampling instances recorded over a collection period. Each sampling instance comprises one or more biomarker readings. The collection period for the sampling set may be defined as multiple sampling instances within a day, multiple sampling instances within a week, multiple sampling instances within consecutive weeks, or multiple sampling instances on consecutive days within a week. The biomarker may relate to the levels of glucose, triglycerides, low density lipids, and high density lipids. In one exemplary embodiment, the biomarker reading is a blood glucose reading, specifically a fasting blood glucose reading. In addition to the biomarker reading, each sampling instance may comprise the biomarker reading and other contextual data associated with the biomarker reading, wherein the contextual data is selected from the group consisting of the time of collection, the date of collection, the time when the last meal was consumed, the recommended dose of insulin, and combinations thereof. United States Publication No. 2011/0319322 entitled “Systems, Methods and Devices for Achieving Glycemic Balance” to Hygieia, discloses a system for optimizing a patient's insulin dosage regimen over time, comprising at least a first memory for storing data inputs corresponding at least to one or more components in a patient's present insulin dosage regimen, and data inputs corresponding at least to the patient's blood-glucose-level measurements determined at a plurality of times, and a processor operatively connected to the at least first memory. The processor is programmed at least to determine from the data inputs corresponding to the patient's blood-glucose-level measurements determined at a plurality of times whether and by how much to vary at least one of the one or more components in the patient's present insulin dosage regimen in order to maintain the patient's future blood-glucose-level measurements within a predefined range. United States Publication No. 2012/0232520 entitled “Multi-Function Analyte Monitor Device and Methods of use” to Abbott Diabetes Care, Inc, discloses methods, systems and devices for detecting an analyte sample, determining an analyte concentration associated with the detected analyte sample, storing the determined analyte concentration and a time associated with the determined analyte concentration, retrieving two or more stored analyte concentrations, and determining an adjusted dose level based at least in part on a current dose level and data associated with the two or more retrieved analyte concentrations are provided. For example, adjustments to dosage levels of long-acting insulin may be provided to assist in the management of diabetes and related conditions In basal insulin therapy, robust and reliable insulin titration algorithms are important, and the glucose levels used to calculate the new dose is important, and given the above background, what is needed in the art are systems and methods that provide improved insulin medicament titration. SUMMARY The present disclosure addresses the need in the art for systems and methods for providing improved insulin medicament titration. The input glucose level used for adjusting the amount of medicament should reflect the critical glucose levels, important for determining the optimal daily basal dose or amount of medicament. Since low glucose values are critical, they are also critical in finding the optimal daily basal dose. In this description, we refer to the glucose level used as input to a titration algorithm as the titration glucose level (TGL). A problem to be solved by the present disclosure is therefore to safely and automatically detect TGL based on data from a continuous glucose monitor (CGM). The continuous or near continuous character of CGM data or autonomously generated time stamped glucose data gives an opportunity for a more refined and dynamic detection of the glucose level to base the basal insulin titration on. In a further aspect TGL is also based on an indication of whether or not an exogenous short acting insulin medicament has been injected and influences the blood glucose level on a short term. A further aspect of the present invention is the provision of systems and methods for safe and automatic detection of a titration glucose level based on continuous or near continuous glucose data. With continuous glucose monitors becoming cheaper and more accurate, it is estimated that the technology will become more widely used, by T1 and T2 diabetes patients. The invention allows determining TGL based on CGM data, which is useful in e.g. basal insulin titration for T2D patients. In one aspect, using the lowest average glucose of CGM data over a period of e.g. 24 hours provides a number of benefits in safety, the lowest glucose readings are the critical values in insulin treatment with a long acting insulin, as opposed to using fasting SMPG measurements, where SMPG measurements only reflect one point in time, and therefore do not ensure capture the lowest fasting glucose. An example is the dawn-effect which causes fasting glucose to rise before waking up, and hence the morning fasting SMPG measurement is higher than the fasting glucose values during the night. As opposed to predefined fasting periods fasting periods vary in real-life and therefore pre-defining a period as fasting is not robust. In an aspect of the present invention, CGM data is analyzed using a set of rules, enabling a fasting-glucose-equivalent to be determined automatically from the data stream. The value, coined “Titration Glucose Level,” is determined by filtering out data that is of inadequate quality, that is affected by prandial artifacts, and that is influenced by insulin on board. Accordingly, one aspect of the present disclosure provides a device for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, wherein the device comprises one or more processors and a memory, the memory comprising: a first data structure that includes the prescribed insulin regimen including a basal insulin medicament dosage regimen, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage, an insulin state indicator, wherein the insulin state indicator can indicate a short-acting-insulin-influence state, wherein the glucose measurements within the primary time window may be influenced by a short acting insulin medicament, and an only-long-acting-insulin-influence state, wherein the glucose measurements within the primary time window can be influenced by a long acting insulin medicament, but the measurements cannot be influenced by a short acting insulin medicament, and instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement in the plurality of glucose measurements, a corresponding timestamp representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level based on small glucose measurements and the state of the insulin state indicator by: (i) obtaining a primary time window within the time course defining the period of time comprising the glucose measurements in the first data set to be used for identifying the titration glucose level, (ii) identifying a titration subset of small glucose measurements, identified as a subset of small glucose measurements within the primary time window, (iii) obtaining the titration glucose level computed as a measure of central tendency of the titration subset of small glucose measurements, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level with the measure of central tendency; (C) adjusting or maintaining the long acting insulin medicament dosage based upon the obtained titration glucose level. Hereby is provided a device that automatically obtains a glucose titration level which can be used as input in an algorithm for adjusting a long acting insulin dose based on the obtained glucose titration level, and wherein the evaluation of the glucose titration level is optimized with respect to characteristics of the blood glucose profile, i.e., whether the blood glucose profile indicates influence of exogenous short acting insulin or not. In other words the evaluation of the titration glucose level is automatic and it adapts to the medical regimen that the subject is following or supposed to follow. The provided device analyzes CGM data by using a set of rules, and thereby enables an automatic determination of a titration glucose level from the data stream. The value of the titration glucose level, is determined by filtering out data that is of inadequate quality, that is affected by prandial artifacts, and that is influenced by insulin on board. In this way the titration subset of small glucose measurements, wherein the small glucose measurements are small, primarily due to the influence of the long-acting insulin medicament, can be identified based on the insulin state indicator. If the insulin state indicator identifies influence by a short acting insulin the small glucose measurements, will be found in a period spaced in time from intake of carbohydrates and bolus insulin, i.e., a fasting period. In a further aspect the obtaining the titration glucose level, in step B, further comprises: based on the status of the insulin state indicator, selecting one of the following evaluation modes: (B1) for the primary time window in a first evaluation mode, (i) obtaining an integer defining the number of glucose measurements to be selected for the titration subset of small glucose measurements, (ii) identifying and selecting the titration subset of small glucose measurements as a subset of smallest glucose measurements, by identifying and selecting the smallest glucose measurements within the primary time window, and ensuring that the number of measurements within the titration subset of small glucose measurements equals the obtained integer, (iii) obtaining the measure of central tendency as a first glucose measure of central tendency, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements, and associating the titration glucose level with the first glucose measure of central tendency, (B2) for the primary time window in a second evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows within the primary time window, wherein each secondary time window comprises a subset of overlapping glucose measurements being a subset of the glucose measurements in the primary time window, for each secondary time window within the plurality of secondary time windows, computing a corresponding second glucose measure of central tendency, and thereby obtaining a plurality of second glucose measures of central tendency, wherein each respective second glucose measure of central tendency is computed as a measure of central tendency of the glucose measurements within the corresponding secondary time window, and thereby obtaining a moving or running period of a measure of central tendency across the glucose measurements in the primary time window, for the plurality of second glucose measures of central tendency, identifying a smallest second glucose measure of central tendency as the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency, whereby the titration subset of small glucose measurements is identified as the subset comprising the glucose measurements within the secondary time window corresponding to the smallest second glucose measure of central tendency, and associating the titration glucose level with the smallest second glucose measure of central tendency, or (B3) for the primary time window in a third evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows within the primary time window, wherein each secondary time window comprises a subset of overlapping glucose measurements being a subset of the glucose measurements in the primary time window, for each secondary time window within the plurality of secondary time windows, computing a corresponding glucose measure of variability, and thereby obtaining a plurality of glucose measures of variability, wherein each respective glucose measure of variability is computed as a measure of variability of the glucose measurements within the corresponding secondary time window, and thereby obtaining a moving period of a measure of variability across the glucose measurements in the primary time window, for the plurality of glucose measures of variability, identifying a smallest glucose measure of variability as the smallest glucose measure of variability within the plurality of glucose measures of variability, whereby the titration subset of small glucose measurements is identified as the subset comprising the glucose measurements within the secondary time window corresponding to the smallest glucose measure of variability, and computing a smallest third glucose measure of central tendency as a measure of central tendency of the titration subset of small glucose measurements, and associating the titration glucose level with the smallest third glucose measure of central tendency. In a further aspect the method further comprises: in response to identifying the state of the insulin state indicator, selecting the first or the second evaluation mode, upon the occurrence that the state of the insulin state indicator is identified as the only-long-acting-insulin-influence state, and thereby using a preferred method for obtaining the titration glucose level, which is preferred for titration with a long acting insulin medicament, when it is ensured that no short acting insulin medicament influences the glucose measurements. In a further aspect the method further comprises: in response to identifying the state of the insulin state indicator, selecting the third evaluation mode, upon the occurrence that the state of the insulin state indicator is identified as the a short-acting-insulin-influence state, and thereby using a preferred method for obtaining the titration glucose level, which is preferred for titration with a long acting insulin medicament, when it is identified that short acting insulin medicament may influence the glucose measurements. In a further aspect, the measure of variability is the variance, and in a further aspect, the measure of central tendency is the mean value. In a further aspect, the method is repeated on a recurring basis. In a further aspect, the method further comprises: obtaining a second data set from one or more insulin pens used by the subject to apply the prescribed insulin regimen, the second data set comprising a plurality of insulin medicament records over the time course, each insulin medicament record in the plurality of medicament records comprising: (i) a respective insulin medicament injection event representing an insulin medicament injection into the subject using a respective insulin pen in the one or more insulin pens and (ii) a corresponding electronic timestamp that is automatically generated by the respective insulin pen upon occurrence of the respective insulin medicament injection event; for the first glucose measures of central tendency in the first evaluation mode, associating the first glucose measure of central tendency with a tertiary time window representing an evaluation period, wherein a most recent end point of the tertiary time window is synchronized with a most recent end point of the primary time window, and wherein the primary and the tertiary windows are of the same length, for the plurality of second glucose measures of central tendency in the second evaluation mode, associating each respective second glucose measure of central tendency with a time indicator representing the time of evaluation of the respective second glucose measure of central tendency, and thereby obtaining a plurality of time indicators defining a tertiary time window representing an evaluation period, wherein a most recent end point of the tertiary time window is synchronized with a most recent end point of the primary time window, and wherein the length of the tertiary time window is smaller than the length of the primary time window, or for the plurality of glucose measures of variability in the third evaluation mode, associating each respective glucose measure of variability with a time indicator representing the time of evaluation of the respective glucose measure of variability, and thereby obtaining a plurality of time indicators defining a tertiary time window representing an evaluation period, wherein a most recent end point of the tertiary time window is synchronized with a most recent end point of the primary time window, and wherein the length of the tertiary time window is smaller than the length of the primary time window; and associating the titration glucose level with the tertiary time window; applying a first characterization to the tertiary time window, wherein the first characterization is one of basal regimen adherent and basal regimen nonadherent, the tertiary time window is deemed basal regimen adherent when the second data set includes one or more medicament records that establish, on a temporal and quantitative basis, adherence with the prescribed basal insulin medicament dosage regimen during the respective tertiary time window, and the tertiary time window is deemed basal regimen nonadherent when the second data set fails to include one or more medicament records that establish, on a temporal and quantitative basis, adherence with the prescribed basal insulin medicament dosage regimen during the tertiary time window; and wherein the adjusting the long acting insulin medicament dosage in the basal insulin medicament dosage regimen for the subject is based upon a titration glucose level that is represented by a tertiary time window that is deemed basal regimen adherent and by excluding a titration glucose level that is represented by a tertiary time window that is deemed basal regimen nonadherent. In a further aspect, the first data structure comprises, a plurality of consecutive epochs, wherein each respective epoch is associated with a basal insulin medicament dosage, indicating when the basal insulin medicament is to be injected within the respective epoch, and how much of the basal insulin medicament is to be injected, and thereby providing a temporal and quantitative basis for the first characterization. In a further aspect the length of the tertiary window is longer than or the same as the length of each of the epochs. In a further aspect, the end point of the tertiary time window is synchronized with an end point of a current epoch, wherein the current epoch is the most recent completed epoch within the plurality of epochs. In a further aspect, each respective epoch of the plurality of epochs is associated with a tertiary time window, and thereby obtaining a plurality of tertiary time windows, wherein each tertiary time window represents an evaluation period, wherein each tertiary window is aligned with the respective epoch on a temporal bases, and wherein each tertiary time window is associated with a titration glucose level. In a further aspect, the first data structure comprises a specification of temporal and quantitative basis for administration of the long acting insulin medicament, for each of the epochs within the plurality of epochs. In a further aspect, the quantitative basis for the long acting insulin medicament is a function of the titration glucose level. In a further aspect, the temporal basis is specified as one injection for each epoch within the plurality of epochs. In a further aspect each epoch in the plurality of epochs is a calendar day or a calendar week. In a further aspect successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less. In a further aspect, the device further comprises a wireless receiver, and wherein the first data set is obtained wirelessly from a glucose sensor affixed to the subject and/or the second data set is obtained wirelessly from the one or more insulin pens. In a further aspect, the first data structure further comprises a hypoglycemic risk state indicator, wherein the hypoglycemic risk state indicator can indicate a high hypoglycemic risk state, wherein the subject may have a high hypoglycemic risk or wherein a high variability across the plurality of glucose measurements can be observed, and a non-high hypoglycemic risk state, wherein the subject may have a non-high hypoglycemic risk or wherein a low variability across the plurality of glucose measurements can be observed, and wherein the method further comprises: in response to identifying the state of the hypoglycemic risk state indicator, selecting the first evaluation mode, upon the occurrence that the state of the hypoglycemic risk state indicator is identified as the high hypoglycemic risk state, and thereby using a method for obtaining the titration glucose level (246) which is more sensitive to low glucose values and noise. In a further aspect, the secondary time window is 50 minutes to 70 minutes, 60 minutes to 120 minutes, 120 minutes to 180 minutes or 180 minutes to 300 minutes. In a further aspect, successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 4 minutes to 6 minutes, and wherein the secondary time window is 50 minutes to 70 minutes. In a further aspect, successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 40 minutes to 80 minutes, and wherein the secondary time window is 180 minutes to 310 minutes. In another aspect is provided, a method for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, at a computer comprising one or more processors and a memory: the memory storing: a first data structure that includes the prescribed insulin regimen including a basal insulin medicament dosage regimen, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage, an insulin state indicator, wherein the insulin state indicator can indicate a short-acting-insulin-influence state, wherein the glucose measurements within the primary time window may be influenced by a short acting insulin medicament, and an only-long-acting-insulin-influence state, wherein the glucose measurements within the primary time window can be influenced by a long acting insulin medicament, but the measurements cannot be influenced by a short acting insulin medicament, and the memory further storing instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement in the plurality of glucose measurements, a corresponding timestamp representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level based on small glucose measurements and the state of the insulin state indicator by: (i) obtaining a primary time window within the time course defining the period of time comprising the glucose measurements in the first data set to be used for identifying the titration glucose level, (ii) identifying a titration subset of small glucose measurements, identified as a subset of small glucose measurements within the primary time window, (iii) obtaining the titration glucose level computed as a measure of central tendency of the titration subset of small glucose measurements, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level with the measure of central tendency; (C) adjusting or maintaining the long acting insulin medicament dosage based upon the obtained titration glucose level. In another aspect is provided a computer program comprising instructions that, when executed by a computer having one or more processors and a memory, perform the method described above. In another aspect is provided a computer-readable data carrier having stored thereon the computer program as described above. In another aspect is provided a device for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, wherein the device comprises one or more processors and a memory, the memory comprising: a first data structure that includes the prescribed insulin regimen including a basal insulin medicament dosage regimen, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage, and instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement in the plurality of glucose measurements, a corresponding timestamp representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level by: (i) obtaining a primary time window within the time course defining the period of time comprising the glucose measurements in the first data set to be used for identifying the titration glucose level, (ii) identifying a titration subset of small glucose measurements, identified as a subset of small glucose measurements within the primary time window, (iii) obtaining the titration glucose level computed as a measure of central tendency of the titration subset of small glucose measurements, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level with the measure of central tendency; (C) adjusting or maintaining the long acting insulin medicament dosage based upon the obtained titration glucose level. In a further aspect of the device, the obtaining the titration glucose level, in step B, further comprises: selecting one of the following evaluation modes: (B1) for the primary time window in a first evaluation mode, (i) obtaining an integer defining the number of glucose measurements to be selected for the subset of glucose measurements, (ii) identifying and selecting the titration subset of small glucose measurements as a subset of smallest glucose measurements, by identifying and selecting the smallest glucose measurements within the primary time window, and ensuring that the number of measurements within the titration subset of small glucose measurements equals the obtained integer, (iii) obtaining the measure of central tendency as a first glucose measure of central tendency, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements, and associating the titration glucose level with the first glucose measure of central tendency, (B2) for the primary time window in a second evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows within the primary time window, wherein each secondary time window comprises a subset of overlapping glucose measurements being a subset of the glucose measurements in the primary time window, for each secondary time window within the plurality of secondary time windows, computing a corresponding second glucose measure of central tendency, and thereby obtaining a plurality of second glucose measures of central tendency, wherein each respective second glucose measure of central tendency is computed as a measure of central tendency of the glucose measurements within the corresponding secondary time window, and thereby obtaining a moving period of a measure of central tendency across the glucose measurements in the primary time window, for the plurality of second glucose measures of central tendency, identifying a smallest second glucose measure of central tendency as the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency, whereby the titration subset of small glucose measurements is identified as the subset comprising the glucose measurements within the secondary time window corresponding to the smallest second glucose measure of central tendency, and associating the titration glucose level with the smallest second glucose measure of central tendency, or (B3) for the primary time window in a third evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows within the primary time window, wherein each secondary time window comprises a subset of overlapping glucose measurements being a subset of the glucose measurements in the primary time window, for each secondary time window within the plurality of secondary time windows, computing a corresponding glucose measure of variability, and thereby obtaining a plurality of glucose measures of variability, wherein each respective glucose measure of variability is computed as a measure of variability of the glucose measurements within the corresponding secondary time window, and thereby obtaining a moving period of a measure of variability across the glucose measurements in the primary time window, for the plurality of glucose measures of variability, identifying a smallest glucose measure of variability as the smallest glucose measure of variability within the plurality of glucose measures of variability, whereby the titration subset of small glucose measurements is identified as the subset comprising the glucose measurements within the secondary time window corresponding to the smallest glucose measure of variability, and computing a smallest third glucose measure of central tendency as a measure of central tendency of the titration subset of small glucose measurements, and associating the titration glucose level with the smallest third glucose measure of central tendency. Hereby is provided a device that automatically obtain a glucose titration level which can be used as input in an algorithm for adjusting a long acting insulin dose based on the obtained glucose titration level, and wherein the evaluation of the glucose titration level can be optimized with respect characteristics of the blood glucose profile, i.e., the evaluation can be optimized if the blood glucose profile indicates influence of exogenous short acting insulin or not. In other words the evaluation of the titration glucose level is automatic and the evaluation can be made dependent on the medical regimen that the subject is following or supposed to follow. In a further aspect the glucose measurements comprised in the titration subset of small glucose measurements have values smaller than a lower percentile of the glucose measurements, wherein the lower percentile ranges from the 0.1th percentile to the 50th percentile, wherein a Pth percentile is defined as the lowest glucose measurement that is greater than P % of the glucose measurements in the first data set to be used for identifying the titration subset within the primary time window In a further aspect is provided, a basal titration adjustment device for autonomously adjusting a long acting insulin medicament dosage in a prescribed basal insulin regimen for a subject, wherein the device comprises one or more processors and a memory, the memory comprising: a prescribed insulin regimen including a basal insulin medicament dosage regimen, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage, and instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement in the plurality of glucose measurements, a corresponding timestamp representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level being the glucose level used as input to an algorithm for maintaining or adjusting the long acting insulin medicament dosage, wherein the titration glucose level is based on a titration subset of small glucose measurements, by: (i) obtaining a primary time window within the time course defining the period of time comprising the glucose measurements in the first data set to be used for identifying the titration subset of small glucose measurements and for obtaining the titration glucose level for the primary time window, wherein each of the glucose measurements has a timestamp 232 within the primary time window, (ii) identifying the titration subset of small glucose measurements identified as a subset of small glucose measurements within the primary time window, (iii) obtaining the titration glucose level 246, computed as a measure of central tendency of the titration subset of small glucose measurements wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level 246 with the measure of central tendency by assigning the value of measure of central tendency to the titration glucose level; (C) adjusting or maintaining the long acting insulin medicament dosage based upon the obtained titration glucose level. Hereby is provided a device adapted for adjusting or maintaining the long actin insulin medicament dosage based upon the obtained titration glucose level for a prescribed basal insulin regimen, i.e., the subject is treated with long acting insulin only, and we hereby know that the lowest glucose levels arise from the influence of the long acting insulin medicament. In a further aspect, the obtaining the titration glucose level, in step B, further comprises: selecting one of the following evaluation modes: (B1) for the primary time window in a first evaluation mode, (i) obtaining an integer defining the number of glucose measurements to be selected for the subset of glucose measurements, (ii) identifying and selecting the titration subset of small glucose measurements as a subset of smallest glucose measurements, by identifying and selecting the smallest glucose measurements within the primary time window, and ensuring that the number of measurements within the titration subset of small glucose measurements equals the obtained integer, (iii) obtaining the measure of central tendency as a first glucose measure of central tendency, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements, and associating the titration glucose level with the first glucose measure of central tendency. In a further aspect, the obtaining the titration glucose level, in step B, further comprises: (B2) for the primary time window in a second evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows within the primary time window, wherein each secondary time window comprises a subset of overlapping glucose measurements being a subset of the glucose measurements in the primary time window, for each secondary time window within the plurality of secondary time windows, computing a corresponding second glucose measure of central tendency, and thereby obtaining a plurality of second glucose measures of central tendency, wherein each respective second glucose measure of central tendency is computed as a measure of central tendency of the glucose measurements within the corresponding secondary time window, and thereby obtaining a moving period of a measure of central tendency across the glucose measurements in the primary time window, for the plurality of second glucose measures of central tendency, identifying a smallest second glucose measure of central tendency as the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency, whereby the titration subset of small glucose measurements is identified as the subset comprising the glucose measurements within the secondary time window corresponding to the smallest second glucose measure of central tendency, and associating the titration glucose level with the smallest second glucose measure of central tendency. In a further aspect, the obtaining the titration glucose level, in step B, further comprises: obtaining a percentage defining a titration percentile defining the number of glucose measurements to be selected for the titration subset of small glucose measurements, (ii) identifying and selecting the titration subset of small glucose measurements as a subset of smallest glucose measurements, by identifying and selecting the glucose measurements defined by the titration percentile of the glucose measurements, wherein the titration percentile ranges from the 0.1th percentile to the 50th percentile, and is smaller than or equal to the lower percentile, and wherein a Pth percentile is defined as the lowest glucose measurement that is greater than P % of the glucose measurements in the first data set to be used for identifying the titration subset within the primary time window, (iii) obtaining the measure of central tendency as a fourth glucose measure of central tendency, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements, and associating the titration glucose level with the fourth glucose measure of central tendency, by assigning the value of the fourth measure of central tendency to the titration glucose level. In a further aspect is provided, a basal titration adjustment device for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, wherein the device comprises one or more processors and a memory, the memory comprising: a prescribed insulin regimen including a basal insulin medicament dosage regimen, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage, and instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement in the plurality of glucose measurements, a corresponding timestamp representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level being the glucose level used as input to an algorithm for maintaining or adjusting the long acting insulin medicament dosage, wherein the titration glucose level is based on a titration subset of small glucose measurements, by: (i) obtaining a primary time window within the time course defining the period of time comprising the glucose measurements in the first data set to be used for identifying the titration subset of small glucose measurements and for obtaining the titration glucose level for the primary time window, wherein each of the glucose measurements has a timestamp within the primary time window, (ii) identifying the titration subset of small glucose measurements identified as a subset of small glucose measurements within the primary time window (iii) obtaining the titration glucose level, computed as a measure of central tendency of the titration subset of small glucose measurements, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level with the measure of central tendency, by assigning the value of measure of central tendency to the titration glucose level; (C) adjusting or maintaining the long acting insulin medicament dosage based upon the obtained titration glucose level. BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 illustrates an exemplary system topology that includes a basal titration adjustment device for automatically adjusting, maintaining or optimizing a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, a data collection device for collecting patient data, one or more glucose sensors that measure glucose data from the subject, and one or more insulin pens that are used by the subject to inject insulin medicaments in accordance with the prescribed insulin medicament regimen, where the above-identified components are interconnected, optionally through a communications network, in accordance with an embodiment of the present disclosure. FIGS. 2A, 2B, 2C and 2D collectively illustrate a basal titration adjustment device for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen in accordance with an embodiment of the present disclosure. FIGS. 3A, 3B, 3C and 3D collectively illustrate a basal titration adjustment device for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen in accordance with another embodiment of the present disclosure. FIGS. 4A, 4B, 4C, and 4D collectively provide a flow chart of processes and features of a basal titration adjustment device for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen in accordance with various embodiments of the present disclosure. FIG. 5 illustrates an example integrated system of connected insulin pen(s), continuous glucose monitor(s), memory and a processor for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen in accordance with an embodiment of the present disclosure. FIG. 6A illustrates the temporal relationship between a primary time window, secondary time windows, a tertiary time window, an epoch and time course wherein the plurality of glucose measurements have been obtained in accordance with embodiments of the present disclosure. FIG. 6B illustrates the steps of identifying a titration subset of smallest glucose measurements within a primary time window, and how to obtain the corresponding glucose titration level in accordance with an embodiment of the present disclosure. The illustrated method is particularly suitable, when glucose levels are not influenced by short acting insulin. FIG. 6C illustrates the steps of identifying a titration subset of small glucose measurements within a primary time window, and the step of obtaining the corresponding glucose titration level in accordance with another embodiment of the present disclosure. The illustrated method is particularly suitable, when glucose levels are not influenced by short acting insulin. FIG. 6D illustrates the steps of identifying a titration subset of small glucose measurements within a primary time window, and the step of obtaining the corresponding glucose titration level in accordance with another embodiment of the present disclosure. The illustrated method is particularly suitable, when glucose levels can be influenced by short acting insulin. FIGS. 6E and 6F collectively illustrate that the relation between tertiary windows and a present time 610, where a user requests an evaluation of the titration glucose level. The figures also illustrate the temporal alignment of tertiary windows, epochs and calendar units, e.g., a calendar day. FIGS. 7A, 7B, 7C, 7D, 7E and 7F collectively illustrate examples of identifying a titration subset of small glucose measurements according embodiment of the present disclosure. FIGS. 7A and 7B illustrate an embodiment wherein the titration subset is identified as the lowest glucose measurements are identified within the primary window. FIGS. 7C and 7D illustrate an embodiment wherein a running average, for three different time windows (length of secondary time window), of the glucose measurements is calculated within the primary time window. A titration subset can be identified as the subset corresponding to the lowest running average, for a given time window. The identified lowest running average corresponds to the titration glucose level. FIGS. 7E and 7F illustrate an embodiment wherein a running evaluation of the variance, for three different time windows (length of secondary time window), of the glucose measurements is calculated within the primary time window. A titration subset can be identified as the subset corresponding to the lowest value of the running evaluation of the variance, for a given time window. The titration glucose level is obtained as the average glucose value of the titration subset. FIGS. 7A, 7C and 7E illustrate examples where the blood glucose in the primary window is influenced by long acting insulin only. FIGS. 7B, 7D and 7F illustrate examples where the blood glucose in the primary time window is influenced by a short acting insulin. The methods illustrated in FIGS. 7A to 7D are most suitable when the blood glucose in influenced by long acting insulin only, whereas the methods illustrated in FIG. 7E-7F is suitable when the blood glucose is influenced by short acting insulin or in similar situations wherein the variance of the blood glucose has effected to increase. Like reference numerals refer to corresponding parts throughout the several views of the drawings. DETAILED DESCRIPTION The present disclosure provides robust systems and methods for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject. The present disclosure relies upon the acquisition of data regarding a data set comprising a plurality of glucose measurements of a subject taken over a time course and, for each respective glucose measurement in the plurality of glucose measurements, a corresponding timestamp representing when in the time course the respective glucose measurement was made. FIG. 1 illustrates an example of an integrated system 502 for the acquisition of such data, and FIG. 5 provides more details of such a system 502. The integrated system 502 includes one or more connected insulin pens 104, one or more glucose monitors or glucose sensors 102, memory 506, and a processor (not shown) for performing algorithmic categorization of autonomous glucose data of a subject. In some embodiments, a glucose monitor 102 is a continuous glucose monitor. With the integrated system 502, autonomous timestamped glucose measurements of the subject are obtained 520. Also, in some embodiments, data from the one or more insulin pens 104 used to apply a prescribed insulin regimen to the subject is obtained 540 as a plurality of records. Each record comprises a timestamped event specifying an amount of injected insulin medicament that the subject received as part of the prescribed insulin medicament dosage regimen. The glucose measurements are filtered 504 and stored in non-transitory memory 506. The plurality of glucose measurements of the subject taken over a time are used to determine a titration glucose level of the subject 508. In this way, the glucose data is analyzed to adjust the long acting insulin medicament dosage based upon a the titration glucose level in accordance with the methods of the present disclosure 510. Reference will now be made in detail to embodiments, examples of which are illustrated in the accompanying drawings. In the following detailed description, numerous specific details are set forth in order to provide a thorough understanding of the present disclosure. However, it will be apparent to one of ordinary skill in the art that the present disclosure may be practiced without these specific details. In other instances, well-known methods, procedures, components, circuits, and networks have not been described in detail so as not to unnecessarily obscure aspects of the embodiments. It will also be understood that, although the terms first, second, etc. may be used herein to describe various elements, these elements should not be limited by these terms. These terms are only used to distinguish one element from another. For example, a first subject could be termed a second subject, and, similarly, a second subject could be termed a first subject, without departing from the scope of the present disclosure. The first subject and the second subject are both subjects, but they are not the same subject. Furthermore, the terms “subject” and “user” are used interchangeably herein. By the term insulin pen is meant an injection device suitable for applying discrete doses of insulin, and wherein the injection device is adapted for logging and communicating dose related data. The terminology used in the present disclosure is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used in the description of the invention and the appended claims, the singular forms “a”, “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. It will also be understood that the term “and/or” as used herein refers to and encompasses any and all possible combinations of one or more of the associated listed items. It will be further understood that the terms “comprises” and/or “comprising,” when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. As used herein, the term “if” may be construed to mean “when” or “upon” or “in response to determining” or “in response to detecting,” depending on the context. Similarly, the phrase “if it is determined” or “if [a stated condition or event] is detected” may be construed to mean “upon determining” or “in response to determining” or “upon detecting [the stated condition or event]” or “in response to detecting [the stated condition or event],” depending on the context. A detailed description of a system 48 for adjusting a long acting insulin medicament dosage 216 in a prescribed insulin regimen for a subject in accordance with the present disclosure is described in conjunction with FIGS. 1 through 3. As such, FIGS. 1 through 3 collectively illustrate the topology of the system in accordance with the present disclosure. In the topology, there is a basal titration adjustment device for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject (“basal titration adjustment device 250”) (FIGS. 1, 2, and 3), a device for data collection (“data collection device 200”), one or more glucose sensors 102 associated with the subject (FIGS. 1 and 5), and one or more insulin pens 104 for injecting insulin medicaments into the subject (FIGS. 1 and 5). Throughout the present disclosure, the data collection device 200 and the basal titration adjustment device 250 will be referenced as separate devices solely for purposes of clarity. That is, the disclosed functionality of the data collection device 200 and the disclosed functionality of the basal titration adjustment device 250 are contained in separate devices as illustrated in FIG. 1. However, it will be appreciated that, in fact, in some embodiments, the disclosed functionality of the data collection device 200 and the disclosed functionality of the basal titration adjustment device 250 are contained in a single device. In some embodiments, the disclosed functionality of the data collection device 200 and/or the disclosed functionality of the basal titration adjustment device 250 are contained in a single device and this single device is a glucose monitor 102 or the insulin pen 104. Referring to FIG. 1, the basal titration adjustment device 250 autonomously adjusts a long acting insulin medicament dosage in a prescribed insulin regimen for a subject. To do this, the data collection device 200, which is in electrical communication with the basal titration adjustment device 250, receives autonomous glucose measurements originating from one or more glucose sensors 102 attached to a subject on an ongoing basis. In some embodiments, the data collection device 200 also receives insulin medicament injection data from one or more insulin pens 104 used by the subject to inject insulin medicaments. In some embodiments, the data collection device 200 receives such data directly from the glucose sensor(s) 102 and insulin pens 104 used by the subject. For instance, in some embodiments the data collection device 200 receives this data wirelessly through radio-frequency signals. In some embodiments such signals are in accordance with an 802.11 (WiFi), Bluetooth, or ZigBee standard. In some embodiments, the data collection device 200 receives such data directly, analyzes the data, and passes the analyzed data to the basal titration adjustment device 250. In some embodiments, a glucose sensor 102 and/or insulin pen 104 includes an RFID tag and communicates to the data collection device 200 and/or the basal titration adjustment device 250 using RFID communication. In some embodiments, the data collection device 200 also obtains or receives physiological measurements 247 of the subject (e.g., from wearable physiological measurement devices, from measurement devices within the data collection device 200 such as a magnetometer or thermostat, etc). In some embodiments, the data collection device 200 and/or the basal titration adjustment device 250 is not proximate to the subject and/or does not have wireless capabilities or such wireless capabilities are not used for the purpose of acquiring glucose data, insulin medicament injection data, and/or physiological measurement data. In such embodiments, a communication network 106 may be used to communicate glucose measurements from the glucose sensor 102 to the data collection device 200 and/or the basal titration adjustment device 250, insulin medicament injection data from the one or more insulin pens 104 to the data collection device 200 and/or the basal titration adjustment device 250, and/or physiological measurement data from one or more physiological measurement devices (not shown) to the data collection device 200 and/or the basal titration adjustment device 250. Examples of networks 106 include, are, but not limited to, the World Wide Web (WWW), an intranet and/or a wireless network, such as a cellular telephone network, a wireless local area network (LAN) and/or a metropolitan area network (MAN), and other devices by wireless communication. The wireless communication optionally uses any of a plurality of communications standards, protocols and technologies, including but not limited to Global System for Mobile Communications (GSM), Enhanced Data GSM Environment (EDGE), high-speed downlink packet access (HSDPA), high-speed uplink packet access (HSUPA), Evolution, Data-Only (EV-DO), HSPA, HSPA+, Dual-Cell HSPA (DC-HSPDA), long term evolution (LTE), near field communication (NFC), wideband code division multiple access (W-CDMA), code division multiple access (CDMA), time division multiple access (TDMA), Bluetooth, Wireless Fidelity (Wi-Fi) (e.g., IEEE 802.11a, IEEE 802.11ac, IEEE 802.11ax, IEEE 802.11b, IEEE 802.11g and/or IEEE 802.11n), voice over Internet Protocol (VoIP), Wi-MAX, a protocol for e-mail (e.g., Internet message access protocol (IMAP) and/or post office protocol (POP)), instant messaging (e.g., extensible messaging and presence protocol (XMPP), Session Initiation Protocol for Instant Messaging and Presence Leveraging Extensions (SIMPLE), Instant Messaging and Presence Service (IMPS)), and/or Short Message Service (SMS), or any other suitable communication protocol, including communication protocols not yet developed as of the filing date of the present disclosure. In some embodiments, there is a single glucose sensor 102 attached to the subject and the data collection device 200 and/or the basal titration adjustment device 250 is part of the glucose sensor 102. That is, in some embodiments, the data collection device 200 and/or the basal titration adjustment device 250 and the glucose sensor 102 are a single device. In some embodiments, the data collection device 200 and/or the basal titration adjustment device 250 is part of an insulin pen. That is, in some embodiments, the data collection device 200 and/or the basal titration adjustment device 250 and an insulin pen 104 are a single device. Of course, other topologies of the system 48 are possible. For instance, rather than relying on a communications network 106, the one or more glucose sensors 102 and the one or more insulin pens 104 may wirelessly transmit information directly to the data collection device 200 and/or basal titration adjustment device 250. Further, the data collection device 200 and/or the basal titration adjustment device 250 may constitute a portable electronic device, a server computer, or in fact constitute several computers that are linked together in a network or be a virtual machine in a cloud computing context. As such, the exemplary topology shown in FIG. 1 merely serves to describe the features of an embodiment of the present disclosure in a manner that will be readily understood to one of skill in the art. Referring to FIGS. 2A, 2B, 2C and 2D, in typical embodiments, the basal titration adjustment device 250 comprises one or more computers. For purposes of illustration in FIG. 2A, the basal titration adjustment device 250 is represented as a single computer that includes all of the functionality for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject. However, the disclosure is not so limited. In some embodiments, the functionality for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject is spread across any number of networked computers and/or resides on each of several networked computers and/or is hosted on one or more virtual machines at a remote location accessible across the communications network 106. One of skill in the art will appreciate that any of a wide array of different computer topologies are used for the application and all such topologies are within the scope of the present disclosure. Turning to FIG. 2 with the foregoing in mind, an exemplary basal titration adjustment device 250 for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject comprises one or more processing units (CPU's) 274, a network or other communications network interface 284, a memory 192 (e.g., random access memory), one or more magnetic disk storage and/or persistent devices 290 optionally accessed by one or more controllers 288, one or more communication busses 213 for interconnecting the aforementioned components, and a power supply 276 for powering the aforementioned components. In some embodiments, data in memory 192 is seamlessly shared with non-volatile memory 290 using known computing techniques such as caching. In some embodiments, memory 192 and/or memory 290 includes mass storage that is remotely located with respect to the central processing unit(s) 274. In other words, some data stored in memory 192 and/or memory 290 may in fact be hosted on computers that are external to the basal titration adjustment device 250 but that can be electronically accessed by the basal titration adjustment device 250 over an Internet, intranet, or other form of network or electronic cable (illustrated as element 106 in FIG. 2) using network interface 284. In some embodiments, the memory 192 of the basal titration adjustment device 250 for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject stores: an operating system 202 that includes procedures for handling various basic system services; a basal titration adjustment module 204; a first data structure 210, the first data structure comprising a prescribed insulin regimen 212 for the subject comprising a basal insulin medicament dosage regimen 214. Optionally, the basal insulin medicament dosage regimen 214 comprises a long acting insulin medicament dosage indicator 2116 and an associated epoch 218. The indicator 216 indicates, when the basal insulin medicament is to be injected within the associated epoch 218. Also optionally, the prescribed insulin regimen comprises an insulin state indicator 211, wherein the insulin state indicator 211 can indicate a short-acting-insulin-influence state, wherein the glucose measurements may be influenced by a short acting insulin medicament, and an only-long-acting-insulin-influence state, wherein the glucose measurements can be influenced by a long acting insulin medicament only, and the glucose measurements cannot be influenced by a short acting insulin medicament; a first data set 220, the first data set representing a time course and comprising a plurality of glucose measurements of the subject over the time course, and for each respective glucose measurement 230 in the plurality of glucose measurements, a timestamp 232 representing when the respective glucose measurement was made; a primary time window 233 defined within the time course, and thereby comprising a glucose measurements 236 from the first data set 220, the primary time window further comprises a submodule SM1 247, SM2 248 or SM3 275 defining further data structures used to identify a titration subset of small glucose measurements (240, 269, 273), and to obtain a glucose titration level 246 associated with the primary time window 233; submodule SM1 247 comprises an integer 234 defining the number of glucose measurements to be selected for a titration subset of small glucose measurements 240, the titration subset of small glucose measurements 240 being a subset of smallest glucose measurements within the primary time window 233. The submodule further comprises a first glucose measure of central tendency 244 being a measure of central tendency of the titration subset of small glucose measurements 240. The titration glucose level 246 is to be associated with the first glucose measure of central tendency; submodule SM2 248 defines a plurality of contemporaneously overlapping secondary time windows 260, wherein each secondary time window 262 comprises glucose measurements 266 defining a subset of overlapping glucose measurements 264 within the primary time window and a second glucose measure of central tendency being the measure of central tendency of the corresponding subset of overlapping glucose measurements, whereby the submodule defines a plurality of second glucose measures of central tendency. The submodule further defines a smallest second glucose measure of central tendency 268 being the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency. The titration glucose level 246 is to be associated with smallest second glucose measure of central tendency 268; submodule SM3 275 comprises a plurality of contemporaneously overlapping secondary time windows 260 within the primary time window 233, wherein each secondary time window 262 comprises a subset of overlapping glucose measurements 264 defining a subset of the glucose measurements 236 within the primary time window 233 and a glucose measure of variability 271 being a measure of variability of the corresponding subset of overlapping glucose measurements 264, and thereby obtaining a plurality of glucose measures of variability, whereby the submodule defines a plurality of glucose measure of variability. The submodule 275 further comprises a smallest glucose measure of variability 272 being the smallest glucose measure of variability within the plurality of glucose measures of variability, and a titration subset of small glucose measurements 273 being the subset comprising the glucose measurements within the secondary time window 262 corresponding to the smallest glucose measure of variability 272. The submodule further comprises a smallest third glucose measure of central tendency 274 being a measure of central tendency of the titration subset of small glucose measurements 273. The titration glucose level 246 is to be associated with the smallest third glucose measure of central tendency 274. In some embodiments, the basal titration adjustment module 204 is accessible within any browser (phone, tablet, laptop/desktop). In some embodiments the basal titration adjustment module 204 runs on native device frameworks, and is available for download onto the basal titration adjustment device 250 running an operating system 202 such as Android or iOS. In some implementations, one or more of the above identified data elements or modules of the basal titration adjustment device 250 for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject are stored in one or more of the previously described memory devices, and correspond to a set of instructions for performing a function described above. The above-identified data, modules or programs (e.g., sets of instructions) need not be implemented as separate software programs, procedures or modules, and thus various subsets of these modules may be combined or otherwise re-arranged in various implementations. In some implementations, the memory 192 and/or 290 optionally stores a subset of the modules and data structures identified above. Furthermore, in some embodiments the memory 192 and/or 290 stores additional modules and data structures not described above. In some embodiments, a basal titration adjustment device 250 for autonomously adjusting a long acting insulin medicament dosage 216 in a prescribed insulin regimen 212 for a subject is a smart phone (e.g., an iPHONE), laptop, tablet computer, desktop computer, or other form of electronic device (e.g., a gaming console). In some embodiments, the basal titration adjustment device 250 is not mobile. In some embodiments, the basal titration adjustment device 250 is mobile. FIGS. 3A, 3B, 3C and 3D provides a further description of a specific embodiment of a basal titration adjustment device 250 that can be used with the instant disclosure. The basal titration adjustment device 250 illustrated in FIG. 3A through 3D has one or more processing units (CPU's) 274, peripherals interface 370, memory controller 368, a network or other communications interface 284, a memory 192 (e.g., random access memory), a user interface 278, the user interface 278 including a display 282 and input 280 (e.g., keyboard, keypad, touch screen), an optional accelerometer 317, an optional GPS 319, optional audio circuitry 372, an optional speaker 360, an optional microphone 362, one or more optional intensity sensors 364 for detecting intensity of contacts on the basal titration adjustment device 250 (e.g., a touch-sensitive surface such as a touch-sensitive display system 282 of the basal titration adjustment device 250), an optional input/output (I/O) subsystem 366, one or more optional optical sensors 373, one or more communication busses 213 for interconnecting the aforementioned components, and a power supply 276 for powering the aforementioned components. In some embodiments, the input 280 is a touch-sensitive display, such as a touch-sensitive surface. In some embodiments, the user interface 278 includes one or more soft keyboard embodiments. The soft keyboard embodiments may include standard (QWERTY) and/or non-standard configurations of symbols on the displayed icons. The basal titration adjustment device 250 illustrated in FIG. 3 optionally includes, in addition to accelerometer(s) 317, a magnetometer (not shown) and a GPS 319 (or GLONASS or other global navigation system) receiver for obtaining information concerning the location and orientation (e.g., portrait or landscape) of the basal titration adjustment device 250 and/or for determining an amount of physical exertion by the subject. It should be appreciated that the basal titration adjustment device 250 illustrated in FIG. 3 is only one example of a multifunction device that may be used for autonomously adjusting a long acting insulin medicament dosage (216) in a prescribed insulin regimen for a subject, and that the basal titration adjustment device 250 optionally has more or fewer components than shown, optionally combines two or more components, or optionally has a different configuration or arrangement of the components. The various components shown in FIG. 3 are implemented in hardware, software, firmware, or a combination thereof, including one or more signal processing and/or application specific integrated circuits. Memory 192 of the basal titration adjustment device 250 illustrated in FIG. 3 optionally includes high-speed random access memory and optionally also includes non-volatile memory, such as one or more magnetic disk storage devices, flash memory devices, or other non-volatile solid-state memory devices. Access to memory 192 by other components of the basal titration adjustment device 250, such as CPU(s) 274 is, optionally, controlled by the memory controller 368. In some embodiments, the memory 192 of the basal titration adjustment device 250 illustrated in FIG. 3 optionally includes a third data set 246 comprising a plurality of physiological measurements, and each such physiological measurement 247 includes a measurement value 248. In some embodiments, the physiological measurement 247 is body temperature of the subject. In some embodiments, the physiological measurement 247 is a measurement of activity of the subject. In some embodiments, these physiological measurements serve as an additional glycaemic risk measure. In some embodiments, the optional accelerometer 317, optional GPS 319, and/or magnetometer (not shown) of the basal titration adjustment device 250 or such components optionally within the one or more glucose monitors 102 and/or the one or more pens 104 is used to acquire such physiological measurements 247. The peripherals interface 370 can be used to couple input and output peripherals of the device to CPU(s) 274 and memory 192. The one or more processors 274 run or execute various software programs and/or sets of instructions stored in memory 192, such as the insulin regimen monitoring module 204, to perform various functions for the basal titration adjustment device 250 and to process data. In some embodiments, the peripherals interface 370, CPU(s) 274, and memory controller 368 are, optionally, implemented on a single chip. In some other embodiments, they are, optionally, implemented on separate chips. RF (radio frequency) circuitry of network interface 284 receives and sends RF signals, also called electromagnetic signals. In some embodiments, the first data structure 210, the first data set 228, the optional second data set 320 is received using this RF circuitry from one or more devices such as a glucose sensor 102 associated with a subject, an insulin pen 104 associated with the subject and/or the data collection device 200. In some embodiments, the RF circuitry 108 converts electrical signals to/from electromagnetic signals and communicates with communications networks and other communications devices, glucose sensors 102, and insulin pens 104 and/or the data collection device 200 via the electromagnetic signals. The RF circuitry 284 optionally includes well-known circuitry for performing these functions, including but not limited to an antenna system, an RF transceiver, one or more amplifiers, a tuner, one or more oscillators, a digital signal processor, a CODEC chipset, a subscriber identity module (SIM) card, memory, and so forth. RF circuitry 284 optionally communicates with the communication network 106. In some embodiments, the circuitry 284 does not include RF circuitry and, in fact, is connected to the network 106 through one or more hard wires (e.g., an optical cable, a coaxial cable, or the like). In some embodiments, the audio circuitry 372, the optional speaker 360, and the optional microphone 362 provide an audio interface between the subject and the basal titration adjustment device 250. The audio circuitry 372 receives audio data from peripherals interface 370, converts the audio data to electrical signals, and transmits the electrical signals to speaker 360. The speaker 360 converts the electrical signals to human-audible sound waves. The audio circuitry 372 also receives electrical signals converted by the microphone 362 from sound waves. The audio circuitry 372 converts the electrical signal to audio data and transmits the audio data to peripherals interface 370 for processing. Audio data is, optionally, retrieved from and/or transmitted to the memory 192 and/or the RF circuitry 284 by the peripherals interface 370. In some embodiments, the power supply 276 optionally includes a power management system, one or more power sources (e.g., battery, alternating current (AC)), a recharging system, a power failure detection circuit, a power converter or inverter, a power status indicator (e.g., a light-emitting diode (LED)) and any other components associated with the generation, management and distribution of power in portable devices. In some embodiments, the basal titration adjustment device 250 optionally also includes one or more optical sensors 373. The optical sensor(s) 373 optionally include charge-coupled device (CCD) or complementary metal-oxide semiconductor (CMOS) phototransistors. The optical sensor(s) 373 receive light from the environment, projected through one or more lens, and converts the light to data representing an image. The optical sensor(s) 373 optionally capture still images and/or video. In some embodiments, an optical sensor is located on the back of the basal titration adjustment device 250, opposite the display 282 on the front of the basal titration adjustment device 250, so that the input 280 is enabled for use as a viewfinder for still and/or video image acquisition. In some embodiments, another optical sensor 373 is located on the front of the basal titration adjustment device 250 so that the subject's image is obtained (e.g., to verify the health or condition of the subject, to determine the physical activity level of the subject, or to help diagnose a subject's condition remotely, to acquire visual physiological measurements 247 of the subject, etc.). As illustrated in FIG. 3, a basal titration adjustment device 250 preferably comprises an operating system 202 that includes procedures for handling various basic system services. The operating system 202 (e.g., iOS, DARWIN, RTXC, LINUX, UNIX, OS X, WINDOWS, or an embedded operating system such as VxWorks) includes various software components and/or drivers for controlling and managing general system tasks (e.g., memory management, storage device control, power management, etc.) and facilitates communication between various hardware and software components. In some embodiments the basal titration adjustment device 250 is a smart phone. In other embodiments, the basal titration adjustment device 250 is not a smart phone but rather is a tablet computer, desktop computer, emergency vehicle computer, or other form or wired or wireless networked device. In some embodiments, the basal titration adjustment device 250 has any or all of the circuitry, hardware components, and software components found in the basal titration adjustment device 250 depicted in FIG. 2 or 3. In the interest of brevity and clarity, only a few of the possible components of the basal titration adjustment device 250 are shown in order to better emphasize the additional software modules that are installed on the basal titration adjustment device 250. In some embodiments, and as illustrated in FIG. 3A through 3D, the memory 192 of the basal titration adjustment device 250 for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject further stores: a second data set 320 obtained from one or more insulin pens 104 used by the subject to apply the prescribed insulin regimen, the second data set comprising a plurality of insulin medicament records over the time course, each insulin medicament record 321 in the plurality of medicament records comprising: (i) a respective insulin medicament injection event 322 representing an insulin medicament injection into the subject using a respective insulin pen in the one or more insulin pens 104 and (ii) a corresponding electronic timestamp 323 that is automatically generated by the respective insulin pen upon occurrence of the respective insulin medicament injection event; and for each first glucose measures of central tendency 244 is associated a tertiary time window 330 representing an evaluation period, for the plurality of second glucose measures of central tendency, each respective second glucose measure of central tendency 267 is associated with a time indicator 331 representing the time of evaluation of the respective second glucose measure of central tendency, and thereby obtaining a plurality of time indicators defining a tertiary time window 330 representing an evaluation period, or for the plurality of glucose measures of variability in the third evaluation mode, each respective glucose measure of variability 267 is associated with a time indicator 331 representing the time of evaluation of the respective glucose measure of variability, and thereby obtaining a plurality of time indicators defining a tertiary time window 330 representing an evaluation period; and the titration glucose level 246 is associated with the tertiary time window 330; a first characterization 335 applied to the tertiary time window 330, wherein the first characterization 335 is one of basal regimen adherent and basal regimen nonadherent, the tertiary time window 330 is deemed basal regimen adherent when the second data set 320 includes one or more medicament records that establish, on a temporal and quantitative basis, adherence with the prescribed basal insulin medicament dosage regimen 212 during the respective tertiary time window 330, and the tertiary time window is deemed basal regimen nonadherent when the second data set 320 fails to include one or more medicament records that establish, on a temporal and quantitative basis, adherence with the prescribed basal insulin medicament dosage regimen 212 during the tertiary time window 330. While the system 48 disclosed in FIG. 1 can work standalone, in some embodiments it can also be linked with electronic medical records to exchange information in any way. Now that details of a system 48 for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject have been disclosed, details regarding a flow chart of processes and features of the system, in accordance with an embodiment of the present disclosure, are disclosed with reference to FIGS. 4A through 4E. In some embodiments, such processes and features of the system are carried out by the basal titration adjustment module 204 illustrated in FIGS. 2 and 3. Block 402. Block 402 illustrates the beginning of the process. Block 410. With reference to block 410 of FIG. 4A, the goal of insulin therapy in subjects with either type 1 diabetes mellitus or type 2 diabetes mellitus is to match as closely as possible normal physiologic insulin secretion to control fasting and postprandial plasma glucose. As illustrated in FIG. 2, a basal titration adjustment device 250 comprises one or more processors 274 and a memory 192/290. The memory stores instructions that, when executed by the one or more processors, perform a method. In the method, a first data set 228 is obtained. The first data set 228 comprises glucose measurements 230 of the subject from one or more glucose sensors 102. FIG. 2 illustrates. Each such glucose measurement 230 is timestamped with a glucose measurement timestamp 232 to represent when the respective measurement was made. In some embodiments, the glucose measurements 230 are autonomously measured. The FREESTYLE LIBRE CGM by ABBOTT (“LIBRE”) is an example of a glucose sensor that may be used as a glucose sensor 102 in order to make autonomous glucose measurements of a subject. The LIBRE allows calibration-free glucose measurements with an on-skin coin-sized sensor, which can send up to eight hours of data to a reader device (e.g., the data collection device 200 and/or the basal titration adjustment device 250) via near field communications, when brought close together. The LIBRE can be worn for fourteen days in all daily life activities. Referring to block 410, in some embodiments, the glucose measurements are taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less. In some embodiments, the glucose measurements are taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less, over a time period of a day or more, two days or more, a week or more, or two weeks or more. In some embodiments, the glucose measurements are autonomously taken (e.g., without human effort, without human intervention, etc.). In some embodiments, the glucose measurements are manually taken (e.g., with manual human effort, with human intervention, etc.). The basal titration adjustment device 250 accesses and/or stores a first data structure 210 that includes the prescribed insulin regimen 212 including a basal insulin medicament dosage regimen 214, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage 216, an insulin state indicator 211, wherein the insulin state indicator 211 can indicate a short-acting-insulin-influence state, wherein the glucose measurements within the primary time window may be influenced by a short acting insulin medicament, and an only-long-acting-insulin-influence state, wherein the glucose measurements within the primary time window can be influenced by a long acting insulin medicament, but the glucose measurements cannot be influenced by a short acting insulin medicament. In some embodiments, the long acting insulin medicament specified by the basal insulin medicament dosage regimen 214 consists of a single insulin medicament having a duration of action that is between 12 and 24 hours or a mixture of insulin medicaments that collectively have a duration of action that is between 12 and 24 hours. Examples of such long acting insulin medicaments include, but are not limited to Insulin Degludec (developed by NOVO NORDISK under the brand name Tresiba), NPH (Schmid, 2007, “New options in insulin therapy. J Pediatria (Rio J). 83(Suppl 5): S146-S155), Glargine (LANTUS, Mar. 2, 2007, insulin glargine [rDNA origin] injection, [prescribing information], Bridgewater, N.J.: Sanofi-Aventis), and Determir (Plank et al., 2005, “A double-blind, randomized, dose-response study investigating the pharmacodynamic and pharmacokinetic properties of the long-acting insulin analog detemir,” Diabetes Care 28:1107-1112). In some embodiments, the prescribed regimen may also comprise a bolus insulin medicament dosage regimen specifying the amount of short acting insulin medicament dosage. The short acting insulin medicament specified by the bolus insulin medicament dosage regimen comprises a single insulin medicament having a duration of action that is between three to eight hours or a mixture of insulin medicaments that collectively have a duration of action that is between three to eight hours. Examples of such short acting insulin medicaments include, but are not limited, to Lispro (HUMALOG, May 18, 2001, insulin lispro [rDNA origin] injection, [prescribing information], Indianapolis, Ind.: Eli Lilly and Company), Aspart (NOVOLOG, July 2011, insulin aspart [rDNA origin] injection, [prescribing information], Princeton, N.J., NOVO NORDISK Inc., July, 2011), Glulisine (Helms Kelley, 2009, “Insulin glulisine: an evaluation of its pharmacodynamic properties and clinical application,” Ann Pharmacother 43:658-668), and Regular (Gerich, 2002, “Novel insulins: expanding options in diabetes management,” Am J Med. 113:308-316). Block 420. Referring to block 410 of FIG. 4A, the method continues with the process step (b) of obtaining a titration glucose level 246 based on small glucose measurements and the state of the insulin state indicator 211. Block 422. Referring to block 422 of FIG. 4A, the method continues with (i) obtaining a primary time window 233 within the time course defining the period of time comprising the glucose measurements 236 in the first data set to be used for identifying the titration glucose level. Block 424. Referring to block 424 of FIG. 4A, the method continues with (ii) identifying a titration subset of small glucose measurements 240, 269, 273, identified as a subset of small glucose measurements within the primary time window 233. Block 426. Referring to block 426 of FIG. 4A, the method continues with (iii) obtaining the titration glucose level 246 computed as a measure of central tendency 244, 268, 274 of the titration subset of small glucose measurements 240, 269, 273, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window. Block 428. Referring to block 428 of FIG. 4A, the method continues with (iv) associating the titration glucose level 246 with the measure of central tendency 244, 268, 274, and thereby completing the process step (b) of obtaining the titration glucose level. Block 440. Referring to block 440 of FIG. 4A, the method continues with the process step (C) of adjusting or maintaining the long acting insulin medicament dosage 216 based upon the obtained titration glucose level 246. Block 404. Block 404 illustrates the end of the process. As appears from FIG. 4A the method can be divided into the overall steps: step A wherein the glucose measurements are obtained, step B wherein the titration glucose level is obtained, and step C wherein the long acting insulin is adjusted based on the glucose titration level. Referring to FIG. 4B, in some embodiments according to the disclosure the obtaining the titration glucose level 246, in step B, further comprises, based on the status of the insulin state indicator, selecting one of the following evaluation modes: a first evaluation mode illustrated by subprocess B1, a second evaluation mode illustrated by subprocess B2 and a third evaluation mode illustrated by subprocess B3. Block 450. Referring to block 450 of FIG. 4C, the method can continue in a first evaluation mode with the subprocess B1, wherein the method further comprises, for the primary time window (i) obtaining an integer 234 defining the number of glucose measurements to be selected for the titration subset of small glucose measurements 240. Block 452. Referring to block 452 of FIG. 4C, the method continues with (ii) identifying and selecting the titration subset of small glucose measurements 240 as a subset of smallest glucose measurements, by identifying and selecting the smallest glucose measurements within the primary time window 233, and ensuring that the number of measurements within the titration subset of small glucose measurements 240 equals the obtained integer 234. Block 454. Referring to block 454 of FIG. 4C, the method continues with (iii) obtaining a first glucose measure of central tendency 244, and computed as a measure of central tendency of the glucose measurements within the titration subset of small glucose measurements 240. Block 456. Referring to block 456 of FIG. 4C, the method continues with associating the titration glucose level 246 with the first glucose measure of central tendency 244, whereby the titration glucose level 246 can be fed into the method step C as illustrated on FIG. 4B. The process of obtaining the titration glucose level according to the first evaluation mode is also schematically illustrated in FIG. 6B. An integer 234 is obtained, the titration subset of small glucose measurements 240 is obtained, the first glucose measure of central tendency can be evaluated, and the titration glucose level 246 is obtained. Block 460. Referring to block 460 of FIG. 4C, the method can continue in a second evaluation mode with the subprocess B2, wherein the method further comprises, for the primary time window 233, obtaining a plurality of contemporaneously overlapping secondary time windows 260 within the primary time window 233, wherein each secondary time window 262 comprises a subset of overlapping glucose measurements 264 being a subset of the glucose measurements 236 in the primary time window 233 Block 462. Referring to block 462 of FIG. 4C, the method continues with, for each secondary time window 262 within the plurality of secondary time windows 260, computing a corresponding second glucose measure of central tendency 267, and thereby obtaining a plurality of second glucose measures of central tendency, wherein each respective second glucose measure of central tendency 267 is computed as a measure of central tendency of the glucose measurements within the corresponding secondary time window 262. Hereby is obtaining a moving period of a measure of central tendency across the glucose measurements in the primary time window. Block 464. Referring to block 464 of FIG. 4C, the method continues with, for the plurality of second glucose measures of central tendency, identifying a smallest second glucose measure of central tendency 268 as the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency, whereby the titration subset of small glucose measurements 269 is identified as the subset comprising the glucose measurements within the secondary time window 262 corresponding to the smallest second glucose measure of central tendency 268. Block 466. Referring to block 466 of FIG. 4C, the method continues with associating the titration glucose level 246 with the smallest second glucose measure of central tendency 268, whereby the titration glucose level 246 can be fed into the method step C as illustrated on FIG. 4B. The process of obtaining the titration glucose level according to the second evaluation mode is also schematically illustrated in FIG. 6C. A plurality of secondary time windows is obtained and a second glucose measure of central tendency 267 is associated with each of the secondary time windows, and thereby creating a plurality of second glucose measures of central tendency. Next, the smallest second glucose measure of central tendency, in the illustrated example the second glucose measure of central tendency 267-2, is identified as a smallest second glucose measure of central tendency 268 among the plurality of second glucose measures of central tendency, and the corresponding secondary time window 262-2 is identified as the titration subset of small glucose measurements 269, the smallest second glucose measure of central tendency 268 can be associated with the titration glucose level 246, whereby the titration glucose level is obtained, and can be used for adjusting the dose of the long acting insulin medicament. Block 470. Referring to block 470 of FIG. 4D, the method can continue in a third evaluation mode with the subprocess B3, wherein the method further comprises, for the primary time window 233, obtaining a plurality of contemporaneously overlapping secondary time windows 260 within the primary time window 233, wherein each secondary time window 262 comprises a subset of overlapping glucose measurements 264 being a subset of the glucose measurements 236 in the primary time window 233. Block 472. Referring to block 472 of FIG. 4D, the method continues with for each secondary time window 262 within the plurality of secondary time windows 260, computing a corresponding glucose measure of variability 271, and thereby obtaining a plurality of glucose measures of variability, wherein each respective glucose measure of variability 271 is computed as a measure of variability of the glucose measurements within the corresponding secondary time window 262, and thereby obtaining a moving period of a measure of variability across the glucose measurements in the primary time window. Block 474. Referring to block 474 of FIG. 4D, the method continues with, for the plurality of glucose measures of variability, identifying a smallest glucose measure of variability 272, as the smallest glucose measure of variability within the plurality of glucose measures of variability, whereby the titration subset of small glucose measurements 273 is identified as the subset comprising the glucose measurements within the secondary time window 262 corresponding to the smallest glucose measure of variability 272. Block 476. Referring to block 476 of FIG. 4D, the method continues with computing a smallest third glucose measure of central tendency 274 as a measure of central tendency of the titration subset of small glucose measurements 273. Block 478. Referring to block 478 of FIG. 4D, the method continues with associating the titration glucose level 246 with the smallest third glucose measure of central tendency 274. The process of obtaining the titration glucose level according to the third evaluation mode is also schematically illustrated in FIG. 6D. A plurality of secondary time windows is obtained and a glucose measure of variability 271 is associated with each of the secondary time windows, and thereby creating a plurality of glucose measures of variability. Next, the smallest glucose measure of variability, in the illustrated example it is the secondary time window 262-2, is identified as a smallest glucose measure of variability 272 among the plurality of glucose measures of variability, and the corresponding secondary time window 262-2 is identified as the titration subset of small glucose measurements 273, a third glucose measure of central tendency 274 is evaluated based on the titration subset of small glucose measurements 273 and associated with the titration glucose level 246, whereby the titration glucose level is obtained, and can be used for adjusting the dose of the long acting insulin medicament. Block 430. Referring to block 430 of FIG. 4C, in some embodiments according to the present disclosure the method further comprises, in response to identifying the state of the insulin state indicator 211, selecting the first or the second evaluation mode, upon the occurrence that the state of the insulin state indicator is identified as the only-long-acting-insulin-influence state, and thereby using a preferred method for obtaining the titration glucose level 246, which is preferred for titration with a long acting insulin medicament, when it is ensured that no short acting insulin medicament influences the glucose measurements 236. Block 430. Referring to block 430 of FIG. 4C, in some embodiments according to the present disclosure the method further comprises, in response to identifying the state of the insulin state indicator 310, selecting the third evaluation mode, upon the occurrence that the state of the insulin state indicator is identified as the a short-acting-insulin-influence state, and thereby using a preferred method for obtaining the titration glucose level 246, which is preferred for titration with a long acting insulin medicament, when it is identified that short acting insulin medicament influences the glucose measurements 236. Block 432. Referring to block 432 of FIG. 4C, in some embodiments according to the present disclosure the first data structure further comprises a hypoglycemic risk state indicator, wherein the hypoglycemic risk state indicator can indicate a high hypoglycemic risk state, wherein the subject may have a high hypoglycemic risk or wherein a high variability across the plurality of glucose measurements can be observed, and a non-high hypoglycemic risk state, wherein the subject may have a non-high hypoglycemic risk or wherein a low variability across the plurality of glucose measurements can be observed, and wherein the method further comprises: in response to identifying the state of the hypoglycemic risk state indicator, selecting the first evaluation mode, upon the occurrence that the state of the hypoglycemic risk state indicator is identified as the high hypoglycemic risk state, and thereby using a method for obtaining the titration glucose level 246 which may be beneficial in situation with low glucose values and noise. Block 432. Referring to block 432 of FIG. 4C, in some embodiments according to the present disclosure the first data structure further comprises a hypoglycemic risk state indicator, wherein the hypoglycemic risk state indicator can indicate a high hypoglycemic risk state, wherein the subject may have a high hypoglycemic risk or wherein a high variability across the plurality of glucose measurements can be observed, and a non-high hypoglycemic risk state, wherein the subject may have a non-high hypoglycemic risk or wherein a low variability across the plurality of glucose measurements can be observed, and wherein the method further comprises: in response to identifying the state of the hypoglycemic risk state indicator, selecting the second evaluation mode, upon the occurrence that the state of the hypoglycemic risk state indicator is identified as the non-high hypoglycemic risk state, and thereby using a method for obtaining the titration glucose level 246 which may be beneficial under these circumstances. In some embodiments of the present disclosure, the measure of variability 271 is the variance. For instance, in some embodiments a moving measure of variability is a moving period of variance σk2 across the glucose measurements, where: σ k 2 = ( 1 M ⁢ ∑ i = k - M + 1 k ⁢ ( G i - G _ ) ) 2 and where, Gi is the ith glucose measurement in the portion k of the plurality of glucose measurements considered, M is a number of glucose measurements in the plurality of glucose measurements and represents a contiguous predetermined time span, i.e., the secondary time window, G is the mean of the M glucose measurements selected from the plurality of glucose measurements of the first data set 228, and k is within the first time window. In some embodiments of the present disclosure the measure of central tendency 244, 268, 274 is the mean value. For instance, in some embodiments a moving measure of central tendency is a moving mean or moving average μk across the glucose measurements, where: μ k = 1 M ⁢ ∑ i = k - M + 1 k ⁢ G i and where, Gi is the ith glucose measurement in the portion k of the plurality of glucose measurements considered, M is a number of glucose measurements in the plurality of glucose measurements and represents a contiguous predetermined time span, i.e., the secondary time window, and k is within the first time period. In some embodiments of the present disclosure the method is repeated on a recurring basis. For example, whether and how much to adjust the dose of a long-acting insulin medicament can be requested on a daily basis, which means that the titration glucose level is evaluated on a daily basis. In some embodiments of the present disclosure, the method further comprises obtaining a second data set 320 from one or more insulin pens 104 used by the subject to apply the prescribed insulin regimen, as illustrated in FIGS. 3A through 3D, the second data set comprises a plurality of insulin medicament records over the time course, each insulin medicament record 321 in the plurality of medicament records comprises: (i) a respective insulin medicament injection event 322 representing an insulin medicament injection into the subject using a respective insulin pen in the one or more insulin pens 104 and (ii) a corresponding electronic timestamp 323 that is automatically generated by the respective insulin pen upon occurrence of the respective insulin medicament injection event. For the first glucose measures of central tendency 244 in the first evaluation mode, the first glucose measure of central tendency is associated with a tertiary time window 330 representing an evaluation period, wherein a most recent end point 614 of the tertiary time window is synchronized with a most recent end point 612 of the primary time window 233, and wherein the primary and the tertiary windows are of the same length, as illustrated on FIG. 6A in the marked area 604. Alternatively, for the plurality of second glucose measures of central tendency in the second evaluation mode, each respective second glucose measure of central tendency 267 is associated with a time indicator 331 representing the time of evaluation of the respective second glucose measure of central tendency, and thereby obtaining a plurality of time indicators defining a tertiary time window 330 representing an evaluation period, wherein a most recent end point 614 of the tertiary time window is synchronized with a most recent end point 612 of the primary time window, and wherein the length 615 of the tertiary time window 330 is smaller than the length 613 of the primary time window 233, as illustrated on FIG. 6A in the marked area 602. Alternatively, for the plurality of glucose measures of variability in the third evaluation mode, each respective glucose measure of variability 267 is associated with a time indicator 331 representing the time of evaluation of the respective glucose measure of variability, and thereby obtaining a plurality of time indicators defining a tertiary time window 330 representing an evaluation period, wherein a most recent end point 614 of the tertiary time window is synchronized with a most recent end point (612) of the primary time window, and wherein the length 615 of the tertiary time window 330 is smaller than the length 613 of the primary time window 233. This also illustrated on FIG. 6A in the marked area 604. The method continues by associating the titration glucose level 246 with the tertiary time window 330, as illustrated on FIGS. 6E and 6F, and applying a first characterization 335 to the tertiary time window 330 (not illustrated), wherein the first characterization 335 is one of basal regimen adherent and basal regimen nonadherent, the tertiary time window 330 is deemed basal regimen adherent when the second data set 320 includes one or more medicament records that establish, on a temporal and quantitative basis, adherence with the prescribed basal insulin medicament dosage regimen 212 during the respective tertiary time window 330, and the tertiary time window is deemed basal regimen nonadherent when the second data set 320 fails to include one or more medicament records that establish, on a temporal and quantitative basis, adherence with the prescribed basal insulin medicament dosage regimen 212 during the tertiary time window 330. Hereafter the method may continue with adjusting the long acting insulin medicament dosage 216 in the basal insulin medicament dosage regimen 214 for the subject based upon a titration glucose level 244 represented by a tertiary time window 330 that is deemed basal regimen adherent and by excluding a titration glucose level 244 represented by a tertiary time window 330 that is deemed basal regimen nonadherent. As illustrated on FIG. 2A and FIG. 6A, in some embodiments the first data structure 210 comprises a plurality of consecutive epochs, wherein each respective epoch 218 is associated with a basal insulin medicament dosage 216, indicating when the basal insulin medicament is to be injected within the respective epoch 218, and how much of the basal insulin medicament is to be injected. Thereby the first data structure 210 provides a temporal and quantitative basis for the first characterization. In some embodiments the length 615 of the tertiary window 330 is longer than or the same as the length 619 of each of the epochs 218. In some embodiments the end point 614 of the tertiary time window is synchronized with an end point 618 of a current epoch, wherein the current epoch is the most recent completed epoch within the plurality of epochs. As illustrated on FIGS. 6E and 6F, in some embodiments each respective epoch 218 of the plurality of epochs is associated with a tertiary time window 330, and thereby obtaining a plurality of tertiary time windows, wherein each tertiary time window represents an evaluation period, wherein each tertiary window is aligned with the respective epoch on a temporal bases, and wherein each tertiary time window 330 is associated with a titration glucose level 246. In some embodiments the first data structure comprises a specification of temporal and quantitative basis for administration of the long acting insulin medicament, for each of the epochs 218 within the plurality of epochs. In some embodiments the quantitative basis for the long acting insulin medicament is a function of the titration glucose level. In some embodiments the temporal basis is specified as one injection for each epoch 218 within the plurality of epochs. In some embodiments each epoch 218 in the plurality of epochs is a calendar day or a calendar week, as illustrated in FIGS. 6E and 6F. In some embodiments successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less. In some embodiments the device further comprises a wireless receiver 284, and wherein the first data set is obtained wirelessly from a glucose sensor 102 affixed to the subject and/or the second data set is obtained wirelessly from the one or more insulin pens 104. In some embodiments, the first data structure further comprises a hypoglycemic risk state indicator 311, wherein the hypoglycemic risk state indicator 311 can indicate a high hypoglycemic risk state, wherein the subject may have a high hypoglycemic risk or wherein a high variability across the plurality of glucose measurements can be observed, and a non-high hypoglycemic risk state, wherein the subject may have a non-high hypoglycemic risk or wherein a low variability across the plurality of glucose measurements can be observed, and wherein the method further comprises. Referring to Block 432 FIG. 4B, in response to identifying the state of the hypoglycemic risk state indicator, the method further comprises selecting the first evaluation mode, upon the occurrence that the state of the hypoglycemic risk state indicator is identified as the high hypoglycemic risk state, and thereby using a method for obtaining the titration glucose level 246 which is more beneficial in case of low glucose values and noise. In some embodiments, the secondary time window is 50 minutes to 70 minutes, 60 minutes to 120 minutes, 120 minutes to 180 minutes or 180 minutes to 300 minutes. In some embodiments, successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 4 minutes to 6 minutes, and wherein the secondary time window is 50 minutes to 70 minutes. In some embodiments successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 40 minutes to 80 minutes, and wherein the secondary time window is 180 minutes to 310 minutes. A flow diagram of the subprocesses B1, B2 and B2 applying a first, second and third evaluation mode, respectively, has been illustrated on FIG. 4B through 4D, and FIG. 7A through FIG. 7F further illustrates by example how the titration subset of small glucose measurements is obtained in order to obtain the titration glucose level. FIGS. 7A, 7C and 7E illustrate a first case, where a patient is only on basal insulin and therefore the low glucose values can only be caused by basal insulin injections or endogenous insulin or other natural causes. In the second case, illustrated in FIGS. 7B, 7D and 7F, the patient is on multiple daily injections (MDI) treatment which involves injections with fast acting insulin, and small glucose values are therefore not necessarily related to the basal insulin injections or endogenous insulin production, but could also be caused by the fast acting insulin used to account for the carbohydrates ingested during a meal or to correct for a high glucose level after a meal (correction bolus). The examples of FIG. 7A through 7F illustrate glucose data obtained during a time course of 24 hours, and calendar hours is illustrated by rectangles 710. The primary time window 233, the tertiary time window 330, the epoch 218 and the request time 610, where a user requests the calculation of a titration glucose level, is also indicated on the figures. The patterned rectangle 702 indicates the upper and lower boundaries defining a desired range for the blood glucose level. Referring to FIGS. 7A and 7B, the top panel 704 illustrates the glucose level as a function of time obtained from the plurality of glucose measurements within the time course. The second panel from the top 706, the third panel from the top 708 and the fourth panel from the top 712, respectively, shows the lowest glucose values corresponding to 5 hours, 3 hours, and 1 hours sampling, wherein the sampling rate has been 1 sample or measurement per 5 minutes, i.e., 12 measurements per hour. Therefore 5 hours sampling corresponds to 60 measurements, and the second panel from the top shows the 60 smallest or lowest measurements, 3 hours sampling corresponds to 36 measurements and the third panel from the top shows the 36 smallest measurements, and 1 hour corresponds to 12 measurements and the fourth panel from the top (the bottom panel) shows the 12 smallest measurements. Each of the sets containing a number of smallest glucose measurements can be used as the titration subset of small glucose measurements 269, and can be used to evaluate the titration glucose level 246. The integer 234 used to define the number of measurements in the titration subset of small glucose measurements is here obtained as a product of the sampling rate and the time window, e.g., 12 measurements per hour for 1 hour is 12 measurements. The integer could also be specified as a nearest integer of a fraction of the total number of measurements within the primary time window. For the glucose data illustrated in FIG. 7A the patient is on basal insulin only, hence low glucose values only relate to endogenous insulin and long acting insulin. Here the lowest average is found at around 9:00 on January 20. For the glucose data illustrated in FIG. 7B the patient takes a correction bolus after dinner which causes glucose to decrease at around 20:00. Here the lowest average is found at around 20:00 on January 19. Referring to FIGS. 7C and 7D, the top panel 714 illustrates the glucose level as a function of time obtained from the plurality of glucose measurements within the time course. The second panel 716 shows the running average glucose with different window sizes of 5 hours, 3 hours and 1 hour, respectively. The lowest evaluated average can be used as the titration glucose level. The illustrated tertiary window 330 corresponds to the running average using a 5 hours window (secondary time window is 5 hours). For the glucose data illustrated in FIG. 7C the patient is on basal insulin only, hence low glucose values only relate to endogenous insulin and long acting insulin. Here the smallest average for the 1 hour window 730 is found at around 9:00 on January 20. The smallest average for the 3 hour window 731 is found at around 9:30. The smallest average for the 5 hour window 732 is found around 10:00. For the glucose data illustrated in FIG. 7D the patient takes a correction bolus after dinner which causes glucose to decrease at around 20:30. Here the smallest average for the 1 hour window 733 is found at around 20:30 on January 19. The smallest average for the 3 hour window 734 is found at around 17:00 (beginning of tertiary period for 3 hour window). The smallest average for the 5 hour window 735 is found at around 19:00 (beginning of tertiary period 330 for the 5 hour window). Referring to FIGS. 7E and 7F, the top panel 718 illustrates the glucose level as a function of time obtained from the plurality of glucose measurements within the time course. The second panel 720 shows the running variance of the glucose measurements with different window sizes of 5 hours, 3 hours and 1 hour, respectively. The smallest evaluated variance can be used to identify the titration subset of small glucose values, which can be used to evaluate the titration glucose level. The illustrated tertiary window 330 corresponds to the running average using a 5 hours window (secondary time window is 5 hours). For the glucose data illustrated in FIG. 7E the patient is on basal insulin only, hence low glucose values only relate to endogenous insulin and long acting insulin. In this case we are interested in the lowest glucose value rather than low variance since we know that basal insulin or endogenous insulin is causing the lowest values. The lowest running variance for the 1 hour window 736 is around 3:30. For the 3 hours window, the smallest running variance for the 3 hour window 737 is found around 4:45. For the 5 hours window, the lowest running variance for the 5 hour window 738 is found around 5:15. For the glucose data illustrated in FIG. 7F the patient is on basal and bolus insulin. The patient takes a correction bolus after dinner which causes glucose to decrease at around 20:00. In this case we are not interested in the lowest glucose value since it is caused by a bolus and not the basal insulin. The lowest running variance for the 1 hour window 739 is around 13:00. For the 3 hours window, the smallest running variance for the 3 hour window 740 is found around 03:20. The 3 hour window also has a dip at around 01:00 which is almost as low as the dip around 03:20. For the 5 hours window, the lowest running variance for the 5 hour window 741 is found around 03:40. TABLE 1 Time detection of minimum values for running average of glucose measurements and running variance for glucose measurements. Minimum for Minimum for Minimum for 1 hour 3 hour 5 hour window window window FIG. 7C 09:00 09:30 10:00 (running average of glucose) FIG. 7D 20:30 17:00 19:00 (running average of glucose) FIG. 7E 03:30 04:45 05:15 (running variance of glucose) FIG. 7F 13:00 03:20 03:40 (running variance of glucose) Comparing FIG. 7C where running average is used with FIG. 7E, where running variance is used, the smallest variance was found at around 4:00 depending on the time window, whereas the smallest average is found between 9 and 10 also depending on the time window. In this case we are interested in the lowest value rather than low variance since we know that basal insulin or endogenous insulin is causing the lowest values. Comparing FIG. 7D where running average is used with FIG. 7F, where running variance is used, the smallest variance was found at around 3:30 in the night (with frame size of 3 or 5 hours, the timing actually depends on the frame size), whereas the smallest running average is found around 17:00 and 19-20:30, also depending on the window or frame size. In this case we are not interested in the lowest glucose value since it is caused by a bolus and not the basal insulin. Prior dose guidance algorithms for long acting insulin base their calculations on fasting glucose levels, where the glucose measurement for example has been tagged as a fasting glucose measurement. In the embodiment disclosed on FIGS. 7E and 7F we use a third evaluation mode to determine periods of fasting periods based on periods of low variance in glucose. The argumentation for identifying a titration glucose level in this way, is that glucose variance is typically higher during periods of external glucose entering the system than during fasting. This approach is created with MDI treatment in mind, where the algorithm can utilize information or identify that glucose levels are influenced e.g. by external short acting insulin. Therefore, a different approach is presented according to the embodiment disclosed in FIGS. 7A and 7B where we use a second evaluation mode, where a titration glucose level for dose guidance algorithms for long acting insulin is determined using low glucose values. The definition titration glucose level is used to emphasize that this is not necessarily a fasting glucose value, rather the glucose titration level to base dose calculations on is low glucose values. Instead of using periods of low variance in glucose, we use the information that the glucose is not influenced by short acting insulin, and we use periods of low average glucose within a specific time window. For example, we use a running average window of 3 hours and choose the window with the lowest value as the titration glucose level. This is useful in long acting insulin treatment since we are certain that there is no fast acting insulin causing the lowest values; all lowered glucose values are due to the long acting insulin or physiological changes. We detect titration glucose level by selecting the lowest average of glucose levels during a predefined time interval (i.e. if the time predefined interval is 60 minutes and frequency of measurements is 5 minutes, choose the lowest of 288 12 consecutive measurements). This approach calculates a moving average (MA) of N consecutive measurements, MA period , i = 1 N ⁢ ∑ k = i - ( N - 1 ) i ⁢ G k where G is a glucose measurement, the subscript i is the sample at a specific point in time, N is the number of samples in each average, and the titration glucose level of a period is determined by FG=min(MAperiod) As opposed to methods that assume fasting occurs at predefined times, this approach mitigates people with irregular daily routines (shift workers etc.) and people whose daily routines deviate from normal practice (e.g. not having breakfast), see FIG. 7A. The frequency of continuous glucose measurements can vary, and can span for example from a near continuous signal and up to 1 hour. Example 1: First Evaluation Mode—Subset of Lowest Measurements This method is similar to the running average in that it targets low glucose levels. Therefore it is relevant in basal insulin treatment. This method is more sensitive to low values and noise than the moving average, which might be beneficial in scenarios of e.g. patients with high hyporisk and high glucose variability. As previously mentioned examples of an embodiment can be seen in FIGS. 7A and 7B. Example 2: Second Evaluation Mode—Running or Moving Average In basal insulin titration, typically fasting glucose levels from the past three days are used to determine a next dose size. In this embodiment we use DexCom G5 as an example. The CGM outputs glucose levels every 5 minutes, which represent the average glucose levels of the past 5 minutes. We detect TGL by selecting the lowest average glucose level over the past 0-24, 24-48 and 48-72 hours to obtain TGL of the past three days. For each 24 hour long time interval, a window of 60 minutes, or 12 consecutive CGM measurements, runs through the glucose data and average of each window is calculated. This approach calculates a moving average (MA) of 12 consecutive measurements of 5 minute intervals using the following equation, MA 0 ⁢ - ⁢ 24 ⁢ h , i = 1 12 ⁢ ∑ k = i - 11 i ⁢ G k where G is a glucose measurement and the subscript i refers to the current sample, and the fasting glucose of time interval 0-24 hours is determined by FG0-24h=min(MA0-24h) As previously mentioned, two examples of results are shown in FIGS. 7C and 7D for time frames of 1, 3 and 5 hours. In the first case, a patient is only on basal insulin and therefore the low glucose values can only be caused by basal insulin injections or endogenous insulin (or other natural causes). In the second case the patient is on MDI treatment and therefore low glucose values are not necessarily related to the basal injection or endogenous production, but could also be caused by a meal or correction bolus. Example 3: Third Evaluation Mode—Moving Variance Dose guidance algorithms for MDI treatment including long and fast acting insulin base their calculations on fasting glucose as well as pre-prandial glucose levels. For the purpose of determining periods of fasting to be used as a titration glucose level we detect periods of low variance in glucose. We do this because glucose variance is typically higher during periods of external glucose entering the system than during fasting. This method is similar to the moving average except that instead of calculating average in a time frame we calculate variance within the time frame. As previously mentioned example of embodiments can be seen in FIGS. 7E and 7F. LISTS OF EMBODIMENTS In a first list of embodiments is provided a basal titration adjustment device, adapted for adjusting a long acting insulin regimen, wherein the device is adapted for obtaining a titration glucose level based on small glucose measurements and the state of an insulin state indicator. 1. A basal titration adjustment device 250 for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, wherein the device comprises one or more processors 274 and a memory 192/290, the memory comprising: a first data structure 210 that includes the prescribed insulin regimen 212 including a basal insulin medicament dosage regimen 214, wherein the basal insulin medicament dosage regimen specifies the long acting insulin medicament dosage 216, an insulin state indicator 211, wherein the insulin state indicator 211 can indicate a short-acting-insulin-influence state, wherein the glucose measurements within the primary time window may be influenced by a short acting insulin medicament, and an only-long-acting-insulin-influence state, wherein the glucose measurements within the primary time window can be influenced by a long acting insulin medicament, but the measurements cannot be influenced by a short acting insulin medicament, and instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set 228, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement 230 in the plurality of glucose measurements, a corresponding timestamp 232 representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level 246 based on small glucose measurements and the state of the insulin state indicator 211 by: (i) obtaining a primary time window 233 within the time course defining the period of time comprising the glucose measurements 236 in the first data set to be used for identifying the titration glucose level, (ii) identifying a titration subset of small glucose measurements 240, 269, 273, identified as a subset of small glucose measurements within the primary time window 233, (iii) obtaining the titration glucose level 246 computed as a measure of central tendency 244, 268, 274 of the titration subset of small glucose measurements 240, 269, 273, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level 246 with the measure of central tendency 244, 268, 274; (C) adjusting or maintaining the long acting insulin medicament dosage 216 based upon the obtained titration glucose level 246. 2. The device of embodiment 1, wherein the obtaining the titration glucose level 246, in step B, further comprises: based on the status of the insulin state indicator, selecting one of the following evaluation modes: (B1) for the primary time window in a first evaluation mode, (i) obtaining an integer 234 defining the number of glucose measurements to be selected for the titration subset of small glucose measurements 240, (ii) identifying and selecting the titration subset of small glucose measurements 240 as a subset of smallest glucose measurements, by identifying and selecting the smallest glucose measurements within the primary time window 233, and ensuring that the number of measurements within the titration subset of small glucose measurements 240 equals the obtained integer 234, (iii) obtaining the measure of central tendency as a first glucose measure of central tendency 244, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements 240, and associating the titration glucose level 246 with the first glucose measure of central tendency 244, (B2) for the primary time window 233 in a second evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows 260 within the primary time window 233, wherein each secondary time window 262 comprises a subset of overlapping glucose measurements 264 being a subset of the glucose measurements 236 in the primary time window 233, for each secondary time window 262 within the plurality of secondary time windows 260, computing a corresponding second glucose measure of central tendency 267, and thereby obtaining a plurality of second glucose measures of central tendency, wherein each respective second glucose measure of central tendency 267 is computed as a measure of central tendency of the glucose measurements within the corresponding secondary time window 262, and thereby obtaining a moving period of a measure of central tendency across the glucose measurements in the primary time window, for the plurality of second glucose measures of central tendency, identifying a smallest second glucose measure of central tendency 268 as the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency, whereby the titration subset of small glucose measurements 269 is identified as the subset comprising the glucose measurements within the secondary time window 262 corresponding to the smallest second glucose measure of central tendency 268, and associating the titration glucose level 246 with the smallest second glucose measure of central tendency 268, or (B3) for the primary time window 233 in a third evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows 260 within the primary time window 233, wherein each secondary time window 262 comprises a subset of overlapping glucose measurements 264 being a subset of the glucose measurements 236 in the primary time window 233, for each secondary time window 262 within the plurality of secondary time windows 260, computing a corresponding glucose measure of variability 271, and thereby obtaining a plurality of glucose measures of variability, wherein each respective glucose measure of variability 271 is computed as a measure of variability of the glucose measurements within the corresponding secondary time window 262, and thereby obtaining a moving period of a measure of variability across the glucose measurements in the primary time window, for the plurality of glucose measures of variability, identifying a smallest glucose measure of variability 272 as the smallest glucose measure of variability within the plurality of glucose measures of variability, whereby the titration subset of small glucose measurements 273 is identified as the subset comprising the glucose measurements within the secondary time window 262 corresponding to the smallest glucose measure of variability 272, and computing a smallest third glucose measure of central tendency 274 as a measure of central tendency of the titration subset of small glucose measurements 273, and associating the titration glucose level 246 with the smallest third glucose measure of central tendency 274. 3. The device of any of the embodiments 1 or 2, wherein the method further comprises: in response to identifying the state of the insulin state indicator 211, selecting the first or the second evaluation mode, upon the occurrence that the state of the insulin state indicator is identified as the only-long-acting-insulin-influence state, and thereby using a preferred method for obtaining the titration glucose level 246, which is preferred for titration with a long acting insulin medicament, when it is ensured that no short acting insulin medicament influences the glucose measurements 236. 4. The device of any of embodiments 1 or 2, wherein the method further comprises: in response to identifying the state of the insulin state indicator 310, selecting the third evaluation mode, upon the occurrence that the state of the insulin state indicator is identified as the a short-acting-insulin-influence state, and thereby using a preferred method for obtaining the titration glucose level 246, which is preferred for titration with a long acting insulin medicament, when it is identified that short acting insulin medicament may influence the glucose measurements 236. 5. The device of any one of the embodiments 2-4, wherein the measure of variability 271 is the variance. 6. The device of any one of the previous embodiments, wherein the measure of central tendency 244, 268, 274 is the mean value. 7. The device of any one of the previous embodiments, wherein the method is repeated on a recurring basis. 8. The device of any one of the embodiments 2-7, wherein the method further comprises: obtaining a second data set 320 from one or more insulin pens 104 used by the subject to apply the prescribed insulin regimen, the second data set comprising a plurality of insulin medicament records over the time course, each insulin medicament record 321 in the plurality of medicament records comprising: (i) a respective insulin medicament injection event 322 representing an insulin medicament injection into the subject using a respective insulin pen in the one or more insulin pens 104 and (ii) a corresponding electronic timestamp 323 that is automatically generated by the respective insulin pen upon occurrence of the respective insulin medicament injection event; for the first glucose measures of central tendency 244 in the first evaluation mode, associating the first glucose measure of central tendency with a tertiary time window 330 representing an evaluation period, wherein a most recent end point 614 of the tertiary time window is synchronized with a most recent end point 612 of the primary time window 233, and wherein the primary and the tertiary windows are of the same length, for the plurality of second glucose measures of central tendency in the second evaluation mode, associating each respective second glucose measure of central tendency 267 with a time indicator 331 representing the time of evaluation of the respective second glucose measure of central tendency, and thereby obtaining a plurality of time indicators defining a tertiary time window 330 representing an evaluation period, wherein a most recent end point 614 of the tertiary time window is synchronized with a most recent end point 612 of the primary time window, and wherein the length 615 of the tertiary time window 330 is smaller than the length 613 of the primary time window 233, or for the plurality of glucose measures of variability in the third evaluation mode, associating each respective glucose measure of variability 267 with a time indicator 331 representing the time of evaluation of the respective glucose measure of variability, and thereby obtaining a plurality of time indicators defining a tertiary time window 330 representing an evaluation period, wherein a most recent end point 614 of the tertiary time window is synchronized with a most recent end point 612 of the primary time window, and wherein the length 615 of the tertiary time window 330 is smaller than the length 613 of the primary time window 233; and associating the titration glucose level 246 with the tertiary time window 330; applying a first characterization 335 to the tertiary time window 330, wherein the first characterization 335 is one of basal regimen adherent and basal regimen nonadherent, the tertiary time window 330 is deemed basal regimen adherent when the second data set 320 includes one or more medicament records that establish, on a temporal and quantitative basis, adherence with the prescribed basal insulin medicament dosage regimen 212 during the respective tertiary time window 330, and the tertiary time window is deemed basal regimen nonadherent when the second data set 320 fails to include one or more medicament records that establish, on a temporal and quantitative basis, adherence with the prescribed basal insulin medicament dosage regimen 212 during the tertiary time window 330; and wherein the adjusting the long acting insulin medicament dosage 216 in the basal insulin medicament dosage regimen 214 for the subject is based upon a titration glucose level 244 that is represented by a tertiary time window 330 that is deemed basal regimen adherent and by excluding a titration glucose level 244 that is represented by a tertiary time window 330 that is deemed basal regimen nonadherent. 9. The device according to any of the previous embodiments, wherein the first data structure comprises, a plurality of consecutive epochs, wherein each respective epoch 218 is associated with a basal insulin medicament dosage 216, indicating when the basal insulin medicament is to be injected within the respective epoch 218, and how much of the basal insulin medicament is to be injected, and thereby providing a temporal and quantitative basis for the first characterization. 10. The device according to embodiments 8 and 9, wherein the length 615 of the tertiary window 330 is longer than or the same as the length 619 of each of the epochs 218. 11. The device of any of the embodiments 8-10, wherein the end point 614 of the tertiary time window is synchronized with an end point 618 of a current epoch, wherein the current epoch is the most recent completed epoch within the plurality of epochs. 12. The device of embodiment 9, wherein each respective epoch 218 of the plurality of epochs is associated with a tertiary time window 330, and thereby obtaining a plurality of tertiary time windows, wherein each tertiary time window represents an evaluation period, wherein each tertiary window is aligned with the respective epoch on a temporal bases, and wherein each tertiary time window 330 is associated with a titration glucose level 246. 13. The device of any of the previous embodiments, wherein the first data structure comprises a specification of temporal and quantitative basis for administration of the long acting insulin medicament, for each of the epochs 218 within the plurality of epochs. 14. The device of any of embodiments 8-13, wherein the quantitative basis for the long acting insulin medicament is a function of the titration glucose level. 15. The device of any of embodiments 9-14, wherein the temporal basis is specified as one injection for each epoch 218 within the plurality of epochs. 16. The device of any of the embodiments 9-15, wherein each epoch 218 in the plurality of epochs is a calendar day or a calendar week. 17. The device of any of the previous embodiments, wherein successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less. 18. The device of any of the previous embodiments, wherein the device further comprises a wireless receiver 284, and wherein the first data set is obtained wirelessly from a glucose sensor 102 affixed to the subject and/or the second data set is obtained wirelessly from the one or more insulin pens 104. 19. The device of any of the previous embodiments, wherein the first data structure further comprises a hypoglycemic risk state indicator, wherein the hypoglycemic risk state indicator can indicate a high hypoglycemic risk state, wherein the subject may have a high hypoglycemic risk or wherein a high variability across the plurality of glucose measurements can be observed, and a non-high hypoglycemic risk state, wherein the subject may have a non-high hypoglycemic risk or wherein a low variability across the plurality of glucose measurements can be observed, and wherein the method further comprises: in response to identifying the state of the hypoglycemic risk state indicator, selecting the first evaluation mode, upon the occurrence that the state of the hypoglycemic risk state indicator is identified as the high hypoglycemic risk state, and thereby using a method for obtaining the titration glucose level 246 which is more sensitive to low glucose values and noise. 20. The device of any of the embodiments 2-19, wherein the secondary time window is 50 minutes to 70 minutes, 60 minutes to 120 minutes, 120 minutes to 180 minutes, 180 minutes to 300 minutes, or 300-500 minutes. 21. The device of any of the embodiments 2-20, wherein successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 4 minutes to 6 minutes. 22. The device of any of embodiments 2-19, wherein successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 4 minutes to 6 minutes, and wherein the secondary time window is 50 minutes to 70 minutes. 23. The device of any of embodiments 2-19, wherein successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 4 minutes to 6 minutes, and wherein the secondary time window is 180 minutes to 300 minutes. 24. The device of any of embodiments 2-19, wherein successive measurements in the plurality of glucose measurements are autonomously taken from the subject at an interval rate of 40 minutes to 80 minutes, and wherein the secondary time window is 180 minutes to 310 minutes. 25. The device of any one of the previous embodiments, wherein the method further comprises: obtaining a second data set (320) from one or more insulin pens (104) used by the subject to apply the prescribed insulin regimen, the second data set comprising a plurality of insulin medicament records over the time course, each insulin medicament record (321) in the plurality of medicament records comprising: (i) a respective insulin medicament injection event (322) representing an insulin medicament injection into the subject using a respective insulin pen in the one or more insulin pens (104) and (ii) a corresponding electronic timestamp (323) that is automatically generated by the respective insulin pen upon occurrence of the respective insulin medicament injection event, (iii) the type of insulin medicament indicating whether the injected insulin medicament is a short acting insulin medicament type or a long acting insulin medicament type. 26. The device of any of the previous embodiments, wherein the prescribed insulin regimen further comprises a bolus insulin medicament dosage regimen, wherein the bolus insulin medicament dosage regimen specifies the [amount of] short acting insulin dosage. 27. The device according to any of the embodiments 25-26, wherein the prescribed insulin regimen 212 further comprises a duration of action for the short acting insulin medicament, wherein the duration of action of a medicament specifies the duration that a measurable medicament effect persist, and thereby influences the glucose level. 28. The device according to any of embodiments 25-27, wherein the prescribed insulin regimen 212 comprises a duration of action for the long acting insulin medicament, wherein the duration of action of a medicament specifies the duration that a measurable medicament effect persist, and thereby influences the glucose level. 29. The device of any of the embodiments 25-27, wherein the insulin state indicator 211 indicates the short-acting-insulin-influence state based on the second data set and the duration of action for the short acting insulin medicament. 30. The device of any of the embodiments 25-29, wherein the insulin state indicator 211 indicates the only-long-acting-insulin-influence state based on the second data set and the duration of action for the short acting insulin medicament. 31. The device of any of the embodiments 25-30, wherein the insulin state indicator 211 indicates the only-long-acting-insulin-influence state further based on the duration of action for the long acting insulin medicament. 32. The device of any of the previous embodiments, wherein the titration glucose level 246 is associated with the first glucose measure of central tendency 244 by assigning the value of the first measure of central tendency to the titration glucose level. 33. The device of any of the previous embodiments, wherein the measurements in the plurality of glucose measurements are autonomously obtained by a continuous glucose monitor. 34. The device according to any of the previous embodiments, wherein the titration glucose level 246 is the glucose level used as input to an algorithm for maintaining or adjusting the long acting insulin medicament dosage 216. 35. The device according to any of the previous embodiments, wherein the glucose measurements 236 in the first data set to be used for identifying the titration glucose level, comprises the titration subset of small glucose measurements 240, 269, 273. 36. The device according to any of the previous embodiments, wherein the glucose measurements comprised in the titration subset of small glucose measurements 240, 269, 273 have values smaller than a lower percentile of the glucose measurements 236, wherein the lower percentile ranges from the 0.1th percentile to the 50th percentile, wherein a Pth percentile is defined as the lowest glucose measurement that is greater than P % of the glucose measurements 236 in the first data set to be used for identifying the titration subset 240, 269, 273 within the primary time window 233. (iii) obtaining the titration glucose level 246 computed as a measure of central tendency 244, 268, 274 of the titration subset of small glucose measurements 240, 269, 273, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level 246 with the measure of central tendency 244, 268, 274; 37. The device of any of the previous embodiments, wherein the obtaining the titration glucose level 246, in step B, further comprises: verifying that the values of the glucose measurements comprised in the titration subset of small glucose measurements 240, 269, 273 are smaller than the lower percentile of the glucose measurements 236. 38. The device of any of the embodiments 36-37, wherein the lower percentile is the 5th percentile, wherein a 5th percentile is defined as the lowest glucose measurement that is greater than 5% of the glucose measurements 236 in the first data set to be used for identifying the titration subset 240, 269, 273. 39. A method for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, at a computer comprising one or more processors and a memory: the memory storing: a first data structure 210 that includes the prescribed insulin regimen 212 including a basal insulin medicament dosage regimen 214, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage 216, an insulin state indicator 211, wherein the insulin state indicator 211 can indicate a short-acting-insulin-influence state, wherein the glucose measurements within the primary time window may be influenced by a short acting insulin medicament, and an only-long-acting-insulin-influence state, wherein the glucose measurements within the primary time window can be influenced by a long acting insulin medicament, but the measurements cannot be influenced by a short acting insulin medicament, and the memory further storing instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set 228, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement 230 in the plurality of glucose measurements, a corresponding timestamp 232 representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level 246 based on small glucose measurements and the state of the insulin state indicator 211 by: (i) obtaining a primary time window 233 within the time course defining the period of time comprising the glucose measurements 236 in the first data set to be used for identifying the titration glucose level, (ii) identifying a titration subset of small glucose measurements 240, 269, 273, identified as a subset of small glucose measurements within the primary time window (233), (iii) obtaining the titration glucose level 246 computed as a measure of central tendency 244, 268, 274 of the titration subset of small glucose measurements 240, 269, 273, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level 246 with the measure of central tendency 244, 268, 274; (C) adjusting or maintaining the long acting insulin medicament dosage 216 based upon the obtained titration glucose level 246. 40. A computer program comprising instructions that, when executed by a computer having one or more processors and a memory, perform the method of embodiment 39. 41. A computer-readable data carrier having stored thereon the computer program according to embodiment 39. In a second list of embodiments is provided a basal titration adjustment device, adapted for adjusting a long acting insulin regimen, wherein the device is adapted for obtaining a titration glucose level based on small glucose measurements. 1. A basal titration adjustment device 250 for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, wherein the device comprises one or more processors 274 and a memory 192/290, the memory comprising: a first data structure 210 that includes the prescribed insulin regimen 212 including a basal insulin medicament dosage regimen 214, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage 216, and instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set 228, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement 230 in the plurality of glucose measurements, a corresponding timestamp 232 representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level 246 by: (i) obtaining a primary time window 233 within the time course defining the period of time comprising the glucose measurements 236 in the first data set to be used for identifying the titration glucose level, (ii) identifying a titration subset of small glucose measurements 240, 269, 273, identified as a subset of small glucose measurements within the primary time window 233, (iii) obtaining the titration glucose level 246 computed as a measure of central tendency 244, 268, 274 of the titration subset of small glucose measurements 240, 269, 273, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level 246 with the measure of central tendency 244, 268, 274; (C) adjusting or maintaining the long acting insulin medicament dosage 216 based upon the obtained titration glucose level 246. 2. The device of embodiment 1, wherein the obtaining the titration glucose level (246), in step B, further comprises: selecting one of the following evaluation modes: (B1) for the primary time window in a first evaluation mode, (i) obtaining an integer 234 defining the number of glucose measurements to be selected for the subset of glucose measurements 240, (ii) identifying and selecting the titration subset of small glucose measurements 240 as a subset of smallest glucose measurements, by identifying and selecting the smallest glucose measurements within the primary time window 233, and ensuring that the number of measurements within the titration subset of small glucose measurements 240 equals the obtained integer 234, (iii) obtaining the measure of central tendency as a first glucose measure of central tendency 244, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements 240, and associating the titration glucose level 246 with the first glucose measure of central tendency 244, (B2) for the primary time window 233 in a second evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows 260 within the primary time window (233), wherein each secondary time window 262 comprises a subset of overlapping glucose measurements 264 being a subset of the glucose measurements 236 in the primary time window 233, for each secondary time window 262 within the plurality of secondary time windows 260, computing a corresponding second glucose measure of central tendency 267, and thereby obtaining a plurality of second glucose measures of central tendency, wherein each respective second glucose measure of central tendency 267 is computed as a measure of central tendency of the glucose measurements within the corresponding secondary time window 262, and thereby obtaining a moving period of a measure of central tendency across the glucose measurements in the primary time window, for the plurality of second glucose measures of central tendency, identifying a smallest second glucose measure of central tendency 268 as the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency, whereby the titration subset of small glucose measurements 269 is identified as the subset comprising the glucose measurements within the secondary time window 262 corresponding to the smallest second glucose measure of central tendency 268, and associating the titration glucose level 246 with the smallest second glucose measure of central tendency 268, or (B3) for the primary time window 233 in a third evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows 260 within the primary time window 233, wherein each secondary time window 262 comprises a subset of overlapping glucose measurements 264 being a subset of the glucose measurements (236) in the primary time window 233, for each secondary time window 262 within the plurality of secondary time windows 260, computing a corresponding glucose measure of variability 271, and thereby obtaining a plurality of glucose measures of variability, wherein each respective glucose measure of variability 272 is computed as a measure of variability of the glucose measurements within the corresponding secondary time window 262, and thereby obtaining a moving period of a measure of variability across the glucose measurements in the primary time window, for the plurality of glucose measures of variability, identifying a smallest glucose measure of variability 272 as the smallest glucose measure of variability within the plurality of glucose measures of variability, whereby the titration subset of small glucose measurements 273 is identified as the subset comprising the glucose measurements within the secondary time window 262 corresponding to the smallest glucose measure of variability 272, and computing a smallest third glucose measure of central tendency 274 as a measure of central tendency of the titration subset of small glucose measurements 273, and associating the titration glucose level 246 with the smallest third glucose measure of central tendency 274. In a third list of embodiments is provided a basal titration adjustment device, adapted for adjusting a long acting insulin regimen, wherein the device is adapted for obtaining a titration glucose level based on small glucose measurements. 1. A basal titration adjustment device 250 for autonomously adjusting a long acting insulin medicament dosage in a prescribed basal insulin regimen for a subject, wherein the device comprises one or more processors 274 and a memory 192/290, the memory comprising: a prescribed insulin regimen 212 including a basal insulin medicament dosage regimen 214, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage 216, and instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set 228, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement 230 in the plurality of glucose measurements, a corresponding timestamp 232 representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level 246 being the glucose level used as input to an algorithm for maintaining or adjusting the long acting insulin medicament dosage, wherein the titration glucose level is based on a titration subset of small glucose measurements, by: (i) obtaining a primary time window 233 within the time course defining the period of time comprising the glucose measurements 236 in the first data set to be used for identifying the titration subset of small glucose measurements 240, 269, 273 and for obtaining the titration glucose level for the primary time window 233, wherein each of the glucose measurements 236 has a timestamp 232 within the primary time window 233, (ii) identifying the titration subset of small glucose measurements 240, 269, 273, identified as a subset of small glucose measurements within the primary time window 233, (iii) obtaining the titration glucose level 246, computed as a measure of central tendency 244, 268, 274 of the titration subset of small glucose measurements 240, 269, 273, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level 246 with the measure of central tendency 244, 268, 274, by assigning the value of measure of central tendency to the titration glucose level; (C) adjusting or maintaining the long acting insulin medicament dosage 216 based upon the obtained titration glucose level 246. 2. The device of embodiment 1, wherein the obtaining the titration glucose level (246), in step B, further comprises: selecting one of the following evaluation modes: (B1) for the primary time window in a first evaluation mode, (i) obtaining an integer 234 defining the number of glucose measurements to be selected for the subset of glucose measurements 240, (ii) identifying and selecting the titration subset of small glucose measurements 240 as a subset of smallest glucose measurements, by identifying and selecting the smallest glucose measurements within the primary time window 233, and ensuring that the number of measurements within the titration subset of small glucose measurements 240 equals the obtained integer 234, (iii) obtaining the measure of central tendency as a first glucose measure of central tendency 244, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements 240, and associating the titration glucose level 246 with the first glucose measure of central tendency 244. 3. The device of embodiment 1, wherein the obtaining the titration glucose level (246), in step B, further comprises: (B2) for the primary time window 233 in a second evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows 260 within the primary time window (233), wherein each secondary time window 262 comprises a subset of overlapping glucose measurements 264 being a subset of the glucose measurements 236 in the primary time window 233, for each secondary time window 262 within the plurality of secondary time windows 260, computing a corresponding second glucose measure of central tendency 267, and thereby obtaining a plurality of second glucose measures of central tendency, wherein each respective second glucose measure of central tendency 267 is computed as a measure of central tendency of the glucose measurements within the corresponding secondary time window 262, and thereby obtaining a moving period of a measure of central tendency across the glucose measurements in the primary time window, for the plurality of second glucose measures of central tendency, identifying a smallest second glucose measure of central tendency 268 as the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency, whereby the titration subset of small glucose measurements 269 is identified as the subset comprising the glucose measurements within the secondary time window 262 corresponding to the smallest second glucose measure of central tendency 268, and associating the titration glucose level 246 with the smallest second glucose measure of central tendency 268. 4. The device of embodiment 1, wherein the obtaining the titration glucose level (246), in step B, further comprises: obtaining a percentage defining a titration percentile defining the number of glucose measurements to be selected for the titration subset of small glucose measurements, (ii) identifying and selecting the titration subset of small glucose measurements as a subset of smallest glucose measurements, by identifying and selecting the glucose measurements defined by the titration percentile of the glucose measurements (236), wherein the titration percentile ranges from the 0.1th percentile to the 50th percentile, and is smaller than or equal to the lower percentile, and wherein a Pth percentile is defined as the lowest glucose measurement that is greater than P % of the glucose measurements (236) in the first data set to be used for identifying the titration subset within the primary time window (233), (iii) obtaining the measure of central tendency as a fourth glucose measure of central tendency, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements (240), and associating the titration glucose level (246) with the fourth glucose measure of central tendency (244), by assigning the value of the fourth measure of central tendency to the titration glucose level. 5. The device of any of the previous embodiments, wherein the measurements in the plurality of glucose measurements are autonomously obtained by a continuous glucose monitor from the subject In a fourth list of embodiments is provided a basal titration adjustment device, adapted for adjusting a long acting insulin regimen, wherein the device is adapted for obtaining a titration glucose level based on small glucose measurements. 1. A basal titration adjustment device 250 for autonomously adjusting a long acting insulin medicament dosage in a prescribed insulin regimen for a subject, wherein the device comprises one or more processors 274 and a memory 192/290, the memory comprising: a prescribed insulin regimen 212 including a basal insulin medicament dosage regimen 214, wherein the basal insulin medicament dosage regimen specifies the amount of long acting insulin medicament dosage 216, and instructions that, when executed by the one or more processors, perform a method of: (A) obtaining, a first data set 228, the first data set comprising a plurality of glucose measurements of the subject taken over a time course and, for each respective glucose measurement 230 in the plurality of glucose measurements, a corresponding timestamp 232 representing when in the time course the respective glucose measurement was made; (B) obtaining a titration glucose level 246 being the glucose level used as input to an algorithm for maintaining or adjusting the long acting insulin medicament dosage, wherein the titration glucose level is based on a titration subset of small glucose measurements, by: (i) obtaining a primary time window 233 within the time course defining the period of time comprising the glucose measurements 236 in the first data set to be used for identifying the titration subset of small glucose measurements 240, 269, 273 and for obtaining the titration glucose level for the primary time window 233, wherein each of the glucose measurements 236 has a timestamp 232 within the primary time window 233, (ii) identifying the titration subset of small glucose measurements 240, 269, 273, identified as a subset of small glucose measurements within the primary time window 233, (iii) obtaining the titration glucose level 246, computed as a measure of central tendency 244, 268, 274 of the titration subset of small glucose measurements 240, 269, 273, wherein the measure of central tendency represents a measure of small glucose measurements for the primary time window, and (iv) associating the titration glucose level 246 with the measure of central tendency 244, 268, 274, by assigning the value of measure of central tendency to the titration glucose level; (C) adjusting or maintaining the long acting insulin medicament dosage 216 based upon the obtained titration glucose level 246. 2. The device of embodiment 1, wherein the obtaining the titration glucose level (246), in step B, further comprises: selecting one of the following evaluation modes: (B1) for the primary time window in a first evaluation mode, (i) obtaining an integer 234 defining the number of glucose measurements to be selected for the subset of glucose measurements 240, (ii) identifying and selecting the titration subset of small glucose measurements 240 as a subset of smallest glucose measurements, by identifying and selecting the smallest glucose measurements within the primary time window 233, and ensuring that the number of measurements within the titration subset of small glucose measurements 240 equals the obtained integer 234, (iii) obtaining the measure of central tendency as a first glucose measure of central tendency 244, and computed as a measure of central tendency of the glucose measurements within the subset of small glucose measurements 240, and associating the titration glucose level 246 with the first glucose measure of central tendency 244. 3. The device of embodiment 1, wherein the obtaining the titration glucose level (246), in step B, further comprises: (B2) for the primary time window 233 in a second evaluation mode, obtaining a plurality of contemporaneously overlapping secondary time windows 260 within the primary time window (233), wherein each secondary time window 262 comprises a subset of overlapping glucose measurements 264 being a subset of the glucose measurements 236 in the primary time window 233, for each secondary time window 262 within the plurality of secondary time windows 260, computing a corresponding second glucose measure of central tendency 267, and thereby obtaining a plurality of second glucose measures of central tendency, wherein each respective second glucose measure of central tendency 267 is computed as a measure of central tendency of the glucose measurements within the corresponding secondary time window 262, and thereby obtaining a moving period of a measure of central tendency across the glucose measurements in the primary time window, for the plurality of second glucose measures of central tendency, identifying a smallest second glucose measure of central tendency 268 as the smallest second glucose measure of central tendency within the plurality of second glucose measures of central tendency, whereby the titration subset of small glucose measurements 269 is identified as the subset comprising the glucose measurements within the secondary time window 262 corresponding to the smallest second glucose measure of central tendency 268, and associating the titration glucose level 246 with the smallest second glucose measure of central tendency 268. 4. The device according to any of the embodiments 1-3, wherein the prescribed insulin regimen 212 further comprises a duration of action for the short acting insulin medicament, wherein the duration of action of a medicament specifies the duration that a measurable medicament effect persist, and thereby influences the glucose level. 5. The device according to any of embodiments 1-4, wherein the prescribed insulin regimen 212 comprises a duration of action for the long acting insulin medicament, wherein the duration of action of a medicament specifies the duration that a measurable medicament effect persist, and thereby influences the glucose level. 6. The device of any one of the embodiments 1-5, wherein the method further comprises: obtaining a second data set 320 from one or more insulin pens 104 used by the subject to apply the prescribed insulin regimen, the second data set comprising a plurality of insulin medicament records over the time course, each insulin medicament record 321 in the plurality of medicament records comprising: (i) a respective insulin medicament injection event 322 representing an insulin medicament injection into the subject using a respective insulin pen in the one or more insulin pens 104 and (ii) a corresponding electronic timestamp 323 that is automatically generated by the respective insulin pen upon occurrence of the respective insulin medicament injection event, (iii) the type of insulin medicament indicating whether the injected insulin medicament is a short acting insulin medicament type or a long acting insulin medicament type, wherein the method further comprises (i) using the second data set and the duration of action to obtain the glucose measurements within the primary time window 233 that are not influenced by the short acting insulin, wherein the titration subset of small glucose measurements are identified within the glucose measurements within the primary time window 233 that are not influenced by the short acting insulin. 7. The device of any of the previous embodiments, wherein the measurements in the plurality of glucose measurements are autonomously obtained by a continuous glucose monitor from the subject REFERENCES CITED AND ALTERNATIVE EMBODIMENTS All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. The present invention can be implemented as a computer program product that comprises a computer program mechanism embedded in a nontransitory computer readable storage medium. For instance, the computer program product could contain the program modules shown in any combination of FIG. 1, 2, or 3 and/or described in FIG. 4. These program modules can be stored on a CD-ROM, DVD, magnetic disk storage product, or any other non-transitory computer readable data or program storage product. Many modifications and variations of this invention can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only. The embodiments were chosen and described in order to best explain the principles of the invention and its practical applications, to thereby enable others skilled in the art to best utilize the invention and various embodiments with various modifications as are suited to the particular use contemplated. The invention is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled. 16622326 novo nordisk a/s USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open Apr 12th, 2022 11:42AM Apr 12th, 2022 11:42AM Novo Nordisk Health Care Pharmaceuticals & Biotechnology
nyse:nvo Novo Nordisk Apr 5th, 2022 12:00AM Aug 14th, 2017 12:00AM https://www.uspto.gov?id=US11295847-20220405 Systems and methods for adjusting basal administration timing Systems and methods are provided for optimizing basal administration timing in a standing basal insulin regimen of a subject. The regimen specifies a total amount of basal insulin medicament, one or more basal injection event types for a recurring period, and an apportionment of the total amount of medicament between the injection event types. Time stamped glucose measurements of the subject are obtained over a past time course comprising a plurality of instances of the recurring period. When the glucose measurements satisfy a stop condition, a recommended adjustment is determined that comprises a change in the number of injection event types in the regimen and/or a change in the apportionment of insulin medicament between injection event types. The recommended adjustment is communicated to the subject for manual adjustment of the regimen, an insulin pen charged with delivering the regimen to the subject, or a health care practitioner associated with the subject. 11295847 1. A device for optimizing basal administration timing in a standing basal insulin regimen for a subject, wherein the device comprises one or more processors and a memory, the memory storing instructions that, when executed by the one or more processors, perform a method of: obtaining the standing basal insulin regimen for the subject, wherein the standing basal insulin regimen specifies (i) a total amount of basal insulin medicament for a recurring period, (ii) one or more basal injection event types in a set of basal injection event types for the recurring period, and (iii) a respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types; obtaining, in the memory, from a continuous glucose monitor coupled with the subject, communicatively linked to the memory, and configured to continuously and autonomously provide physiological measurements comprising blood glucose of the subject, a first data set, the first data set comprising a plurality of glucose measurements of the subject over a past time course, the past time course comprising a first plurality of instances of the recurring period and, for each respective glucose measurement in the plurality of glucose measurements, a glucose measurement timestamp representing when the respective measurement was made; evaluating the plurality of glucose measurements over the past time course using a stop condition, wherein, when the stop condition is satisfied, the method further comprises: determining a recommended adjustment comprising a change in the number of basal injection event types in the standing basal insulin regimen and/or a change in the respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types; communicating the recommended adjustment to: (i) the subject for manual adjustment of the standing basal insulin regimen, (ii) the insulin pen charged with delivering the standing basal insulin regimen to the subject, or (iii) a health care practitioner associated with the subject; applying the recommended adjustment to a next recurring period, and wherein the standing basal insulin regimen specifies a single basal injection event type for the recurring period, and the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises: obtaining one or more fasting events in the past time course, wherein each fasting event is associated with a different instance of the recurring period in the first plurality of instances of the recurring period, comparing, for each respective fasting event in the one or more fasting events, (i) one or more first glucose measurements of the subject in the first data set occurring at a first time slot that is a first predetermined amount of time prior to a beginning of the respective fasting event to (ii) one or more second glucose measurements of the subject in the first data set occurring at a second time slot that is at a predetermined point within or after the respective fasting event, thereby obtaining one or more comparisons, wherein, the stop condition is satisfied when the one or more comparisons indicate that the respective one or more first glucose measurements deviate from the corresponding respective one or more second glucose measurements by more than a threshold amount, and the recommended adjustment is to increase the number of basal injection event types to two basal injection event types and to apportion the total amount of basal insulin medicament between the two basal injection event types. 2. The device of claim 1, wherein the standing basal insulin regimen specifies a single basal injection event type for the recurring period, and the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises: obtaining a plurality of fasting events in the past time course, wherein each fasting event is associated with a different instance of the recurring period in the first plurality of instances of the recurring period, obtaining, for each respective fasting event in the plurality of fasting events, (i) a first glucose measurements of the subject in the first data set occurring at a first time slot that is a first predetermined amount of time prior to a beginning of the respective fasting, and (ii) a second glucose measurements of the subject in the first data set occurring at a second time slot that is at a predetermined point within or after the respective fasting event, obtaining a first measure of central tendency of the first glucose measurement of each fasting event in the plurality of fasting event, and a second measure of central tendency of the second glucose measurement of each fasting in the plurality of fasting events, and comparing the first measure of central tendency to the second measure of central tendency, and thereby obtaining a comparison, wherein, the stop condition is satisfied when the comparison indicate that the respective first measure of central tendency deviate from the second measure of central tendency by more than a threshold amount, and  the recommended adjustment is to increase the number of basal injection event types to two basal injection event types and to apportion the total amount of basal insulin medicament between the two basal injection event types. 3. The device of claim 1, wherein the obtaining the one or more fasting events comprises identifying a first fasting event in a first recurring period in the first plurality of recurring periods by computing a moving period of variance σk2 across the plurality of glucose measurements, wherein: σ k 2 = ( 1 M ⁢ ∑ i = k - M + 1 k ⁢ ( G i - G ¯ ) ) 2 wherein, Gi is the ith glucose measurement in a portion k of the plurality of glucose measurements, M is a number of glucose measurements in the plurality of glucose measurements and represents the past time course, G is the mean of the glucose measurements selected from the plurality of glucose measurements, and k is within the first recurring period; and associating the first fasting event with a region of minimum variance min k ⁢ σ k 2 within the first recurring period. 4. The device of claim 1, wherein the obtaining the one or more fasting events comprises receiving an indication of each fasting event in the one or more fasting events from the subject. 5. The device of claim 1, wherein the obtaining the one or more fasting events comprises receiving a second data set from a wearable device worn by the subject, and the second data set indicates a physiological metric of the subject during the past time course that is indicative of the one or more fasting events. 6. The device of claim 1, the method further comprising: obtaining a third data set from an insulin pen used by the subject to apply the standing basal insulin regimen, the third data set comprising a plurality of insulin medicament records, each respective insulin medicament record in the plurality of medicament records comprising: (i) a respective insulin medicament injection event including an amount of basal insulin medicament injected into the subject and (ii) a corresponding insulin event electronic timestamp that is automatically generated by the insulin pen upon occurrence of the respective insulin medicament injection event, and using the third data set and the standing basal insulin regimen to determine one or more recurring periods in the past time course that do not comply with the standing basal insulin regimen for the subject; and excluding from the stop condition evaluation those glucose measurements in the one or more recurring periods in the past time course that do not comply with the standing basal insulin regimen. 7. The device of claim 1, wherein the set of basal injection event types for the recurring period consists of “morning basal,” and “night basal,” the standing basal insulin regimen specifies a single basal injection event type for the recurring period of “morning basal,” and the recommended adjustment is to add the “night basal” basal injection event type to the standing basal insulin regimen and to apportion the total amount of basal insulin medicament between the “night basal” basal injection event type and the “morning basal” basal injection event type. 8. The device of claim 1, wherein the method further comprises: obtaining a second data set from an insulin pen used by the subject to apply the standing basal insulin regimen, the second data set comprising a plurality of insulin medicament records over the past time course, each insulin medicament record in the plurality of medicament records comprising: (i) a respective insulin medicament injection event representing an insulin medicament injection of the basal insulin medicament into the subject using the insulin pen and (ii) a corresponding electronic timestamp that is automatically generated by the insulin pen upon occurrence of the respective insulin medicament injection event; and using the first data set and the second data set to simulate a glucose concentration of the subject over a future time course a plurality of times, thereby computing a plurality of simulations of the glucose concentration of the subject over the future time course, wherein each respective simulation in the plurality of simulations is associated with a different apportionment of the total amount of basal insulin medicament across the set of basal injection event types, the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises evaluating the glucose concentration of the subject across the future time course in each respective simulation in the plurality of simulations by calculating a glycaemic risk metric for each simulation, the stop condition is satisfied when a first simulation in the plurality of simulations minimizes the glycaemic risk metric, by more than a threshold amount as compared to one of: (i) a reference simulation of the glucose concentration of the subject over the future time course based upon the apportionment of the total amount of basal insulin medicament across the set of basal injection event types specified in the standing basal insulin regimen and (ii) the glucose concentration of the subject across the first data set, and the apportionment of the total amount of basal insulin medicament across the set of basal injection event types in the first simulation is different than that of the standing basal insulin regimen. 9. The device of claim 8, wherein the glycaemic risk metric comprises: (i) a total glucose level variability observed across the respective simulation, (ii) a variability in a plurality of fasting glucose levels calculated across the respective simulation, (iii) a percentage of time that a total glucose level exceeds a first threshold value or falls below a second threshold value across the respective simulation, or (iv) a percentage of time that an HbA1c level exceeds a third threshold value or falls below a fourth threshold value across the respective simulation. 10. The device of claim 8, the method further comprising: obtaining a meal record data set comprising a plurality of meal records over the past time course for the subject, each respective meal record in the meal record data set comprising: (i) a carbohydrate intake event and (ii) a corresponding electronic carbohydrate timestamp of when the carbohydrate intake event occurred; and wherein the using the first data set and the second data set to simulate a glucose concentration of the subject over a future time course a plurality of times comprises using the first data set, the second data set, and the meal record data set to simulate a glucose concentration of the subject over the future time course the plurality of times. 11. The device of claim 10, the method further comprising: obtaining a fourth data set comprising physical exertion of the subject over the past time course and wherein the using the first data set and the second data set to simulate a glucose concentration of the subject over a future time course a plurality of times comprises using the first data set, the second data set, the third data set and the fourth data set to simulate a glucose concentration of the subject over the future time course the plurality of times. 12. The device of claim 1, wherein the set of basal injection event types for the recurring period consists of “morning basal” and “night basal,” and the recurring period is a day. 13. The device of claim 1, wherein successive measurements in the plurality of glucose measurements in the first data set are autonomously taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less. 14. The device of claim 1, wherein the past time course is the last week, the last two weeks, or the last month and wherein the method is repeated on a recurring basis over time, and the basal insulin medicament consists of a single insulin medicament having a duration of action that is between 12 and 24 hours or a mixture of insulin medicaments that collectively have a duration of action that is between 12 and 24 hours. 15. A method for optimizing basal administration timing in a standing basal insulin regimen for a subject, the method comprising: obtaining the standing basal insulin regimen for the subject, wherein the standing basal insulin regimen specifies (i) a total amount of basal insulin medicament for a recurring period, (ii) one or more basal injection event types in a set of basal injection event types for the recurring period, and (iii) a respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types; obtaining, in a memory, from a continuous glucose monitor coupled with the subject, communicatively linked to the memory, and configured to continuously and autonomously provide physiological measurements comprising blood glucose of the subject, a first data set, the first data set comprising a plurality of glucose measurements of the subject over a past time course, the past time course comprising a first plurality of instances of the recurring period and, for each respective glucose measurement in the plurality of glucose measurements, a timestamp representing when the respective measurement was made; evaluating the plurality of glucose measurements over the past time course using a stop condition, wherein, when the stop condition is satisfied, the method further comprises: determining a recommended adjustment comprising a change in the number of basal injection event types in the standing basal insulin regimen and/or a change in the respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more periodic injection event types; communicating the recommended adjustment to: (i) the subject for manual adjustment of the basal insulin regimen, (ii) an insulin pen charged with delivering the standing basal insulin regimen to the subject, or (iii) a health care practitioner associated with the subject; applying the recommended adjustment to a next recurring period, and wherein the standing basal insulin regimen specifies a single basal injection event type for the recurring period, and the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises: obtaining one or more fasting events in the past time course, wherein each fasting event is associated with a different instance of the recurring period in the first plurality of instances of the recurring period, comparing, for each respective fasting event in the one or more fasting events, (i) one or more first glucose measurements of the subject in the first data set occurring at a first time slot that is a first predetermined amount of time prior to a beginning of the respective fasting event to (ii) one or more second glucose measurements of the subject in the first data set occurring at a second time slot that is at a predetermined point within or after the respective fasting event, thereby obtaining one or more comparisons, wherein, the stop condition is satisfied when the one or more comparisons indicate that the respective one or more first glucose measurements deviate from the corresponding respective one or more second glucose measurements by more than a threshold amount, and the recommended adjustment is to increase the number of basal injection event types to two basal injection event types and to apportion the total amount of basal insulin medicament between the two basal injection event types. 16. A non-transitory computer-readable data carrier having stored thereon computer program code that, when executed, causes a computer to perform steps of optimizing basal administration timing in a standing basal insulin regimen for a subject, comprising: obtaining the standing basal insulin regimen for the subject, wherein the standing basal insulin regimen specifies (i) a total amount of basal insulin medicament for a recurring period, (ii) one or more basal injection event types in a set of basal injection event types for the recurring period, and (iii) a respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types; obtaining, in a memory, from a continuous glucose monitor coupled with the subject, communicatively linked to the memory, and configured to continuously and autonomously provide physiological measurements comprising blood glucose of the subject, a first data set, the first data set comprising a plurality of glucose measurements of the subject over a past time course, the past time course comprising a first plurality of instances of the recurring period and, for each respective glucose measurement in the plurality of glucose measurements, a timestamp representing when the respective measurement was made; evaluating the plurality of glucose measurements over the past time course using a stop condition, wherein, when the stop condition is satisfied, the method further comprises: determining a recommended adjustment comprising a change in the number of basal injection event types in the standing basal insulin regimen and/or a change in the respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more periodic injection event types; and communicating the recommended adjustment to: (i) the subject for manual adjustment of the basal insulin regimen, (ii) an insulin pen charged with delivering the standing basal insulin regimen to the subject, or (iii) a health care practitioner associated with the subject; applying the recommended adjustment to a next recurring period, and wherein the standing basal insulin regimen specifies a single basal injection event type for the recurring period, and the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises: obtaining one or more fasting events in the past time course, wherein each fasting event is associated with a different instance of the recurring period in the first plurality of instances of the recurring period, comparing, for each respective fasting event in the one or more fasting events, (i) one or more first glucose measurements of the subject in the first data set occurring at a first time slot that is a first predetermined amount of time prior to a beginning of the respective fasting event to (ii) one or more second glucose measurements of the subject in the first data set occurring at a second time slot that is at a predetermined point within or after the respective fasting event, thereby obtaining one or more comparisons, wherein, the stop condition is satisfied when the one or more comparisons indicate that the respective one or more first glucose measurements deviate from the corresponding respective one or more second glucose measurements by more than a threshold amount, and the recommended adjustment is to increase the number of basal injection event types to two basal injection event types and to apportion the total amount of basal insulin medicament between the two basal injection event types. 16 CROSS-REFERENCE TO RELATED APPLICATIONS This application is a 35 U.S.C. § 371 National Stage application of International Application PCT/EP2017/070588 (published as WO 2018/036854), filed Aug. 14, 2017, which claims priority to European Patent Application 16185931.9, filed Aug. 26, 2016, the contents of all above-named applications are incorporated herein by reference. TECHNICAL FIELD The present disclosure relates generally to systems and methods for optimizing basal administration timing in a standing insulin regimen for a subject that includes a basal insulin medicament. BACKGROUND Type 2 diabetes mellitus is characterized by progressive disruption of normal physiologic insulin secretion. In healthy individuals, basal insulin secretion by pancreatic β cells occurs continuously to maintain steady glucose levels for extended periods between meals. Also in healthy individuals, there is prandial secretion in which insulin is rapidly released in an initial first-phase spike in response to a meal, followed by prolonged insulin secretion that returns to basal levels after 2-3 hours. Insulin is a hormone that binds to insulin receptors to lower blood glucose by facilitating cellular uptake of glucose, amino acids, and fatty acids into skeletal muscle and fat and by inhibiting the output of glucose from the liver. In normal healthy individuals, physiologic basal and prandial insulin secretions maintain euglycemia, which affects fasting plasma glucose and postprandial plasma glucose concentrations. Basal and prandial insulin secretion is impaired in Type 2 diabetes and early post-meal response is absent. To address these adverse events, subjects with Type 2 diabetes are provided with insulin medicament treatment regimens. Subjects with Type 1 diabetes are also provided with insulin medicament treatment regimens. The goal of these insulin medicament treatment regimens is to maintain a desired fasting blood glucose target level that will minimize estimated risk of hypo- and hyper-glycaemia. Traditional insulin medicament delivery systems have included the use of pump systems that provide a frequent recurrent dosage of insulin medicament. More recently, additional types of delivery systems have been developed, such as insulin pens, which can be used to self-administer insulin medicament treatment regimens in the form of less frequent insulin medicament injections. A common approach to diabetes treatment using such delivery systems is to inject a long acting insulin medicament (basal) dosage in accordance with the standing insulin regimen to maintain glycaemic control independent of meal events. However, the optimal delivery of such long acting insulin medicament (basal) dosage is subject specific. In some subjects it is best to split the basal dosage into two or more dosages per day in order to best maintain glycaemic control whereas in other subjects it is best to administer the basal dosage at a single time each day. In practice, a health care practitioner typically determines a total daily amount of basal insulin medicament a subject should take. Moreover, the health care practitioner determines, based on available medical data about the subject, whether this daily basal insulin medicament allotment should be administered as a single injection event, or in multiple injection events over the course of the day. The determination made by the health care practitioner is formalized as a standing basal insulin regimen which the subject is directed to adhere to until the next visit by the subject to the health care practitioner. The drawback with this conventional practice is twofold. First, the health care practitioner conventionally uses limited data to draw upon in order to establish the standing basal insulin regimen. Thus, the standing basal insulin regimen risks not being optimal for a given subject due to the limited data used to formalize the standing basal insulin regimen. Second, subjects are not able to visit the health care practitioner frequently enough to truly optimize the standing basal insulin regimen. For instance, the optimal standing basal insulin regimen may shift between health care practitioner visits due to health events that occur between visits. These shifts in the optimal standing basal insulin regimen are not discovered until the next health care practitioner visit. These drawbacks with conventional practice result in some subjects administering the basal insulin medicament as more than one dose over the course of the day when, in fact, it would be better for such subjects to administer the basal insulin medicament as a single dose. Conversely, other subjects that are directed to administer their basal insulin medicament in a single injection event each day are better served, in fact, if they were to break up the daily basal insulin medicament dosage into smaller dosages that are injected at multiple times during the day. United States Patent Publication No. 20110098548 entitled “Methods for Modeling Insulin Therapy Requirements” to Abbott Diabetes Care Inc. discloses a system for processing diabetes related information, including glucose information, for predicting future glucose levels as a function of glucose data, carbohydrate intake, insulin delivery history and exercise history and subsequently providing recommendations related to the predicted future glucose levels. However, the 20110098548 provides no teaching on how to apportion a recurring basal insulin medicament dosage into a discrete set of injection events (e.g., a single injection event or multiple injection events) in order to minimize glycaemic risk. International Patent Publication WO2015/169814 entitled “Insulin Dosage Proposal System” to Joanneum Res Forschungs GMBH discloses systems and methods for determining a proposal for an insulin dosage that includes doses and assigned times of the day according to which insulin is to be administered to a diabetes patient. However, like the 20110098458 publication, the WO2015/169814 publication provides no teaching on how to apportion a recurring basal insulin medicament dosage into a discrete set of injection events (e.g., a single injection event or multiple injection events) in order to minimize glycaemic risk. International Patent Publication WO 2010/091129 entitled “Multi-Function Analyte Test Device and Methods Therefore” to Abbott Diabetes Care discloses a health monitoring device that may include instruction to perform a long-acting medication dosage calculation function. A long-acting medication may be a medication wherein a single dose may last for up to 12 hours, 24 hours, or longer. The instructions for a long-acting medication dosage calculation function may be in the form of software stored on the memory device and executed by the processor of the health monitor device. In one aspect, the long-acting medication dosage calculation function may be an algorithm based on the current concentration of an analyte of a patient, wherein the long-acting medication dosage calculation function compares the current analyte concentration value to a predetermined threshold, which may be based on clinically determined threshold levels for a particular analyte, or may be tailored for individual patients by a doctor or other treating professional. If the current analyte concentration is above the predetermined threshold, the long-acting medication dosage calculation function may use the current analyte concentration value to calculate a recommended dosage of a long-acting medication. Once calculated, the recommended medication dosage may be displayed on the display unit of the health monitor device. However, the WO 2010/091129 publication provides no teaching on how to apportion a recurring basal insulin medicament dosage into a discrete set of injection events (e.g., a single injection event or multiple injection events) in order to minimize glycaemic risk. Given the above background, what is needed in the art are systems and methods for optimizing basal administration timing in order to minimize glycaemic risk and for communicating this information to subjects so that the basal insulin medicament is administered with insulin pens in accordance with the optimal basal administration timing. SUMMARY The present disclosure addresses the need in the art for optimizing basal administration timing in a standing basal insulin regimen of a subject specifying a total amount of basal insulin medicament, one or more basal injection event types for a recurring period (e.g., a day), and an apportionment of the total amount of medicament between the injection event types in the recurring period. Timestamped glucose measurements of the subject are obtained over a past time course. This past time course comprises a plurality of instances of the recurring period (e.g., several days, where the recurring period is a single day). When the glucose measurements satisfy a stop condition, a recommended adjustment is determined. This recommended adjustment comprises a change in the number of injection event types in the standing basal insulin regimen and/or a change in the apportionment of insulin medicament between injection event types in the recurring period. The recommended adjustment is communicated to the subject for manual adjustment of the standing basal insulin regimen, an insulin pen charged with delivering the standing basal insulin regimen to the subject, or a health care practitioner associated with the subject. As such, one aspect of the present disclosure provides a device for optimizing basal administration timing in a standing basal insulin regimen for a subject. The device comprises one or more processors and a memory. The memory stores instructions that, when executed by the one or more processors, performs a method. In the method, the standing basal insulin regimen for the subject is obtained. The standing basal insulin regimen specifies (i) a total amount of basal insulin medicament for a recurring period (e.g., a one day period, a two day period, etc.), (ii) one or more basal injection event types in a set of basal injection event types for the recurring period, and (iii) a respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types in a given recurring period. In the method, a first data set is also obtained. The first data set comprises a plurality of glucose measurements of the subject over a past time course. The past time course comprises a first plurality of instances of the recurring period. For instance, if the recurring period is a day, the past time course comprises several days. For each respective glucose measurement in the plurality of glucose measurements, there is a glucose measurement timestamp representing when the respective measurement was made. In the method, the plurality of glucose measurements over the past time course is evaluated using a stop condition. When the stop condition is satisfied, the method further comprises determining a recommended adjustment to the standing basal insulin regimen. As such, the recommended adjustment comprises a change in the number of basal injection event types in the standing basal insulin regimen and/or a change in the respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types. The recommended adjustment is communicated to: (i) the subject for manual adjustment of the basal insulin regimen, (ii) an insulin pen charged with delivering the standing basal insulin regimen to the subject, or (iii) a health care practitioner associated with the subject. In other words the device communicates an apportionment of the total basal insulin between one or more basal injection event types, without increasing the amount of basal insulin, i.e., the total basal insulin. The stop condition indicates that data based on the first data set has been analysed and when the stop condition is evaluated against the analysed data, a satisfied stop condition indicates that the apportionment and/or the number of basal injection event types should be changed. The analysis and the stop condition can be based directly on the measurements or on predictions based on the measurements. The stop condition can be a comparison of glucose measurements related to a fasting period, or the stop condition can be an indication on that a change in apportionment or number of basal injection event types will affect the glycaemic risk to the better or worse. In some embodiments, the standing basal insulin regimen specifies a single basal injection event type for the recurring period and the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises obtaining one or more fasting events in the past time course. Each such fasting event is associated with a different instance of the recurring period in the first plurality of instances of the recurring period. For instance, if the recurring period is a day, each respective fasting period is associated with a different day in the past time course. That is, if the recurring period is one day, an example of a “different instance” of the recurring period would be a particular day, such as Tuesday, May 5. For each respective fasting event in the one or more fasting events, a comparison is made between (i) one or more first glucose measurements of the subject in the first data set occurring at a first time slot that is a first predetermined amount of time prior to a beginning of the respective fasting event and (ii) one or more second glucose measurements of the subject in the first data set occurring at a second time slot that is at a predetermined point within or after the respective fasting event. In this way, one or more comparisons are obtained. In such embodiments, the stop condition is satisfied when the one or more comparisons indicate that the respective one or more first glucose measurements deviate from the corresponding respective one or more second glucose measurements by more than a threshold amount. In this case, the recommended adjustment is to increase the number of basal injection event types to two basal injection event types and to apportion the total amount of basal insulin medicament between the two basal injection event types. In a further or alternative aspect, the standing basal insulin regimen specifies a single basal injection event type for the recurring period, and the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises: obtaining a plurality of fasting events in the past time course, wherein each fasting event is associated with a different instance of the recurring period in the first plurality of instances of the recurring period, obtaining, for each respective fasting event in the plurality of fasting events, (i) a first glucose measurements of the subject in the first data set occurring at a first time slot that is a first predetermined amount of time prior to a beginning of the respective fasting, and (ii) a second glucose measurements of the subject in the first data set occurring at a second time slot that is at a predetermined point within or after the respective fasting event, obtaining a first measure of central tendency of the first glucose measurement of each fasting event in the plurality of fasting event, and a second measure of central tendency of the second glucose measurement of each fasting in the plurality of fasting events, and comparing the first measure of central tendency to the second measure of central tendency, and thereby obtaining a comparison, wherein, the stop condition is satisfied when the comparison indicate that the respective first measure of central tendency deviate from the second measure of central tendency by more than a threshold amount, and the recommended adjustment is to increase the number of basal injection event types to two basal injection event types and to apportion the total amount of basal insulin medicament between the two basal injection event types. In some such embodiments, the obtaining the one or more fasting events comprises identifying a first fasting event in a first recurring period in the first plurality of recurring periods by computing a moving period of variance σk2 across the plurality of glucose measurements, where σ k 2 = ( 1 M ⁢ ∑ i = k - M + 1 k ⁢ ( G i - G ¯ ) ) 2 where Gi is the ith glucose measurement in a portion k of the plurality of glucose measurements, M is a number of glucose measurements in the plurality of glucose measurements and represents the past time course, G is the mean of the glucose measurements selected from the plurality of glucose measurements, and k is within the first recurring period. In such embodiments, the first fasting event is associated with a region of minimum variance min k ⁢ σ k 2 within the first recurring period. In alternative embodiments, the obtaining the one or more fasting events comprises receiving an indication of each fasting event in the one or more fasting events from the subject. In still further alternative embodiments, the obtaining the one or more fasting events comprises receiving a second data set from a wearable device worn by the subject, and the second data set indicates a physiological metric of the subject during the time course that is indicative of the one or more fasting events. In some embodiments, the method further comprises obtaining a third data set from an insulin pen used by the subject to apply the standing basal insulin regimen. The third data set comprises a plurality of insulin medicament records. Each respective insulin medicament record in the plurality of medicament records comprises: (i) a respective insulin medicament injection event including an amount of basal insulin medicament injected into the subject and (ii) a corresponding insulin event electronic timestamp that is automatically generated by the insulin pen upon occurrence of the respective insulin medicament injection event. In the method, the third data set and the standing basal insulin regimen is used to determine one or more recurring periods in the past time course that do not comply with the standing basal insulin regimen for the subject. The glucose measurements taken during such recurring periods that do not comply with the standing basal insulin regimen for the subject are excluded from the stop condition evaluation. In some embodiments, the set of basal injection event types for the recurring period consists of “morning basal” and “night basal.” Further, the standing basal insulin regimen specifies a single basal injection event type for the recurring period. Further still, the recommended adjustment is to add the “night basal” basal injection event type to the standing basal insulin regimen and to apportion the total amount of basal insulin medicament between the “night basal” basal injection event type and the “morning basal” basal injection event type. In some embodiments, the method further comprises obtaining a second data set from an insulin pen used by the subject to apply the standing basal insulin regimen. The second data set comprises a plurality of insulin medicament records over the past time course. Each insulin medicament record in the plurality of medicament records comprises: (i) a respective insulin medicament injection event representing an insulin medicament injection of the basal insulin medicament into the subject using the insulin pen and (ii) a corresponding electronic timestamp that is automatically generated by the insulin pen upon occurrence of the respective insulin medicament injection event. The first data set and the second data set are used to simulate a glucose concentration of the subject over a future time course a plurality of times, thereby computing a plurality of simulations of the glucose concentration of the subject over the future time course. Each respective simulation in the plurality of simulations is associated with a different apportionment of the total amount of basal insulin medicament across the set of basal injection event types. In such embodiments, the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises evaluating the glucose concentration of the subject across the future time course in each respective simulation in the plurality of simulations by calculating a glycaemic risk metric for each simulation. The stop condition is satisfied when a first simulation in the plurality of simulations minimizes the glycaemic risk metric, by more than a threshold amount as compared to one of (i) a reference simulation of the glucose concentration of the subject over the future time course based upon the apportionment of the total amount of basal insulin medicament across the set of basal injection event types specified in the standing basal insulin regimen and (ii) the glucose concentration of the subject across the first data set. In such embodiments, the apportionment of the total amount of basal insulin medicament across the set of basal injection event types in the first simulation is different than that of the standing basal insulin regimen. In some such embodiments, the glycaemic risk metric comprises: (i) a total glucose level variability observed across the respective simulation, (ii) a variability in a plurality of fasting glucose levels calculated across the respective simulation, (iii) a percentage of time that a total glucose level exceeds a first threshold value or falls below a second threshold value across the respective simulation, or (iv) a percentage of time that an HbA1c level exceeds a third threshold value or falls below a fourth threshold value across the respective simulation. In some such embodiments, a meal record data set is obtained. The meal record data set comprises a plurality of meal records over the past time course for the subject. Each respective meal record in the meal record data set comprises: (i) a carbohydrate intake event and (ii) a corresponding electronic carbohydrate timestamp of when the carbohydrate intake event occurred. In such embodiments, the using the first data set and the second data set to simulate a glucose concentration of the subject over a future time course a plurality of times comprises using the first data set, the second data set, and the meal record data set to simulate a glucose concentration of the subject over the future time course the plurality of times. In some such embodiments, the method further comprises obtaining a fourth data set comprising physical exertion of the subject over the past time course. In such embodiments, the using the first data set and the second data set to simulate a glucose concentration of the subject over a future time course a plurality of times comprises using the first data set, the second data set, the third data set and the fourth data set to simulate a glucose concentration of the subject over the future time course the plurality of times. In some embodiments, the set of basal injection event types for the recurring period consists of “morning basal” and “night basal” and the recurring period is a day. In some embodiments, successive measurements in the plurality of glucose measurements in the first data set are autonomously taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less. In some embodiments, the past time course is the last week, the last two weeks, or the last month and the method is repeated on a recurring basis over time. Further, the basal insulin medicament consists of a single insulin medicament having a duration of action that is between 12 and 24 hours or a mixture of insulin medicaments that collectively have a duration of action that is between 12 and 24 hours. Another aspect of the present disclosure provides a method for a method for optimizing basal administration timing in a standing basal insulin regimen for a subject. The method comprises obtaining the standing basal insulin regimen for the subject. The standing basal insulin regimen specifies (i) a total amount of basal insulin medicament for a recurring period, (ii) one or more basal injection event types in a set of basal injection event types for the recurring period, and (iii) a respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types. In the method, a first data set is obtained. The first data set comprises a plurality of glucose measurements of the subject over a past time course. The past time course comprises a first plurality of instances of the recurring period. The first data set further comprises, for each respective glucose measurement in the plurality of glucose measurements, a timestamp representing when the respective measurement was made. In the methods, the plurality of glucose measurements are evaluated over the past time course using a stop condition. When the stop condition is satisfied, the method further comprises determining a recommended adjustment comprising a change in the number of basal injection event types in the standing basal insulin regimen and/or a change in the respective apportionment of all or a portion of the total amount of basal insulin medicament between each respective basal injection event type in the one or more periodic injection event types. The recommended adjustment is communicated to: (i) the subject for manual adjustment of the basal insulin regimen, (ii) an insulin pen charged with delivering the standing basal insulin regimen to the subject, or (iii) a health care practitioner associated with the subject. In a further aspect is provided a computer program comprising instructions that, when executed by one or more processors, perform the method of: obtaining the standing basal insulin regimen for the subject, wherein the standing basal insulin regimen specifies (i) a total amount of basal insulin medicament for a recurring period, (ii) one or more basal injection event types in a set of basal injection event types for the recurring period, and (iii) a respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types; obtaining a first data set, the first data set comprising a plurality of glucose measurements of the subject over a past time course, the past time course comprising a first plurality of instances of the recurring period and, for each respective glucose measurement in the plurality of glucose measurements, a glucose measurement timestamp representing when the respective measurement was made; evaluating the plurality of glucose measurements over the past time course using a stop condition, wherein, when the stop condition is satisfied, the method further comprises: determining a recommended adjustment comprising a change in the number of basal injection event types in the standing basal insulin regimen and/or a change in the respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types; and communicating the recommended adjustment to: (i) the subject for manual adjustment of the standing basal insulin regimen, (ii) an insulin pen charged with delivering the standing basal insulin regimen to the subject, or (iii) a health care practitioner associated with the subject. In a further aspect is provided, a computer-readable data carrier having stored thereon the computer program as described above. BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 illustrates an exemplary system topology that includes a regimen adjustment device for optimizing basal administration timing in a standing basal insulin regimen for a subject, a data collection device for collecting subject data, one or more glucose sensors that measure glucose data from the subject, and one or more insulin pens that are used by the subject to inject insulin medicaments in accordance with the standing basal insulin regimen, where the above-identified components are interconnected, optionally through a communications network, in accordance with an embodiment of the present disclosure. FIG. 2 illustrates a device for optimizing basal administration timing in a standing basal insulin regimen for a subject in accordance with an embodiment of the present disclosure. FIG. 3 illustrates a device for optimizing basal administration timing in a standing basal insulin regimen for a subject in accordance with another embodiment of the present disclosure. FIGS. 4A, 4B, 4C, 4D, and 4E collectively provide a flow chart of processes and features of a device for optimizing basal administration timing in a standing basal insulin regimen for a subject, where optional elements of the flow chart are indicated by dashed boxes, in accordance with various embodiments of the present disclosure. FIG. 5 illustrates an example integrated system of connected insulin pen(s), continuous glucose monitor(s), memory and a processor for optimizing basal administration timing in a standing basal insulin regimen for a subject in accordance with an embodiment of the present disclosure. FIG. 6 illustrates instances where one or more glucose measurements from a second time slot trends higher than one or more glucose measurements from a first time slot (panel A) and where one or more glucose measurements from a second time slot trends lower than one or more glucose measurements from a first time slot (panel B) in accordance with an embodiment of the present disclosure. FIG. 7 illustrates how an optimization algorithm searches for a recommended adjustment comprising a change in a number of basal injection event types in a standing basal insulin regimen and/or a change in the respective apportionment of a total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types by simulating scenarios and measuring outcome in accordance with an embodiment of the present disclosure. The algorithm then suggests the recommended adjustment with the best outcome in accordance with an embodiment of the present disclosure. FIG. 8 illustrates an exemplary algorithm for optimizing basal administration timing in a standing basal insulin regimen for a subject in accordance with an embodiment of the present disclosure. Like reference numerals refer to corresponding parts throughout the several views of the drawings. DETAILED DESCRIPTION The present disclosure provides systems and methods for optimizing basal administration timing in a standing basal insulin regimen for a subject. FIG. 1 illustrates an example of an integrated system 502 for optimizing basal administration timing in a standing basal insulin regimen for a subject, and FIG. 5 provides more details of such a system 502. The integrated system 502 includes one or more connected insulin pens 104, one or more glucose monitors 102, memory 506, and a processor (not shown) for adjusting a basal/bolus ratio in a standing insulin regimen for a subject. In some embodiments, a glucose monitor 102 is a continuous glucose monitor. In FIG. 5, optional physiological measurements 230 in the form of a second data set 228, continuous glucose measurements in the form of a first data set 216, and optional insulin dose/timestamp/type data in the form of a third data set 302 are filtered 504, stored in memory 192/290 (step 506) and a determination on whether to make a recommended adjustment to the standing insulin regimen 206 is made at step 508. When a recommended adjustment to the standing insulin regimen is made it is then communicated to a subject 510 With the integrated system, the basal administration timing in a standing basal insulin regimen is optimized for a subject, where the standing basal insulin regimen specifies (i) a total amount of basal insulin medicament for a recurring period (e.g., a day), (ii) one or more basal injection event types in a set of basal injection event types (e.g., “morning basal,” “evening basal”) for the recurring period, and (iii) a respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types. Timestamped glucose measurements of the subject are obtained over a past time course comprising a plurality of instances of the recurring period. When the glucose measurements satisfy a stop condition, a recommended adjustment is determined that comprises a change in the number of injection event types in the standing basal insulin regimen and/or a change in the apportionment of insulin medicament between injection event types. The recommended adjustment is communicated to the subject for manual adjustment of the standing basal insulin regimen, an insulin pen charged with delivering the standing basal insulin regimen to the subject, or a health care practitioner associated with the subject. Reference will now be made in detail to embodiments, examples of which are illustrated in the accompanying drawings. In the following detailed description, numerous specific details are set forth in order to provide a thorough understanding of the present disclosure. However, it will be apparent to one of ordinary skill in the art that the present disclosure may be practiced without these specific details. In other instances, well-known methods, procedures, components, circuits, and networks have not been described in detail so as not to unnecessarily obscure aspects of the embodiments. It will also be understood that, although the terms first, second, etc. may be used herein to describe various elements, these elements should not be limited by these terms. These terms are only used to distinguish one element from another. For example, a first subject could be termed a second subject, and, similarly, a second subject could be termed a first subject, without departing from the scope of the present disclosure. The first subject and the second subject are both subjects, but they are not the same subject. Furthermore, the terms “subject,” “user,” and “patient” are used interchangeably herein. By the term “insulin pen,” is meant an injection device suitable for applying discrete doses of insulin, where the injection device is adapted for logging and communicating dose related data. By the term “injection event,” is meant the use of an insulin pen to apply a discrete dose of an insulin medicament. The terminology used in the present disclosure is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used in the description of the invention and the appended claims, the singular forms “a”, “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. It will also be understood that the term “and/or” as used herein refers to and encompasses any and all possible combinations of one or more of the associated listed items. It will be further understood that the terms “comprises” and/or “comprising,” when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. As used herein, the term “if” may be construed to mean “when” or “upon” or “in response to determining” or “in response to detecting,” depending on the context. Similarly, the phrase “if it is determined” or “if [a stated condition or event] is detected” may be construed to mean “upon determining” or “in response to determining” or “upon detecting [the stated condition or event]” or “in response to detecting [the stated condition or event],” depending on the context. A detailed description of a system 48 for optimizing basal administration timing in a standing basal insulin regimen for a subject in accordance with the present disclosure is described in conjunction with FIGS. 1, 2, 3, and 5. As such, FIGS. 1, 2, 3, and 5 collectively illustrate the topology of the system in accordance with the present disclosure. In the topology, there is a regimen adjustment device for optimizing basal administration timing in a standing basal insulin regimen for a subject (“regimen adjustment device 250”) (FIGS. 1, 2, and 3), a device for data collection (“data collection device 200”), one or more glucose sensors 102 associated with the subject (FIGS. 1 and 5), and one or more insulin pens 104 for injecting insulin medicaments into the subject (FIGS. 1 and 5). Throughout the present disclosure, the data collection device 200 and the regimen adjustment device 250 will be referenced as separate devices solely for purposes of clarity. That is, the disclosed functionality of the data collection device 200 and the disclosed functionality of the regimen adjustment device 250 are contained in separate devices as illustrated in FIG. 1. However, it will be appreciated that, in fact, in some embodiments, the disclosed functionality of the data collection device 200 and the disclosed functionality of the regimen adjustment device 250 are contained in a single device. In some embodiments, the disclosed functionality of the data collection device 200 and/or the disclosed functionality of the regimen adjustment device 250 are contained in a single device and this single device is a glucose monitor 102 or an insulin pen 104. Referring to FIG. 1, the regimen adjustment device 250 optimizes the basal administration timing in a standing basal insulin regimen for a subject. To do this, the data collection device 200, which is in electrical communication with the regimen adjustment device 250, receives glucose measurements originating from one or more glucose sensors 102 attached to a subject on an ongoing basis. In some embodiments, the data collection device 200 also receives insulin medicament injection data from one or more insulin pens 104 used by the subject to inject insulin medicaments. In some embodiments, the data collection device 200 receives such data directly from the glucose sensor(s) 102 and insulin pens 104 used by the subject. For instance, in some embodiments the data collection device 200 receives this data wirelessly through radio-frequency signals. In some embodiments such signals are in accordance with an 802.11 (WiFi), Bluetooth, or ZigBee standard. In some embodiments, the data collection device 200 receives such data directly, analyzes the data, and passes the analyzed data to the regimen adjustment device 250. In some embodiments, a glucose sensor 102 and/or insulin pen 104 includes an RFID tag and communicates to the data collection device 200 and/or the regimen adjustment device 250 using RFID communication. In some embodiments, referring to FIG. 2, the data collection device 200 also obtains or receives physiological measurements 232 of the subject (e.g., from wearable physiological measurement devices, from measurement devices within the data collection device 200 such as a magnetometer or a thermostat, etc.). In some embodiments, the data collection device 200 and/or the regimen adjustment device 250 is not proximate to the subject and/or does not have wireless capabilities or such wireless capabilities are not used for the purpose of acquiring glucose data, insulin medicament injection data, and/or physiological measurement data. In such embodiments, a communication network 106 may be used to communicate glucose measurements from the glucose sensor(s) 102 to the data collection device 200 and/or the regimen adjustment device 250, insulin medicament injection data from the one or more insulin pens 104 to the data collection device 200 and/or the regimen adjustment device 250, and/or physiological measurement data from one or more physiological measurement devices (not shown) to the data collection device 200 and/or the regimen adjustment device 250. Examples of networks 106 include, but are not limited to, the World Wide Web (WWW), an intranet and/or a wireless network, such as a cellular telephone network, a wireless local area network (LAN) and/or a metropolitan area network (MAN), and other devices by wireless communication. The wireless communication optionally uses any of a plurality of communications standards, protocols and technologies, including but not limited to Global System for Mobile Communications (GSM), Enhanced Data GSM Environment (EDGE), high-speed downlink packet access (HSDPA), high-speed uplink packet access (HSUPA), Evolution, Data-Only (EV-DO), HSPA, HSPA+, Dual-Cell HSPA (DC-HSPDA), long term evolution (LTE), near field communication (NFC), wideband code division multiple access (W-CDMA), code division multiple access (CDMA), time division multiple access (TDMA), Bluetooth, Wireless Fidelity (Wi-Fi) (e.g., IEEE 802.11a, IEEE 802.11ac, IEEE 802.11ax, IEEE 802.11b, IEEE 802.11g and/or IEEE 802.11n), voice over Internet Protocol (VoIP), Wi-MAX, a protocol for e-mail (e.g., Internet message access protocol (IMAP) and/or post office protocol (POP)), instant messaging (e.g., extensible messaging and presence protocol (XMPP), Session Initiation Protocol for Instant Messaging and Presence Leveraging Extensions (SIMPLE), Instant Messaging and Presence Service (IMPS)), and/or Short Message Service (SMS), or any other suitable communication protocol, including communication protocols not yet developed as of the filing date of the present disclosure. In some embodiments, there is a single glucose sensor 102 attached to the subject and the data collection device 200 and/or the regimen adjustment device 250 is part of the glucose sensor 102. That is, in some embodiments, the data collection device 200 and/or the regimen adjustment device 250 and the glucose sensor 102 are a single device. In some embodiments, the data collection device 200 and/or the regimen adjustment device 250 is part of an insulin pen. That is, in some embodiments, the data collection device 200 and/or the regimen adjustment device 250 and an insulin pen 104 are a single device. Of course, other topologies of the system 48 are possible. For instance, rather than relying on a communications network 106, the one or more glucose sensors 102 and the one or more insulin pens 104 may wirelessly transmit information directly to the data collection device 200 and/or regimen adjustment device 250. Further, the data collection device 200 and/or the regimen adjustment device 250 may constitute a portable electronic device, a server computer, or in fact constitute several computers that are linked together in a network or be a virtual machine in a cloud computing context. As such, the exemplary topology shown in FIG. 1 merely serves to describe the features of an embodiment of the present disclosure in a manner that will be readily understood to one of skill in the art. Referring to FIG. 2, in typical embodiments, the regimen adjustment device 250 comprises one or more computers. For purposes of illustration in FIG. 2, the regimen adjustment device 250 is represented as a single computer that includes all of the functionality for optimizing basal administration timing in a standing basal insulin regimen for a subject. However, the disclosure is not so limited. In some embodiments, the functionality for optimizing basal administration timing in a standing basal insulin regimen for a subject is spread across any number of networked computers and/or resides on each of several networked computers and/or is hosted on one or more virtual machines at a remote location accessible across the communications network 106. One of skill in the art will appreciate that any of a wide array of different computer topologies are used for the application and all such topologies are within the scope of the present disclosure. Turning to FIG. 2 with the foregoing in mind, an exemplary regimen adjustment device 250 for optimizing basal administration timing in a standing basal insulin regimen for a subject comprises one or more processing units (CPU's) 274, a network or other communications interface 284, a memory 192 (e.g., random access memory), one or more magnetic disk storage and/or persistent devices 290 optionally accessed by one or more controllers 288, one or more communication busses 213 for interconnecting the aforementioned components, a user interface 278, the user interface 278 including a display 282 and input 280 (e.g., keyboard, keypad, touch screen), and a power supply 276 for powering the aforementioned components. In some embodiments, data in memory 192 is seamlessly shared with non-volatile memory 290 using known computing techniques such as caching. In some embodiments, memory 192 and/or memory 290 includes mass storage that is remotely located with respect to the central processing unit(s) 274. In other words, some data stored in memory 192 and/or memory 290 may in fact be hosted on computers that are external to the regimen adjustment device 250 but that can be electronically accessed by the regimen adjustment device 250 over an Internet, intranet, or other form of network or electronic cable (illustrated as element 106 in FIG. 2) using network interface 284. In some embodiments, the memory 192 of the regimen adjustment device 250 for optimizing basal administration timing in a standing basal insulin regimen for a subject stores: an operating system 202 that includes procedures for handling various basic system services; a basal timing adjustment module 204; a standing insulin regimen 206 for the subject, the standing insulin regimen comprising a recurring period 208 and, for each instance of the recurring period, a total amount of basal insulin medicament 210 that is to be administered in the recurring period through execution of one or more basal injection event types, where each basal injection event type 212 is associated with a respective apportionment 214 of the total amount of basal insulin medicament; a first data set 216, the first data set comprising a plurality of glucose measurements of the subject over a past time course, and for each respective glucose measurement 218 in the plurality of glucose measurements, a glucose measurement timestamp 220 representing when the respective glucose measurement was made; a stop condition 222 that is used to evaluation the plurality of glucose measurements of the subject over the past time course; a fasting event data set 224 comprising a plurality of fasting events that occur in the past time course encompassed by the first data set, each such fasting event 226 associated with a different recurring period instance 208 within the past time course; an optional second data set 228 that comprises one or more physiological metrics and, for each respective physiological metric 230 in the one or more physiological metrics, one or more physiological metric measurements 232 of the subject; a plurality of simulations of the glucose concentration of the subject over a future time course, each respective glucose concentration simulation 234 based upon the first data set and the second data set and associated with a different apportionment 236 of the total amount of basal insulin medicament across the set of basal injection event types thereby providing a glucose concentration over the future time course 238 and an associated glycaemic risk metric 240; and an optional reference glucose simulation 242 across the future time course based upon the apportionment 236 of the total amount of basal insulin medicament as set forth in the standing insulin regimen 206. In some embodiments, the physiological metric measurement 233 is body temperature of the subject. In some embodiments, the physiological metric measurement 233 is a measurement of activity of the subject. In some embodiments, these physiological metric measurements serve as an additional input for optimizing basal administration timing in a standing basal insulin regimen for a subject. In some embodiments, the optional accelerometer 317, optional GPS 319, and/or magnetometer (not shown) of the regimen adjustment device 250 or such components optionally within the one or more glucose monitors 102 and/or the one or more insulin pens 104 is used to acquire such physiological metric measurements 232. In some embodiments, the basal timing adjustment module 204 is accessible within any browser (phone, tablet, laptop/desktop). In some embodiments, the basal timing adjustment module 204 runs on native device frameworks, and is available for download onto the regimen adjustment device 250 running an operating system 202 such as Android or iOS. In some implementations, one or more of the above identified data elements or modules of the regimen adjustment device 250 for optimizing basal administration timing in a standing basal insulin regimen for a subject are stored in one or more of the previously described memory devices, and correspond to a set of instructions for performing a function described above. The above-identified data, modules or programs (e.g., sets of instructions) need not be implemented as separate software programs, procedures or modules, and thus various subsets of these modules may be combined or otherwise re-arranged in various implementations. In some implementations, the memory 192 and/or 290 optionally stores a subset of the modules and data structures identified above. Furthermore, in some embodiments, the memory 192 and/or 290 stores additional modules and data structures not described above. In some embodiments, a regimen adjustment device 250 for optimizing basal administration timing in a standing basal insulin regimen for a subject is a smart phone (e.g., an iPHONE), laptop, tablet computer, desktop computer, or other form of electronic device (e.g., a gaming console). In some embodiments, the regimen adjustment device 250 is not mobile. In some embodiments, the regimen adjustment device 250 is mobile. FIG. 3 provides a further description of a specific embodiment of a regimen adjustment device 250 in accordance with the instant disclosure. The regimen adjustment device 250 illustrated in FIG. 3 has one or more processing units (CPU's) 274, peripherals interface 370, memory controller 368, a network or other communications interface 284, a memory 192 (e.g., random access memory), a user interface 278, the user interface 278 including a display 282 and input 280 (e.g., keyboard, keypad, touch screen), an optional accelerometer 317, an optional GPS 319, optional audio circuitry 372, an optional speaker 360, an optional microphone 362, one or more optional intensity sensors 364 for detecting intensity of contacts on the regimen adjustment device 250 (e.g., a touch-sensitive surface such as a touch-sensitive display system 282 of the regimen adjustment device 250), an optional input/output (I/O) subsystem 366, one or more optional optical sensors 373, one or more communication busses 213 for interconnecting the aforementioned components, and a power supply 276 for powering the aforementioned components. In some embodiments, the input 280 is a touch-sensitive display, such as a touch-sensitive surface. In some embodiments, the user interface 278 includes one or more soft keyboard embodiments. The soft keyboard embodiments may include standard (QWERTY) and/or non-standard configurations of symbols on the displayed icons. The regimen adjustment device 250 illustrated in FIG. 3 optionally includes, in addition to accelerometer(s) 317, a magnetometer (not shown) and a GPS 319 (or GLONASS or other global navigation system) receiver for obtaining information concerning the location and orientation (e.g., portrait or landscape) of the regimen adjustment device 250 and/or for determining an amount of physical exertion by the subject. It should be appreciated that the regimen adjustment device 250 illustrated in FIG. 3 is only one example of a multifunction device that may be used for optimizing basal administration timing in a standing basal insulin regimen for a subject, and that the regimen adjustment device 250 optionally has more or fewer components than shown, optionally combines two or more components, or optionally has a different configuration or arrangement of the components. The various components shown in FIG. 3 are implemented in hardware, software, firmware, or a combination thereof, including one or more signal processing and/or application specific integrated circuits. Memory 192 of the regimen adjustment device 250 illustrated in FIG. 3 optionally includes high-speed random access memory and optionally also includes non-volatile memory, such as one or more magnetic disk storage devices, flash memory devices, or other non-volatile solid-state memory devices. Access to memory 192 by other components of the regimen adjustment device 250, such as CPU(s) 274 is, optionally, controlled by the memory controller 368. In some embodiments, the memory 192 of the regimen adjustment device 250 illustrated in FIG. 3 optionally includes a third data set 302 comprising a plurality of insulin medicament records over the past time course. Each insulin medicament record 304 in the plurality of medicament records comprises: (i) a respective insulin medicament injection event 306 including an amount of insulin medicament injected 308 into the subject using a respective insulin pen 104 in the one or more insulin pens, (ii) a corresponding insulin event electronic timestamp 310 that is automatically generated by the respective insulin pen 104 upon occurrence of the respective insulin medicament injection event, and (iii) a respective type of insulin medicament 312 injected into the subject from one of (a) the basal insulin medicament and (b) the bolus insulin medicament. In some embodiments, the memory 192 of the regimen adjustment device 250 illustrated in FIG. 3 optionally includes a meal record data set 314 comprising a plurality of meal records over the past time course. Each respective meal record 316 in the plurality of meal records comprises: (i) a carbohydrate intake event 318 (e.g., a meal such a breakfast, lunch, or dinner), and (ii) a corresponding carbohydrate timestamp 320 indicating when the respective carbohydrate intake event occurred. In some embodiments, memory 192 of the regimen adjustment device 250 illustrated in FIG. 3 optionally includes a fourth data set 322 comprising the physical exertion of the subject over the past time course. The peripherals interface 370 can be used to couple input and output peripherals of the device to CPU(s) 274 and memory 192. The one or more processors 274 run or execute various software programs and/or sets of instructions stored in memory 192, such as the basal timing adjustment module 204, to perform various functions for the regimen adjustment device 250 and to process data. In some embodiments, the peripherals interface 370, CPU(s) 274, and memory controller 368 are, optionally, implemented on a single chip. In some other embodiments, they are implemented on separate chips. RF (radio frequency) circuitry of network interface 284 receives and sends RF signals, also called electromagnetic signals. In some embodiments, the standing insulin regimen 206, the first data set 218, the fasting event data set 224, the second data set 228, the third data set 302, the meal record data set 314, and/or the fourth data set 310 is received using this RF circuitry from one or more devices such as a glucose sensor 102 associated with a subject, an insulin pen 104 associated with the subject and/or the data collection device 200. In some embodiments, the RF circuitry 108 converts electrical signals to/from electromagnetic signals and communicates with communications networks and other communications devices, glucose sensors 102, and insulin pens 104 and/or the data collection device 200 via the electromagnetic signals. The RF circuitry 284 optionally includes well-known circuitry for performing these functions, including but not limited to an antenna system, an RF transceiver, one or more amplifiers, a tuner, one or more oscillators, a digital signal processor, a CODEC chipset, a subscriber identity module (SIM) card, memory, and so forth. The RF circuitry 284 optionally communicates with the communication network 106. In some embodiments, the circuitry 284 does not include RF circuitry and, in fact, is connected to the network 106 through one or more hard wires (e.g., an optical cable, a coaxial cable, or the like). In some embodiments, the audio circuitry 372, the optional speaker 360, and the optional microphone 362 provide an audio interface between the subject and the adjustment timing device 250. The audio circuitry 372 receives audio data from the peripherals interface 370, converts the audio data to electrical signals, and transmits the electrical signals to the speaker 360. The speaker 360 converts the electrical signals to human-audible sound waves. The audio circuitry 372 also receives electrical signals converted by the microphone 362 from sound waves. The audio circuitry 372 converts the electrical signal to audio data and transmits the audio data to peripherals interface 370 for processing. Audio data is, optionally, retrieved from and/or transmitted to the memory 192 and/or the RF circuitry 284 by the peripherals interface 370. In some embodiments, the power supply 276 optionally includes a power management system, one or more power sources (e.g., battery, alternating current (AC)), a recharging system, a power failure detection circuit, a power converter or inverter, a power status indicator (e.g., a light-emitting diode (LED)) and any other components associated with the generation, management and distribution of power in portable devices. In some embodiments, the regimen adjustment device 250 optionally also includes one or more optical sensors 373. The optical sensor(s) 373 optionally include charge-coupled device (CCD) or complementary metal-oxide semiconductor (CMOS) phototransistors. The optical sensor(s) 373 receive light from the environment, projected through one or more lens, and converts the light to data representing an image. The optical sensor(s) 373 optionally capture still images and/or video. In some embodiments, an optical sensor is located on the back of the regimen adjustment device 250, opposite the display 282 on the front of the regimen adjustment device 250, so that the input 280 is enabled for use as a viewfinder for still and/or video image acquisition. In some embodiments, another optical sensor 373 is located on the front of the regimen adjustment device 250 so that the subject's image is obtained (e.g., to verify the health or condition of the subject, to determine the physical activity level of the subject, to help diagnose a subject's condition remotely, or to acquire visual physiological measurements 312 of the subject, etc.). As illustrated in FIG. 3, a regimen adjustment device 250 preferably comprises an operating system 202 that includes procedures for handling various basic system services. The operating system 202 (e.g., iOS, DARWIN, RTXC, LINUX, UNIX, OS X, WINDOWS, or an embedded operating system such as VxWorks) includes various software components and/or drivers for controlling and managing general system tasks (e.g., memory management, storage device control, power management, etc.) and facilitates communication between various hardware and software components. In some embodiments the regimen adjustment device 250 is a smart phone. In other embodiments, the regimen adjustment device 250 is not a smart phone but rather is a tablet computer, desktop computer, emergency vehicle computer, or other form or wired or wireless networked device. In some embodiments, the regimen adjustment device 250 has any or all of the circuitry, hardware components, and software components found in the regimen adjustment device 250 depicted in FIG. 2 or 3. In the interest of brevity and clarity, only a few of the possible components of the regimen adjustment device 250 are shown in order to better emphasize the additional software modules that are installed on the adjustment timing device 250. While the system 48 disclosed in FIG. 1 can work standalone, in some embodiments it can also be linked with electronic medical records to exchange information in any way. Now that details of a system 48 for optimizing basal administration timing in a standing basal insulin regimen for a subject have been disclosed, details regarding a flow chart of processes and features of the system, in accordance with an embodiment of the present disclosure, are disclosed with reference to FIGS. 4A through 4E. In some embodiments, such processes and features of the system are carried out by the basal timing adjustment module 204 illustrated in FIGS. 2 and 3. Block 402. With reference to block 402 of FIG. 4A, the goal of insulin therapy in subjects with either type 1 diabetes mellitus or type 2 diabetes mellitus is to match as closely as possible normal physiologic insulin secretion to control fasting and postprandial plasma glucose. As illustrated in FIG. 2, a regimen adjustment device 250 comprises one or more processors 274 and a memory 192/290. The memory stores instructions that, when executed by the one or more processors, perform a method for optimizing basal administration timing in a standing basal insulin regimen. Blocks 404-406. Referring to block 404 of FIG. 4A, in the method, a standing basal insulin regimen 206 for the subject is obtained. The standing basal insulin regimen 206 for the subject comprises a total amount of basal insulin medicament 210 for a recurring period 208. An example of a recurring period 208 is one day and in such an example, the total amount of basal insulin medicament 210 is the amount of basal insulin medicament 210 the subject should take each day. The present disclosure is not limited to recurring periods that are one day and longer or shorter recurring periods are within the scope of the present disclosure. The standing basal insulin regimen 206 further contains one or more basal injection event types in a set of basal injection event types for the recurring period. Referring to block 406 of FIG. 4A, examples of basal injection event types include “morning basal” which represents the portion of the total amount of basal insulin medicament 210 that is to be injected as a single injection event using an insulin pen 104 in the morning and “evening basal” which represents the portion of the total amount of basal insulin medicament 210 that is to be injected as a single injection event using an insulin pen 104 in the evening. The standing basal insulin regimen 206 further contains a respective apportionment 214 of the total amount of basal insulin medicament between each respective basal injection event type 212 in the one or more basal injection event types. For instance, in an example where the standing basal insulin regimen 206 consists of two the “morning basal” and “evening basal” injection event types, the respective apportionment 214 will indicate what percentage of the total amount of basal insulin medicament 210 is to be administered as the “morning basal” (e.g., forty percent, fifty percent, sixty percent) and what percentage of the total amount of basal insulin medicament 210 is to be administered as the “evening basal” (e.g., forty percent, fifty percent, sixty percent), where the respective apportionments collectively sum up to the entire total amount of the basal insulin medicament 210 for the recurring period. Thus, in the example where the standing basal insulin regimen 206 consists of two the “morning basal” and “evening basal” injection event types, the respective apportionment of the “morning basal” and “evening basal” sums up to 100 percent of the basal insulin medicament 210 for the recurring period. In some embodiments, the basal insulin medicament specified by the basal insulin medicament dosage regimen 206 consists of a single insulin medicament having a duration of action that is between 12 and 24 hours or a mixture of insulin medicaments that collectively have a duration of action that is between 12 and 24 hours. Examples of such basal insulin medicaments include, but are not limited to, Insulin Degludec (developed by NOVO NORDISK under the brand name Tresiba), NPH (Schmid, 2007, “New options in insulin therapy,” J Pediatria (Rio J). 83(Suppl 5): S146-S155), Glargine (LANTUS, Mar. 2, 2007), Insulin Glargine [rDNA origin] injection (Dunn et al. 2003, “An Updated Review of its Use in the Management of Diabetes Mellitus” Drugs 63: p. 1743), and Determir (Plank et al., 2005, “A double-blind, randomized, dose-response study investigating the pharmacodynamic and pharmacokinetic properties of the long-acting insulin analog detemir,” Diabetes Care 28:1107-1112). Blocks 408-410. Referring to block 408 of FIG. 4A, in the method, a first data set 216 is obtained. The past time course comprises a first plurality of instances of a recurring period. In some embodiments, the recurring period is one day and the past time course comprises a plurality of days (e.g., two days, three days, four days, or five or more days). The first data set 216 comprises a plurality of glucose measurements of the subject taken over the past time course. In typical embodiments, the glucose measurements are from one or more glucose sensors 102. FIG. 2 illustrates. Each such glucose measurement 218 is timestamped with a glucose measurement timestamp 220 to represent when the respective measurement was made. Thus, in some embodiments, the glucose measurements are measured without human intervention. That is, the subject does not manually make the glucose measurements. In alternative embodiments of the present disclosure, the subject or a health care practitioner manually takes glucose measurements and such manual glucose measurements are used as the glucose measurements 218 in the first data set 216. In embodiments where autonomous glucose measurements are used in the first data set 216, devices such as the FREESTYLE LIBRE CGM by ABBOTT (“LIBRE”) may serve as the glucose sensor 102 in order to make the plurality of autonomous glucose measurements of a subject. The LIBRE allows calibration-free glucose measurements with an on-skin coin-sized sensor, which can send up to eight hours of data to a reader device (e.g., the data collection device 200 and/or the regimen adjustment device 250) via near field communications, when brought close together. The LIBRE can be worn for fourteen days in all daily life activities. Referring to block 410 of FIG. 4A, in some embodiments, the glucose measurements 218 are autonomously taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less. In some embodiments, the glucose measurements 218 are taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less, over a time period of a day or more, two days or more, a week or more, or two weeks or more. In some embodiments, the glucose measurements 218 are autonomously taken (e.g., without human effort, without human intervention, etc.). In some embodiments the regimen adjustment device 250 further comprises a wireless receiver and the first data set 216 is obtained wirelessly from a glucose sensor 102 affixed to the subject (e.g., in accordance with an 802.11, Bluetooth, or ZigBee standard). Referring to block 412 of FIG. 4A, in some embodiments, the past time course is the last week, the last two weeks, or the last month. Further, the method disclosed in FIGS. 4A through 4E is repeated on a recurring basis over time. Further still, the basal insulin medicament consists of a single insulin medicament having a duration of action that is between 12 and 24 hours or a mixture of insulin medicaments that collectively have a duration of action that is between 12 and 24 hours. Blocks 414-416. Referring to block 414 of FIG. 4B, the method continues with the evaluation of the plurality of glucose measurements of the first data set 216 over the past time course using a stop condition 222. The purpose of this evaluation is to ensure that the relative apportionment of the total amount of basal insulin medicament between the injection event types is optimal. FIG. 7 illustrates. In the example illustrated in FIG. 7 the glucose measurements of the first data set 218, as well as optionally insulin pen and meal data are used to identify parameters of a model describing insulin-glucose dynamics of the individual subject. The model with identified parameters allows for the simulation of different basal insulin medicament injection event apportionment scenarios, where different timings of basal injections are simulated and the results compared with respect to glycaemic outcome. This practice constitutes evaluation of the plurality of glucose measurements of the first data set 216 over the past time course using a stop condition 222. In some embodiments, included in this evaluation are basal split scenarios (e.g., where the basal injection event types used in the modeling is expanded to include more injection event types) and basal un-splitting scenarios (e.g., where the basal injection event types used in the modeling is compacted to include fewer injection event types). Further still, the apportionment of the total amount of basal insulin medicament across the different basal injection event types can also be determined using optimized using the modeling. In FIG. 7, four different apportionment scenarios are evaluated using a stop condition (1: 100 percent evening basal injection event; 2: 100 percent morning basal injection event; 3: 50 percent evening and 50 percent morning basal injection event; and 4: 30 percent evening and 70 morning basal injection event). Each of these apportionment scenarios is evaluated over a future time course for a stop condition. For instance, each of the apportionment scenarios are evaluated to determine if they better project a subject from glycaemic risks than the apportionment 214 found in the current standing insulin regimen 206. When this is the case, the stop condition is deemed satisfied and the apportionment of the total amount of basal insulin medicament 210 is reapportioned across the basal injection event types 212 in the standing insulin regimen 206 in accordance with the apportionment scenario that better protects the subject from glycaemic risks. Referring to block 416 of FIG. 4B, in some embodiments, the first data set is filtered such that glucose measurements 218 from only those periods of time in which the subject is adhering to the standing insulin regimen are used. In such embodiments, glucose measurements taken during periods in which the subject is not in adherence with the standing insulin regimen 206 are not used. For instance, in some embodiments, a subject is deemed to not be adhering to the standing insulin regimen when the subject is taking the total amount of basal insulin medicament 210 but not according to the apportionment 214 specified by the standing insulin regimen 206. For example, if the standing insulin regimen 206 specifies a “morning basal” injection event type in which fifty percent of the total amount of basal insulin medicament 210 is to be injected with an insulin pen and an “evening basal” injection event type in which the other fifty percent of the total amount of basal insulin medicament 210 is to be injected, but the subject takes 80 percent of total amount of basal insulin medicament 210 in the morning on day, the subject is deemed to not be in adherence with the standing insulin regimen 206 for the day and all glucose measurements 218 taken that day are not used. Thus, in accordance with block 416 of FIG. 4B, to determine standing regimen adherence, a third data set 302 is obtained from an insulin pen 104 used by the subject to apply the basal insulin regimen. The third data set comprises insulin medicament records. Each respective record 304 in the plurality of medicament records comprises: (i) an insulin medicament injection event 306 including an amount of basal insulin medicament injected 308 into the subject and (ii) a corresponding insulin event electronic timestamp 310 that is automatically generated by the insulin pen upon occurrence of the respective insulin medicament injection event. The third data set and the standing basal insulin regimen is then used to determine one or more recurring periods in the past time course that do not comply with the standing basal insulin regimen for the subject. Glucose measurements taken during these days are excluded from the stop condition evaluation. Blocks 418 through 436. Blocks 418 through 436 of FIGS. 4B through 4E provide several different embodiments in accordance with the present disclosure for how the disclosed method proceeds to both evaluate the plurality of glucose measurements over the past time course using the stop condition 222 and what actions are taken when the stop condition is deemed to be satisfied. In particular, referring to block 418, when the stop condition is satisfied, a recommended adjustment is made that comprises a change in the number of basal injection event types in the standing basal insulin regimen and/or a change in the respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types. An example of a recommended adjustment has been presented above in relation to FIG. 7. In FIG. 7, four different apportionment scenarios are evaluated using a stop condition (1: 100 percent evening basal injection event; 2: 100 percent morning basal injection event; 3: 50 percent evening and 50 percent morning basal injection event; and 4: 30 percent evening and 70 morning basal injection event). Each of these apportionment scenarios is evaluated over a future time course for a stop condition. For instance, each of the apportionment scenarios are evaluated to determine if they better project a subject from glycaemic risks than the apportionment 214 found in the current standing insulin regimen 206. When this is the case, the stop condition is deemed satisfied and the apportionment of the total amount of basal insulin medicament 210 is reapportioned across the basal injection event types 212 in the standing insulin regimen 206 in accordance with the apportionment scenario that better protects the subject from glycaemic risks. Block 420 provides another such example of the determination of a recommended adjustment to the standing insulin regimen 206. In this embodiment, the standing basal insulin regimen 206 specifies a single basal injection event type 212 for the recurring period 208. In one example, one hundred percent of the total amount of basal insulin medicament 210 is to be administered with an insulin pen as a single “morning basal” injection event. In this embodiment, the evaluating the glucose measurements over the past time course using the stop condition 222 comprises obtaining fasting events in the past time course. Each fasting event 226 is associated with a different instance of the recurring period in the plurality of instances of the recurring period. For example, in the case where the recurring period is a day, each fasting event 226 is associated with a different day in the multi-day past time course. That is, if the recurring period is one day, an example of a “different instance” of the recurring period would be a particular day, such as Tuesday, May 5. For each respective fasting event 226 in the one or more fasting events, there is compared (i) one or more first glucose measurements (e.g., a single glucose measurement, a measure of central tendency of 2 or more glucose measurements) of the subject in the first data set occurring at a first time slot that is a first predetermined amount of time prior to (e.g., 2 hours before, 4 hours before, 6 hours before, 8 hours before, 10 hours before, 12 hours before, etc.) a beginning of the respective fasting event to (ii) one or more second glucose measurements (e.g., a single glucose measurement, a measure of central tendency of 2 or more glucose measurements) of the subject in the first data set occurring at a second time slot that is at a predetermined point within or after (e.g. 30 minutes after, one hour after, two hours after, three hours after, etc.) the respective fasting event, thereby obtaining one or more comparisons. Such comparisons are premised on the basis that if a subject takes one insulin dose per day and glucose measurements are high before bedtime but low before breakfast or vice versa, this indicates that the basal should be split. FIG. 6 illustrates. Panel A of FIG. 6 illustrates the blood glucose concentration of a subject over a recurring period in which the contribution to glucose levels from meal events and associated short term insulin medicament (bolus) injection events have been subtracted out. Further in panel A, the subject takes 100 percent of the total amount of basal insulin medicament 210 as a morning basal bolus injection event 212. Comparison of the glucose measurements in the first time slot to the glucose measurements in the second time slot (which are obtained from the first data set 216 using the glucose measurement timestamps 220) shows a substantial deviation and indicates that there is insufficient insulin medicament during the first time slot. Panel B of FIG. 6 illustrates the blood glucose concentration of a different subject over a recurring period in which the contribution to glucose levels from meal events and associated bolus injection events has likewise been subtracted. Further in panel B, the subject takes 100 percent of the total amount of basal insulin medicament 210 as an evening basal bolus. Comparison of the glucose measurements in the first time slot to the glucose measurements in the second time slot for the subject of panel B shows a substantial deviation and indicates that there is insufficient insulin medicament during the second time slot. Thus, in both the situation of panel A and panel B of FIG. 6, the basal insulin medicament should be split into two injections events per recurring period. Thus, as illustrated in FIG. 6, the stop condition in the embodiment of block 420 is satisfied when the comparisons indicate that the respective one or more first glucose measurements (of the first time slot) deviate from the corresponding respective one or more second glucose measurements (of the second time slot) by more than a threshold amount (e.g., by five percent or more, 10 percent or more, 15 percent or more, 20 percent or more, 25 percent or more, 30 percent or more, 35 percent or more, 40 percent or more, 45 percent or more, 50 percent or more, 55 percent or more, 60 percent or more, or 65 percent or more). In such embodiments, as discussed above for panels A and B of FIG. 6, the recommended adjustment is to increase the number of basal injection event types 212 to two (e.g., a “morning basal” and an “evening basal”) and apportion the total amount of basal insulin medicament 210 between the two basal injection event types. Embodiments such as that described in block 420 rely on the identification of fasting events during the past time course. Block 422 of FIG. 4C provides one way in accordance with the present disclosure in which such fasting events are identified. In accordance with block 422, a first fasting event 226 is identified in a first recurring period (e.g., a period of 24 hours) in the first plurality of recurring periods in the past time course represented by the first data set encompassed by the plurality of glucose measurements in the first data set 216 by first computing a moving period of variance σk2 across the plurality of glucose measurements, where: σ k 2 = ( 1 M ⁢ ∑ i = k - M + 1 k ⁢ ( G i - G ¯ ) ) 2 and where Gi is the ith glucose measurement in a portion k of the plurality of glucose measurements, M is a number of glucose measurements in the plurality of glucose measurements and represents the past time course, G is the mean of the glucose measurements selected from the plurality of glucose measurements of the first data set 216, and k is within the first recurring time period. As an example, the glucose measurements may span several days or weeks, with glucose measurements taken every five minutes. A first time period k (e.g., one day) within this overall time span is selected and thus the portion k of the plurality of measurements is examined for a period of minimum variance. The first fasting period is deemed to be the period of minimum variance min k ⁢ σ k 2 within the first time period. Next, the process is repeated with portion k of the plurality of glucose measurements by examining the next portion k of the plurality of glucose measurements for another period of minimum variance thereby assigning another fasting period. Moreover, in some embodiments, only those fasting events that are deemed standing insulin regimen 206 adherent are used. Example 1, below, illustrates a way in which a determination is made as to whether a fasting event 226 is standing insulin regimen 206 adherent. Moreover, European Patent Application Number EP16177080.5, entitled “Regimen Adherence Measure for Insulin Treatment Base on Glucose Measurement and Insulin Pen Data,” filed Jun. 30, 2016, which is hereby incorporated by reference, discloses techniques for identifying and classifying fasting events as adherent or nonadherent. In some embodiments, only those fasting events that are classified as “basal regimen adherent” in accordance with European Patent Application Number EP16177080.5 are used to optimizing basal administration timing in a standing basal insulin regimen. Block 424 of FIG. 4C provides another way in accordance with the present disclosure in which fasting events 224 are identified. In accordance with block 424, the obtaining of the one or more fasting events comprises receiving an indication of each fasting event in the one or more fasting events from the subject. In other words, in some embodiments, the identifying the one or more of fasting events comprises receiving an indication of each fasting event 226 in the one or more fasting events from the subject in the form of feed-forward events. Each respective feed-forward event represents an instance where the subject has indicated they are fasting or are about to fast. For instance, the user may indicate through a graphical user interface provided by the basal timing adjustment module 204 when the subject is fasting (e.g. has not eaten a meal in more than five hours, more than six hours, more than seven hours, more than eight hours, more than nine hours, or more than 10 hours, etc.). Block 426 of FIG. 4C provides yet another way in accordance with the present disclosure in which fasting events 224 are identified. In accordance with block 426, the obtaining of the one or more fasting events comprises receiving a second data set 228 from a wearable device (e.g., from wearable physiological measurement devices, from measurement devices within the data collection device 200 such as a magnetometer or a thermostat, etc.) worn by the subject, and the second data set indicates a physiological metric 230 of the subject during the past time course that is indicative of the one or more fasting events. In some embodiments, the physiological metric measurement 230 is body temperature of the subject. In some embodiments, the physiological metric measurement 230 is a measurement of activity of the subject. In some embodiments, the optional accelerometer 317, optional GPS 319, and/or magnetometer (not shown) of the regimen adjustment device 250 or such components optionally within the one or more glucose monitors 102 and/or the one or more insulin pens 104 is used to acquire such physiological metric measurements 232. In some embodiments, both an autonomous fast detection algorithm, such as one disclosed in blocks 422 and/or 426, and the manual (user indicated) fast detection disclosed in block 424 are used for detecting fasting events. For instance, in some embodiments, a fasting event 226 that has been autonomously detected (e.g., using the algorithm of block 422) is then verified using the feed-forward events of block 424 and/or 426. To illustrate, when a fasting event 226 autonomously detected using an algorithm such as one disclosed in block 422 is matched in time (temporally matched) to a feed-forward event in which the subject indicated they are fasting and/or a physiological metric 230 that indicated they are fasting, the fasting event 226 is deemed verified and used in further steps of the present disclosure. In some embodiments, a fasting event 226 must be verified in this manner and also be deemed insulin regimen 206 adherent (e.g., deemed standing insulin regimen 206 adherent as disclosed in Example 1 below). Referring to block 428 of FIG. 4C, in some embodiments, the set of possible basal injection event types 212 for the recurring period specified by the standing insulin regimen 206 consists of “morning basal,” and “night basal.” In some such embodiments, the standing insulin regimen 206 specifies a single basal injection event type for the recurring period, the “morning basal” injection event type, and the recommended adjustment of block 418 is to add the “night basal” basal injection event type to the standing insulin regimen (e.g., in response to the scenario of FIG. 6, panel A) and to apportion the total amount of basal insulin medicament 210 between the “night basal” basal injection event type and the “morning basal” basal injection event type. Referring to block 430 of FIG. 4D, in some embodiments the method further comprises obtaining a data set 302 from an insulin pen used by the subject to apply the standing basal insulin regimen. The data set 302 comprises a plurality of insulin medicament records over the past time course. Each insulin medicament record 304 in the plurality of medicament records comprises: (i) a respective insulin medicament injection event 306 representing an insulin medicament injection of the basal insulin medicament into the subject using the insulin pen and (ii) a corresponding insulin event electronic timestamp 310 that is automatically generated by the insulin pen upon occurrence of the respective insulin medicament injection event. The first data set 216 and the data set 302 are used to simulate a glucose concentration of the subject over a future time course a plurality of times, thereby computing a plurality of simulations of the glucose concentration of the subject over the future time course. FIG. 7 illustrates such simulations. As illustrated in FIG. 7, each respective glucose concentration simulation 234 in the plurality of simulations is associated with a different apportionment 236 of the total amount of basal insulin medicament across the set of basal injection event types. For instance: 100% taken in the morning 100% taken at night 80%/20% morning/night 70%/30% morning/night 60%/40% morning/night 50%/50% morning/night 40%/60% morning/night 30%/70% morning/night 20%/80% morning/night In such embodiments, the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises evaluating the glucose concentration of the subject across the future time course 238 in each respective simulation in the plurality of simulations by calculating a glycaemic risk metric 240 (e.g., blood glucose variance, time in a desired glucose target range, estimated HbA1c) for each simulation. That is, the glucose measurements 218 of the first data set, and optionally the insulin pen data of the third data set 302, and optionally the meal data of the meal record data set (FIG. 3) are used to identify parameters of a model (e.g., insulin sensitivity factor) describing insulin-glucose dynamics of the individual subject. The model with identified parameters allows for the simulation of apportionment scenarios of the total amount of basal insulin medicament 210 across the set of basal injection event types, where different timings of basal injections are simulated and the results compared with respect to glycaemic outcome. The stop condition is satisfied when a first simulation in the plurality of simulations minimizes the glycaemic risk metric, by more than a threshold amount as compared to one of (i) a reference simulation 242 of the glucose concentration of the subject over the future time course based upon the apportionment of the total amount of basal insulin medicament across the set of basal injection event types specified in the standing basal insulin regimen 206 and (ii) the glucose concentration of the subject across the first data set. This threshold amount is application specific, dependent upon the parameters of the model describing insulin-glucose dynamics of the individual subject, as well as the glycaemic risk metric. Thus, consider the case in which the standing insulin regimen 206 specifies that 100% apportionment of the total amount of basal insulin medicament 210 is to be taken as a single basal injection event 212 in the morning in each recurring period. A reference simulation 242 of the glucose concentration of the subject over the future time course using the insulin-glucose dynamics of the individual subject may be taken with this 100% apportionment and the glycaemic risk metric for this reference simulation evaluated. Also a simulation 242 of the glucose concentration of the subject over the future time course using the same insulin-glucose dynamics of the individual subject but with 50%/50% morning/night apportionment of the total amount of basal insulin medicament 210 each recurring period and the glycaemic risk metric for this simulation evaluated. If the 50%/50% morning/night apportionment produces a better value for the glycaemic risk metric than the 100% apportionment of the total amount of basal insulin medicament 210 to morning, than the stop condition is satisfied and the recommended adjustment comprises changing the apportionment of the total amount of basal insulin medicament 210 from 100% morning apportionment per recurring period to a 50%/50% morning/night apportionment per recurring period. Alternatively, again consider the case in which the standing insulin regimen 206 specifies that 100% apportionment of the total amount of basal insulin medicament 210 is to be taken as a single basal injection event 212 in the morning in each recurring period. A reference simulation 242 of the glucose concentration of the subject over the future time course using the insulin-glucose dynamics of the individual subject is not taken with this 100% apportionment. Rather the glucose measurements from the past time course during which the 100% apportionment was imposed in the standing insulin regimen 206 are used to compute the glycaemic risk metric. Also a simulation 242 of the glucose concentration of the subject over the future time course using the insulin-glucose dynamics derived for the individual subject from the data acquired during the past time course (e.g., the glucose measurements 218, etc.) but with 50%/50% morning/night apportionment of the total amount of basal insulin medicament 210 each recurring period and the glycaemic risk metric for this simulation is evaluated. If the 50%/50% morning/night apportionment produces a better value for the glycaemic risk metric than the glycaemic risk metric computed using the glucose measurements from the first data set 216, than the stop condition is satisfied and the recommended adjustment comprises changing the apportionment of the total amount of basal insulin medicament 210 from 100% morning apportionment per recurring period to a 50%/50% morning/night apportionment per recurring period. Referring to block 432 of FIG. 4E, and using FIG. 7 to illustrate examples of first and second threshold values, in some embodiments the glycaemic risk metric comprises (i) a total glucose level variability observed across the respective simulation, (ii) a variability in a plurality of fasting glucose levels calculated across the respective simulation, (iii) a percentage of time that a total glucose level exceeds a first threshold value 702 or falls below a second threshold 704 value across the respective simulation, or (iv) a percentage of time that an HbA1c level exceeds a third threshold value or falls below a fourth threshold value across the respective simulation. In instances where a glycaemic risk metric computed for a simulation is compared to a glycaemic risk metric computed using the glucose measurements in the first data set, the glycaemic risk metric for the first data set is computed using the glucose measurements levels within the first data set. Referring to block 434 of FIG. 4E, and as discussed above, in some embodiments, the method further comprises obtaining a meal record data set 314 comprising a plurality of meal records over the past time course for the subject. Each respective meal record 316 in the meal record data set comprises: (i) a carbohydrate intake event 318 and (ii) a corresponding electronic carbohydrate timestamp 320 of when the carbohydrate intake event occurred. In such embodiments, the using the first data set and the second data set are used to simulate a glucose concentration of the subject over a future time course a plurality of times comprises using the first data set, the second data set, and the meal record data set to simulate a glucose concentration of the subject over the future time course the plurality of times. That is, the glucose measurements of the first data set 218, as well as insulin pen data and meal data are used to identify parameters of a model describing insulin-glucose dynamics of the individual subject. The model with identified parameters is then used to simulated different basal insulin medicament injection event apportionment scenarios, where the different timings of basal injections are simulated and the glycaemic risk metric is calculated for each simulation. Referring to block 436 of FIG. 4E, in some embodiments, a fourth data set 322 comprising physical exertion of the subject over the past time course is obtained. In such embodiments, the using the first data set 216 and the data set 302 to simulate a glucose concentration of the subject over a future time course a plurality of times comprises using the first data set 216, the data set 302, the meal record data set 314 and the fourth data set 322 to simulate a glucose concentration of the subject over the future time course the plurality of times. That is, the glucose measurements of the first data set 218, as well as insulin pen data, meal data and physical exertion data are used to identify parameters of a model describing insulin-glucose dynamics of the individual subject. The model with identified parameters is then used to simulated different basal insulin medicament injection event apportionment scenarios, where the different timings of basal injections are simulated and the glycaemic risk metric is calculated for each simulation. Block 438. Referring to block 438 of FIG. 4E, the method continues by communicating the recommended adjustment to the standing basal insulin regimen, when the determination is made to make the recommended adjustment to the standing basal insulin regimen for the subject, to: (i) the subject for manual adjustment of the standing basal insulin regimen, (ii) each insulin pen 104 in one or more insulin pens charged with delivering the standing basal insulin regimen to the subject, as dosage adjustment instructions, or (iii) a health care practitioner associated with the subject. FIG. 8 illustrates an exemplary embodiment of the method disclosed in FIGS. 4A through 4E. Example 1: Use of glucose measurements to determine whether a fasting event is insulin regimen adherent. In some embodiments, the first data set 216 comprising a plurality of glucose measurements is obtained. In some embodiments the glucose measurements are obtained autonomously, for instance by a continuous glucose monitor 102. In this example, in addition to the autonomous glucose measurements, insulin administration events are obtained in the form of insulin medicament injection events 306 from one or more insulin pens 104 used by the subject to apply the standing insulin regimen 206. These insulin medicament records 304 may be in any format, and in fact may be spread across multiple files or data structures. As such, in some embodiments, the instant disclosure leverages the recent advances of insulin administration pens, which have become “smart” in the sense that they can remember the timing and the amount of insulin medicament administered in the past. One example of such an insulin pen 104 is the NovoPen 5. Such pens assists patients in logging doses and prevent double dosing. It is contemplated that insulin pens will be able to send and receive insulin medicament dose volume and timing, thus allowing the integration of continuous glucose monitors 102, insulin pens 104 and the algorithms of the present disclosure. As such, insulin medicament records 304 from one or more insulin pens 104 is contemplated, including the wireless acquisition of such data from the one or more insulin pens 104. In some embodiments, each insulin medicament record 304 comprises: (i) a respective insulin medicament injection event 306 including an amount of insulin medicament injected 308 into the subject using a respective insulin pen 104 in the one or more insulin pens and (ii) a corresponding insulin event electronic timestamp 310 that is automatically generated by the respective insulin pen 104 upon occurrence of the respective insulin medicament injection event 306. In some embodiments, a fasting event 226 is identified using the glucose measurements 218 of the subject and their associated glucose measurement timestamps 220 in the first data set 216. Once a fasting event is identified, e.g., by a method described in any one of blocks 422-426 above, or any other method, a classification is applied to the fasting event 224. The classification is one of “insulin regimen adherent” and “insulin regimen nonadherent.” A fasting event 226 is deemed insulin regimen adherent when the acquired one or more medicament records 304 establish, on a temporal and quantitative basis, adherence with the standing insulin medicament regimen 206 during the fasting event 226. A fasting event 226 is deemed insulin regimen nonadherent when the acquired one or more medicament records 304 do not include one or more medicament records that establish, on a temporal and quantitative basis, adherence with the standing insulin regimen 206 during the fasting event 226. In some embodiments, the standing insulin medicament regimen 206 specifies that a dosage of the basal insulin medicament is to be taken during each respective recurring period (e.g., day, twelve hour period) in a plurality of recurring periods and that a fasting event 226 is deemed insulin regimen nonadherent when there are no insulin medicament records 304 for the recurring period associated with the fasting event 226. In various embodiments, each recurring period in the plurality of recurring periods is two days or less, one day or less, or 12 hours or less. Thus, consider the case where the first data set 216 is used to identify a fasting event 226 and the standing insulin regimen 206 specifies to take dosage A of a basal insulin medicament every 24 hours. In this example, therefore, the recurring period is one day (24 hours). The fasting event 226 is inherently timestamped because it is derived from a period of minimum variance in timestamped glucose measurements, or by other forms of analysis of the timestamped glucose measurements 218 or by other means as disclosed in blocks 424 and 426 of FIG. 4C. Thus, the glucose measurement timestamp, or period of fasting (fasting event time period 228), represented by a respective fasting event 226 is used as a starting point for examining whether the fasting event is insulin regimen adherent. For instance, if the period of fasting associated with the respective timestamp includes 6:00 AM on Tuesday, May 17, what is sought in the insulin medicament records 304 is evidence that the subject took dosage A of the basal insulin medicament in the 24 hour period (the recurring period) leading up to 6:00 AM on Tuesday, May 17 (and not more or less of the prescribed dosage). If the subject took the prescribed dosage of the basal insulin medicament during this recurring period, and in accordance with the respective apportionments 214 across the basal injection event types 212 for the recurring period, the fasting event is deemed insulin regimen adherent. If the subject did not take the dose of the basal insulin medicament during this recurring period (or took more than the dose of the basal insulin medicament during this period specified by the standing insulin regimen 206, or did not adhere to the respective apportionments 214 across the basal injection event types 212 for the recurring period, the fasting event 224 is deemed to be insulin regimen nonadherent. LIST OF EMBODIMENTS 1. A device (250) for optimizing basal administration timing in a standing basal insulin regimen (206) for a subject, wherein the device comprises one or more processors (274) and a memory (192/290), the memory storing instructions that, when executed by the one or more processors, perform a method of: obtaining the standing basal insulin regimen for the subject, wherein the standing basal insulin regimen specifies (i) a total amount of basal insulin medicament (210) for a recurring period (208), (ii) one or more basal injection event types in a set of basal injection event types for the recurring period, and (iii) a respective apportionment (214) of the total amount of basal insulin medicament between each respective basal injection event type (212) in the one or more basal injection event types; obtaining a first data set (216), the first data set comprising a plurality of glucose measurements of the subject over a past time course, the past time course comprising a first plurality of instances of the recurring period and, for each respective glucose measurement (218) in the plurality of glucose measurements, a glucose measurement timestamp (220) representing when the respective measurement was made; evaluating the plurality of glucose measurements over the past time course using a stop condition (222), wherein, when the stop condition is satisfied, the method further comprises: determining a recommended adjustment comprising a change in the number of basal injection event types in the standing basal insulin regimen and/or a change in the respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types; and communicating the recommended adjustment to: (i) the subject for manual adjustment of the standing basal insulin regimen, (ii) an insulin pen charged with delivering the standing basal insulin regimen to the subject, or (iii) a health care practitioner associated with the subject. 2. The device of embodiment 1, wherein the standing basal insulin regimen specifies a single basal injection event type for the recurring period, and the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises: obtaining one or more fasting events in the past time course, wherein each fasting event (226) is associated with a different instance of the recurring period in the first plurality of instances of the recurring period, comparing, for each respective fasting event in the one or more fasting events, (i) one or more first glucose measurements of the subject in the first data set occurring at a first time slot that is a first predetermined amount of time prior to a beginning of the respective fasting event to (ii) one or more second glucose measurements of the subject in the first data set occurring at a second time slot that is at a predetermined point within or after the respective fasting event, thereby obtaining one or more comparisons, wherein, the stop condition is satisfied when the one or more comparisons indicate that the respective one or more first glucose measurements deviate from the corresponding respective one or more second glucose measurements by more than a threshold amount, and the recommended adjustment is to increase the number of basal injection event types to two basal injection event types and to apportion the total amount of basal insulin medicament between the two basal injection event types. 3. The device of embodiment 1, wherein the standing basal insulin regimen specifies a single basal injection event type for the recurring period, and the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises: obtaining a plurality of fasting events in the past time course, wherein each fasting event (226) is associated with a different instance of the recurring period in the first plurality of instances of the recurring period, obtaining, for each respective fasting event in the plurality of fasting events, (i) a first glucose measurements of the subject in the first data set occurring at a first time slot that is a first predetermined amount of time prior to a beginning of the respective fasting, and (ii) a second glucose measurements of the subject in the first data set occurring at a second time slot that is at a predetermined point within or after the respective fasting event, obtaining a first measure of central tendency of the first glucose measurement of each fasting event in the plurality of fasting event, and a second measure of central tendency of the second glucose measurement of each fasting in the plurality of fasting events, and comparing the first measure of central tendency to the second measure of central tendency, and thereby obtaining a comparison, wherein, the stop condition is satisfied when the comparison indicate that the respective first measure of central tendency deviate from the second measure of central tendency by more than a threshold amount, and the recommended adjustment is to increase the number of basal injection event types to two basal injection event types and to apportion the total amount of basal insulin medicament between the two basal injection event types. 4. The device of embodiment 2 or 3, wherein the obtaining the one or more fasting events comprises identifying a first fasting event in a first recurring period in the first plurality of recurring periods by computing a moving period of variance σk2 across the plurality of glucose measurements, wherein: σ k 2 = ( 1 M ⁢ ∑ i = k - M + 1 k ⁢ ( G i - G ¯ ) ) 2 wherein, Gi is the ith glucose measurement in a portion k of the plurality of glucose measurements, M is a number of glucose measurements in the plurality of glucose measurements and represents the past time course, G is the mean of the glucose measurements selected from the plurality of glucose measurements, and k is within the first recurring period; and associating the first fasting event with a region of minimum variance min k ⁢ σ k 2 within the first recurring period. 5. The device of embodiment 2 or 3, wherein the obtaining the one or more fasting events comprises receiving an indication of each fasting event in the one or more fasting events from the subject. 6. The device of embodiment 2 or 3, wherein the obtaining the one or more fasting events comprises receiving a second data set (228) from a wearable device worn by the subject, and the second data set indicates a physiological metric (230) of the subject during the past time course that is indicative of the one or more fasting events. 7. The device of any one of embodiments 1-6, the method further comprising: obtaining a third data set (302) from an insulin pen (104) used by the subject to apply the standing basal insulin regimen, the third data set comprising a plurality of insulin medicament records, each respective insulin medicament record (304) in the plurality of medicament records comprising: (i) a respective insulin medicament injection event (306) including an amount of basal insulin medicament injected (308) into the subject and (ii) a corresponding insulin event electronic timestamp (310) that is automatically generated by the insulin pen upon occurrence of the respective insulin medicament injection event, and using the third data set and the standing basal insulin regimen to determine one or more recurring periods in the past time course that do not comply with the standing basal insulin regimen for the subject; and excluding from the stop condition evaluation those glucose measurements in the one or more recurring periods in the past time course that do not comply with the standing basal insulin regimen. 8. The device of embodiment 1, wherein the set of basal injection event types for the recurring period consists of “morning basal,” and “night basal,” the standing basal insulin regimen specifies a single basal injection event type for the recurring period of “morning basal,” and the recommended adjustment is to add the “night basal” basal injection event type to the standing basal insulin regimen and to apportion the total amount of basal insulin medicament between the “night basal” basal injection event type and the “morning basal” basal injection event type. 9. The device of embodiment 1, wherein the method further comprises: obtaining a second data set from an insulin pen used by the subject to apply the standing basal insulin regimen, the second data set comprising a plurality of insulin medicament records over the past time course, each insulin medicament record in the plurality of medicament records comprising: (i) a respective insulin medicament injection event representing an insulin medicament injection of the basal insulin medicament into the subject using the insulin pen and (ii) a corresponding electronic timestamp that is automatically generated by the insulin pen upon occurrence of the respective insulin medicament injection event; and using the first data set and the second data set to simulate a glucose concentration of the subject over a future time course a plurality of times, thereby computing a plurality of simulations of the glucose concentration of the subject over the future time course, wherein each respective simulation (234) in the plurality of simulations is associated with a different apportionment (236) of the total amount of basal insulin medicament across the set of basal injection event types, the evaluating the plurality of glucose measurements over the past time course using the stop condition comprises evaluating the glucose concentration of the subject across the future time course (238) in each respective simulation in the plurality of simulations by calculating a glycaemic risk metric (240) for each simulation, the stop condition is satisfied when a first simulation in the plurality of simulations minimizes the glycaemic risk metric, by more than a threshold amount as compared to one of (i) a reference simulation (242) of the glucose concentration of the subject over the future time course based upon the apportionment of the total amount of basal insulin medicament across the set of basal injection event types specified in the standing basal insulin regimen and (ii) the glucose concentration of the subject across the first data set, and the apportionment of the total amount of basal insulin medicament across the set of basal injection event types in the first simulation is different than that of the standing basal insulin regimen. 10. The device of embodiment 9, wherein the glycaemic risk metric comprises: (i) a total glucose level variability observed across the respective simulation, (ii) a variability in a plurality of fasting glucose levels calculated across the respective simulation, (iii) a percentage of time that a total glucose level exceeds a first threshold value or falls below a second threshold value across the respective simulation, or (iv) a percentage of time that an HbA1c level exceeds a third threshold value or falls below a fourth threshold value across the respective simulation. 11. The device of embodiment 9 or 10, the method further comprising: obtaining a meal record data set (314) comprising a plurality of meal records over the past time course for the subject, each respective meal record (316) in the meal record data set comprising: (i) a carbohydrate intake event (318) and (ii) a corresponding electronic carbohydrate timestamp (320) of when the carbohydrate intake event occurred; and wherein the using the first data set and the second data set to simulate a glucose concentration of the subject over a future time course a plurality of times comprises using the first data set, the second data set, and the meal record data set to simulate a glucose concentration of the subject over the future time course the plurality of times. 12. The device of embodiment 11, the method further comprising: obtaining a fourth data set (322) comprising physical exertion of the subject over the past time course and wherein the using the first data set and the second data set to simulate a glucose concentration of the subject over a future time course a plurality of times comprises using the first data set, the second data set, the third data set and the fourth data set to simulate a glucose concentration of the subject over the future time course the plurality of times. 13. The device of embodiment 1, wherein the set of basal injection event types for the recurring period consists of “morning basal” and “night basal,” and the recurring period is a day. 14. The device of any one of embodiments 1-13, wherein successive measurements in the plurality of glucose measurements in the first data set are autonomously taken from the subject at an interval rate of 5 minutes or less, 3 minutes or less, or 1 minute or less. 15. The device of embodiment 1, wherein the past time course is the last week, the last two weeks, or the last month and wherein the method is repeated on a recurring basis over time, and the basal insulin medicament consists of a single insulin medicament having a duration of action that is between 12 and 24 hours or a mixture of insulin medicaments that collectively have a duration of action that is between 12 and 24 hours. 16. A method for optimizing basal administration timing in a standing basal insulin regimen for a subject, the method comprising: obtaining the standing basal insulin regimen for the subject, wherein the standing basal insulin regimen specifies (i) a total amount of basal insulin medicament for a recurring period, (ii) one or more basal injection event types in a set of basal injection event types for the recurring period, and (iii) a respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more basal injection event types; obtaining a first data set, the first data set comprising a plurality of glucose measurements of the subject over a past time course, the past time course comprising a first plurality of instances of the recurring period and, for each respective glucose measurement in the plurality of glucose measurements, a timestamp representing when the respective measurement was made; evaluating the plurality of glucose measurements over the past time course using a stop condition, wherein, when the stop condition is satisfied, the method further comprises: determining a recommended adjustment comprising a change in the number of basal injection event types in the standing basal insulin regimen and/or a change in the respective apportionment of the total amount of basal insulin medicament between each respective basal injection event type in the one or more periodic injection event types; and communicating the recommended adjustment to: (i) the subject for manual adjustment of the basal insulin regimen, (ii) an insulin pen charged with delivering the standing basal insulin regimen to the subject, or (iii) a health care practitioner associated with the subject. 17. A computer program comprising instructions that, when executed by one or more processors, perform the method of embodiment 16. 18. A computer-readable data carrier having stored thereon the computer program according to embodiment 17. REFERENCES CITED AND ALTERNATIVE EMBODIMENTS All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. The present invention can be implemented as a computer program product that comprises a computer program mechanism embedded in a nontransitory computer readable storage medium. For instance, the computer program product could contain the program modules shown in any combination of FIGS. 1, 2, 3, 5 and/or described in FIG. 4. These program modules can be stored on a CD-ROM, DVD, magnetic disk storage product, USB key, or any other non-transitory computer readable data or program storage product. Many modifications and variations of this invention can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only. The embodiments were chosen and described in order to best explain the principles of the invention and its practical applications, to thereby enable others skilled in the art to best utilize the invention and various embodiments with various modifications as are suited to the particular use contemplated. The invention is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled. 16324940 novo nordisk a/s USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open Apr 5th, 2022 04:33PM Apr 5th, 2022 04:33PM Novo Nordisk Health Care Pharmaceuticals & Biotechnology
nyse:nvo Novo Nordisk Apr 5th, 2022 12:00AM Sep 30th, 2016 12:00AM https://www.uspto.gov?id=US11292825-20220405 Protein conjugates The invention relates to protein conjugates and in particular conjugates of more than two protein or polypeptides. The compounds include a trivalent linker moiety that enables efficient production of desired products. 11292825 1. A protein conjugate of the following structure wherein Linker is a chemical moiety, S is a sulfur atom and Protein1 is covalently linked to Protein2 and Protein3 via the Linker and sulfur atoms, and wherein Protein2 and Protein3 are Fc polypeptides and Linker is a trivalent linker comprising —NH—C(═O)—CH2—* linked to —S-Protein2 and —NH—C(═O)—CH2—* linked to —S-Protein3 and -* indicates the attachment site to —S—, wherein the Linker is 5-200 atoms in length. 2. The protein conjugate of claim 1, wherein the conjugate has the following structure wherein U represents a central unit, RR represents a reactive end radical and S1, S2 and S3 represent individual spacers, wherein the central unit comprises a nitrogen atom or a benzene ring. 3. The conjugate according to claim 1, wherein the sulfur atom's (—S—) are part of thioethers (—CH2—S—CH2—). 4. The conjugate according to claim 2, wherein U comprises a nitrogen atom. 5. The conjugate according to claim 1, wherein the conjugate has the structure: Protein1-RR-S1-U-[S2—NH—C(═O)—CH2—S-Fc]2 wherein RR represents a reactive end radical, U represents a central unit, S1 and S2represent individual spacers and Fc is an Fc polypeptide, wherein the central unit comprises a nitrogen atom or a benzene ring. 6. The conjugate according to claim 1, wherein Protein1 is a growth hormone. 7. The conjugate according to claim 1, wherein the conjugate has the structure: GH-RR-S1-U-[S2—NH—C(═O)—CH2—S-Fc]2 wherein GH represents a growth hormone molecule, RR represents a reactive end radical, U represents a central unit, S1 and S2represent individual spacers and Fc is an Fc polypeptide, wherein the central unit comprises a nitrogen atom or a benzene ring. 8. The conjugate according to claim 1, wherein the hinge region of each Fc polypeptide includes a Cys residue. 9. The conjugate according to claim 1, wherein the sulfur atoms (—S—) are derived from protein cysteines, selected from wild type Cys residues or from variant Cys residues. 10. The conjugate according to claim 2, wherein Protein1 and S1 are linked via —S—CH2—C(═O)—NH—. 11. The conjugate according to claim 2, wherein U is a nitrogen atom. 12. The conjugate according to claim 5, wherein U is a nitrogen atom. 13. The conjugate according to claim 7, wherein U is a nitrogen atom. 14. The protein conjugate according to claim 1, wherein the length of the Linker is 10-60 atoms. 15. The protein conjugate according to claim 1, wherein the linker is selected from the group consisting of (S)-4-(2-{2-[((S)-1-{Bis-[2-(2-chloro-acetylamino)-ethyl]-carbamoyl}-3-carboxy-propylcarbamoyl)-methoxy]-ethoxy}-ethylcarbamoyl)-2-(2-bromo-acetylamino)-butyric acid, (2R)-5-[2-[2-[2-[[(1S)-1-[bis[2-[2-[(2-chloroacetyl)amino]ethylamino]-2-oxo-ethyl]carbamoyl]-3-carboxy-propyl]amino]-2-oxo-ethoxy]ethoxy]ethylamino]-2-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-oxo-pentanoic acid, (4S,18S)-4-(bis(2-(2-Chloroacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-bromoacetamido)ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid, 2-(2-bromoacetamido)-N,N-bis(2-(2-chloroacetamido)ethyl)acetamide, 2-(2-(2-(2-(2-bromoacetamido)ethoxy)ethoxy)acetamido)-N,N-bis(2-(2-chloroacetamido)ethyl)acetamide, (13R,18S)-18-(bis(2-(2-Chloroacetamido)ethyl)carbamoyl)-1-bromo-13-carboxy-2,11,16-trioxo-6,9-dioxa-3,12,17-triazahenicosan-21-oic acid, (18R,23S)-23-(Bis(2-(2-chloroacetamido)ethyl)carbamoyl)-1-bromo-18-carboxy-2,7,16,21-tetraoxo-11,14-dioxa-3,8,17,22-tetraazahexacosan-26-oic acid, (R)-4-{2-[2-({Bis-[2-(2-chloro-acetylamino)-ethyl]-carbamoyl}-methoxy)-ethoxy]-thylcarbamoyl}-2-[(S)-2-(2-{2-[2-(2-bromo-acetylamino)-ethoxy]-ethoxy}-acetyl amino)-4-carboxy-butyrylamino]-butyric acid, (4S,18S)-4-(bis(2-(2-Bromoacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-chloroacetamido)ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid, (4S)-5-[bis[3-[(2-chloroacetyl)amino]propyl]amino]-4-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-oxo-pentanoic acid, (2R)-6-[bis[2-[(2-chloroacetyl)amino]ethyl]carbamoylamino]-2-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]hexanoic acid, N-[2-[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]ethylcarbamoyl-[2-[(2-chloroacetyl)amino]ethyl]amino]ethyl]-2-chloro-acetamide, and (2R)-2-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-[2-[2-[2-[[(1S)-3-carboxy-1-[2-[(2-chloroacetyl)-[2-[(2-chloroacetyl)amino]ethyl]amino]ethylcarbamoyl]propyl]amino]-2-oxo-ethoxy]ethoxy]ethylamino]-5-oxo-pentanoic acid. 16. The protein conjugate according to claim 15, wherein Protein1 is a growth hormone compound. 16 CROSS-REFERENCE TO RELATED APPLICATIONS This application is a 35 U.S.C. § 371 National Stage application of International Application PCT/EP2016/073470 (WO2017/055582), filed Sep. 30, 2016, which claims priority to European Patent Application 15187937.6, filed Oct. 1, 2015; the contents of which are incorporated herein by reference. In accordance with 37 C.F.R. § 1.52(e)(5), Applicants enclose herewith the Sequence Listing for the above-captioned application entitled “150010US01_SeqList.txt”, created on Mar. 20, 2018. The Sequence Listing is made up of 7 kilobytes, and the information contained in the attached “SEQUENCE LISTING” is identical to the information in the specification as originally filed. No new matter is added. TECHNICAL FIELD The field of the invention is protein conjugates and methods of preparing such. BACKGROUND Protein conjugates are useful in multiple situations and the identification and development of biological therapeutic compounds of increasing complexity have increase the focus on attractive methods for preparing such compounds. Difficulties with linkage of two or more proteins arise as proteins are not as stable as traditional chemical moieties and traditional reaction chemistry can usually not be applied without damaging the proteins. Conjugation of a protein with a property modifying agent has been obtained by various methods linking to various amino acid residues such as the N-terminal, the C-terminal, and internal amino acid residues such as Cys, Lys, Gln and Ser that can be reacted with various reactive groups e.g. placed at the end of the property modifying agent. Traditionally proteins have been linked recombinant by expression of fusion proteins possibly linked by a peptide linker. This strategy may result in expression of very large protein molecules which may encounter production problems and thus limits the scope of compounds that can be efficiently produced. In an alternative to fusion proteins, WO2005001025 also describes native ligations e.g. linkage of a thioester to an N-terminal cysteine resulting in amide bond formation. Again, such linkage is limited to the N-terminal of the protein. Chemical linkage of proteins have been explored using di-halomethylene-benzene, ‘Click’ chemistry between an azide and an acetylene unit and PEG linkers with propionaldehyde at the ends such as described in WO201001196. Further exploration of linkage technologies is desired to broaden the scope of compound formats that can be easily and effectively produced. SUMMARY The present invention relates to protein conjugates and methods of preparing such conjugates. The methods may be useful to covalently link two or more protein(s) in an ordered and regio-selective fashion. The protein conjugates may comprise one or more therapeutic proteins as well as one or more effector protein(s). The present invention provides an efficient process for protein-protein conjugation by means of thiol reactive linkers. By using e.g. halo-acetamide with different leaving groups the linkage of reactants can be controlled and linkage via one or more thiol-ether(s) obtained. Examples of such conjugates are: As demonstrated herein the method has been successfully employed for the formation of Fc-conjugates. Fc domains holds two Fc polypeptides and by using a trivalent linker the Fc domains can be covalently bond to a protein of interest via two cysteine residues, e.g. one in each Fc polypeptide representing two individual proteins in the general structure. An aspect of the invention relates to a protein-Linker-Fc conjugate comprising covalent linkage between a Linker and each of the polypeptides of the Fc-domain. The invention thus relates to a compound of structure The linkages to the Fc polypeptides are via sulfur atom's (—S—) derived from cysteine residues in the Fc polypeptides. The Linker is a chemical moiety and Protein1 is thus covalently linked to Protein2 and Protein3 via the linker and the sulfur atoms. An aspect of the invention relates to trivalent linkers as used herein for preparing various protein conjugates. The linker in an embodiment includes a central unit referred to as -U=which hold at least three bonding opportunities. Other features of the linker are spacer elements 1-3 (S1-S3) that links the central unit with the reactive ends (R1-R3), which are used to enable conjugation of the linker with the protein. In one embodiment the trivalent linker has the structure: wherein U represent a central unit, S1, S2 and S3 represent individual spacers and R1, R2 and R3 individually represent a reactive end. Examples provided herein demonstrate that a Nitrogen atom is a suitable central unit and that thiol reactive ends are suitable for linkage to free cysteines in one or more of the proteins to be conjugated. An aspect of the invention relates to a method for preparing protein conjugates where at least two proteins are to be conjugated. The use of linkers having two different reactive ends enables an ordered reaction process increasing specificity, purity and/or yield. In one embodiment the reactive ends of the linker holds two halo-acetamides with different halogens providing reactive ends with different reactivity. After a first conjugation step the conjugate intermediate can be reacted with the 2 (or 3) protein. The efficacy of the method is increased if one of the halogen of the halo-acetamides is exchanged from Cl to I. The method may be employed for conjugation of two or more proteins and also if two of the proteins are identical and to be conjugated to the linker at the same time. An embodiment of the invention relates to a method for preparation of a protein conjugate, wherein Protein1-SH, Protein2-SH and a thiol reactive linker are coupled together obtaining a protein conjugate of Formula II Protein1-S—CH2—C(═O)—NH-Linker-NH—C(═O)—CH2—S-Protein2  (Formula II) wherein the thiol reactive linker has the structure: LG1-CH2—C(═O)—NH-Linker-NH—C(═O)—CH2-LG2, wherein LG1 has a higher reactivity than LG2, the method comprising the steps of: a) reacting Protein1-SH with —NH—C(═O)—CH2-LG1 of the linker b) obtaining a conjugate intermediate: Protein1-S—CH2—C(═O)—NH-Linker-NH—C(═O)—CH2-LG2 c) performing a leaving group exchange reaction increasing the reactivity of LG2. d) reacting the intermediate of c) with Protein2-SH e) obtaining the protein conjugate. As can be foreseen from the disclosure herein, the method, linkers and compounds disclosed may have multiple uses, such as in the development of therapeutic products. BRIEF DESCRIPTION OF DRAWINGS FIG. 1 shows a schematic illustration of a protein to Fc conjugation according to the invention. The trivalent linker includes, the central unit (here illustrated by triangle), independent spacer element S1, S2 and S3 and leaving groups LG1, LG2 and LG3. The method is described as follows: 1) Optionally reducing a mixed disulfide (I) of the protein to be conjugated obtaining a protein with a free Cys (—SH) (II) 2) Alkylating the free Cys (—SH) (II) with the trivalent linker (III) affording a Cys conjugated protein linker intermediate conjugate (IV) 3) Activating leaving groups LG2 and LG3 of the conjugate intermediate (IV) via an aqueous Finkelstein iodine exchange reaction (V) affording a iodine activated conjugate intermediate (VI) 4) Selective reduction of a Fc-domain disulfide bridge (VII) affording an Fc-domain with two reduced cysteines (—SH) (VIII) 5) Coupling of said Fc-domain (VIII) with the iodine activated conjugate intermediate (VI) affording a protein-Fc conjugate (IX). FIG. 2 shows a schematic illustration of an Fc protein conjugation according to the invention. The trivalent linker includes, the central unit (here illustrated by triangle), independent spacer element S1, S2 and S3 and leaving groups LG1, LG2 and LG3. The method is described as follows: 1) Selective reduction of a Fc-domain disulfide bridge (VII) affording an Fc-domain with two reduced cysteines (—SH) (VIII) 2) Alkylating the Fc-domain (VIII) with an trivalent linker (III) affording an LG1-A-B-Fc conjugate intermediate (X) 3) Optionally reducing a mixed disulfide (I) of the protein to be conjugated obtaining a protein with a free Cys (—SH) (II) 4) Activating leaving groups LG1 of the conjugate intermediate (X) via an aqueous Finkelstein iodine exchange reaction (V) affording a iodine activated conjugate intermediate (XI) 5) Coupling said protein with a free Cys (—SH) (II) with the iodine activated conjugate intermediate (XI) affording a protein-Fc conjugate (IX). SEQUENCE INFORMATION Standard Fc polypeptide sequences for IgG1 and IgG4 are provided in the sequence listing and replicated here for ease of information. SEQ ID NO 1: IgG1 C2-C3 Corresponding to AA231-447 of full length heavy chain according to EU numbering    APELLGG PSVFLFPPKP KDTLMISRTP EVTCVVVDVS HEDPEVKFNW YVDGVEVHNA KTKPREEQYN STYRVVSVLT VLHQDWLNGK EYKCKVSNKA LPAPIEKTIS KAKGQPREPQ VYTLPPSREE MTKNQVSLTC LVKGFYPSDI AVEWESNGQP ENNYKTTPPV LDSDGSFFLY SKLTVDKSRW QQGNVFSCSV MHEALHNHYT QKSLSLSPGK Amino acid residues underlined correspond to L234, L235, G237 and A330 and P331 SEQ ID NO 2: IgG1 hinge Corresponding to AA217-230 of full length heavy chain according to EU numbering PKSCDKTHTCPPCP SEQ ID NO 3: IgG4 C2-C3 Corresponding to AA231-447 of full length heavy chain according to EU numbering APEFLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSQED PEVQFNWYVD GVEVHNAKTK PREEQFNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKGLPS SIEKTISKAK GQPREPQVYT LPPSQEEMTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSRL TVDKSRWQEG NVFSCSVMHE ALHNHYTQKS LSLSLGK SEQ ID NO 4: IgG4 hinge Corresponding to AA217-230 of full length heavy chain according to EU numbering SKYGPPCPSCP S in bold and underlined corresponds to S228 of full length IgG4 heavy chain according to EU numbering. DESCRIPTION The present invention relates to protein conjugates and method of preparing such conjugates. The methods may be useful to covalently link two or more polypeptides. Protein Conjugates The present invention relates to protein conjugates e.g. compound including two or more proteins or polypeptides that are covalently linked by post translational chemical reactions. Such compounds and molecules may find use in multiple areas and in particular in relation to the development of therapeutic compounds. Protein/Polypeptide The proteins or polypeptides may be any proteins or polypeptides that a skilled person wishes to covalently link together. The present invention thus goes beyond the exemplified compounds as a skilled person can easily adapt the method to other proteins and polypeptides. In the following focus will be on conjugations involving a therapeutic protein and an effector protein aimed at modifying the properties of the therapeutic protein. Again alternative uses of compounds of the invention are foreseen. As can be seen herein the technologies developed are functional for proteins/polypeptides of various sizes. It is well-know that handling of proteins/polypeptides is substantially more challenging than handling of small peptides that can be treated more or less as small molecules. In one embodiment one or more or the proteins/polypeptides are at least 40 amino acids long, such as at least 60, 80 or 100 amino acids long. In one embodiment the proteins/polypeptides are all at least 40 amino acids long, such as at least 60, 80 or 100 amino acids long. Therapeutic Protein A therapeutic protein is a protein or polypeptide e.g. an amino acid sequence that is useful in a method of treatment of a disease or disorder. Growth Hormone The term “growth hormone compound” as used herein collectively refers to a growth hormone molecule retaining substantially the functional characteristics of mature human growth hormone identified by SEQ ID NO 5. The compound may thus be a growth hormone, a growth hormone fusion protein, a growth hormone variant or analogue or a growth hormone conjugate or derivative including also acylated or alkylated growth hormone. The ability of a growth hormone compound to stimulate signalling through the growth hormone receptor (GHR) may be measured in an in vitro cell based assay such as a BAF assay (Assay 2 herein). As a GH variant or GH compound comprising a variant amino acid sequence or other modification may have other advantageous, the GH activity as measured in a BAF assay can be lower than for human growth hormone (hGH) while the variant or compound is still an attractive molecules as long as the molecules are able to stimulate the receptor and proliferation of the cells to a reasonable degree. In one such embodiment the in vitro activity is measured in a BAF assay. In one embodiment the GH variant has an equal in vitro activity in a BAF assay compared to hGH identified by SEQ ID NO 5. As described in Assay 2, the result of the BAF assay (BAF ratio) may be expressed as the ratio between EC50 of the test compound (variant/GH compound) and EC50 for the reference (hGH/GH compound w. hGH sequence). In one embodiment the in vitro activity of the GH variant or the GH compound is comparable to the in vitro activity of hGH or the equivalent GH compound comprising the hGH sequence. Comparable here means that the ratio of BAF activity is within the interval of 1/100-100/1 or such as 1/10-10/1. Rat models are frequently used to test biological effect of GH variants and compounds. Testing may be performed in normal rats and/or in hypophysectomised rats. The Sprague Dawley rat is frequently used and methods for testing are described in Assay 3 and 4. Such testing may provide information on several pharmacokinetic parameters such as the AUC, T1/2 (half-life) and MRT (mean residence time) which are relevant in order to determine the total exposure and the duration of the presence of a given compound in the blood of a recipient. Furthermore an induction of the IGF-1 response, one of more characteristics for the biological effects of hGH, can be measured (Assay 5). As an alternative or supplement minipigs may be used as described in Assay 6. In one embodiment the GH conjugate has an increased half-life compared to hGH (SEQ ID NO 5). In one embodiment the GH conjugate according to the invention has an increased in vivo T1/2 compared to hGH (SEQ ID NO 5). It is noted that hGH has a T1/2 of approximately 12-14 minutes in the described assay 3 herein. Although not equivalent with half-life in humans, it is contemplated that an increased in vivo T1/2 in rats or minipigs will also translate into an extended in vivo presence in a therapeutic setting. In one embodiment the GH conjugate has a T1/2 above 30 minutes, or above 60 minutes, or above 90 minutes or above 120 minutes. In further embodiments T1/2 is above 60 minutes or 1 hour, such as above 2 hours or preferably above 4 hours. In on embodiment the GH conjugate has a T1/2 of 2-10 hours, such as 4-8 hours. In one embodiment the extended T1/2 is measure after intravenous (iv.) or subcutaneous (sc.) administration to rats or minipigs. The skilled person will know how such assay can be modified, depending on the tools available for detection of the GH variant or GH compound. In one embodiment the GH compound has an increased half-life compared to hGH. In one embodiment the GH compound has a T1/2 of more than 8 hours, such as more than 12 hours, such as more than 24 hours. In one embodiment the GH compound has a T1/2 of more than 8 hours, such as more than 12 hours, such as more than 24 hours, when measured after a single i.v. dose of 15 nmol to normal rats. In one embodiment the GH compound has a T1/2 of more than 8 hours, such as more than 12 hours, such as more than 24 hours, when measured after a single i.v. dose of 15 nmol to hypophysectomised rats (see Assay 4 herein). In one embodiment the GH compound has a T1/2 of more than 48 hours, such as more than 60 hours, such as more than 72 hours, when measured after a single iv. dose of 15 nmol to hypophysectomised rats. The IGF-1 response may be measured after dosing of a GH compound such as described in Assay 5, herein, although the skilled person will know to apply alternative methods as well. The plasma concentration of IGF-1 in rats after a single dose a GH should preferably increase over a period of time corresponding to the increased plasma concentration of the GH compound. In one embodiment the GH compound according to the invention is capable of inducing an IGF-1 response. An IGF-1 response may thus be stronger than the response observed for hGH by reaching a higher plasma concentration of IGF-1. The concentration of plasma IGF-1 may be detected within 72 hours, such as within 48 hours, such as within 36 hours, such as within 24 hours. To compare effects of different compounds values may be measured at different time points and compared at each individual time point, such as by either of 6, 12, 24, 36, 48, 72, 96, 144, 192, 240, 288, 336 hours after a dosage. In one embodiment the GH compound induces an increased IGF-1 response. In one embodiment the GH compound induces an IGF-1 response, wherein the IGF-1 response is detected as an increased plasma IGF-1 concentration at up to 96 hours, or such as 6, 12, 24, 36, 48, 72 hours after a single dose of said GH variant or compound. In one embodiment the GH compound induces an extended IGF-1 response. If the plasma concentration of IGF-1 remains high over an extended period of time compared to hGH, the GH compound induces an extended an IGF-1 response. In one embodiment the GH compound induces an extended IGF-1 response compared to the IGF response of wt hGH. In one embodiment the IGF-1 response lasts more than 24 hours, such as more than 48 hours. The structure of growth hormone proteins is composed of four helixes (helix 1-4) connected by three loops (L1-3), and a C-terminal segment. In human growth hormone (SEQ ID NO 5) helix 1 is defined by AA residue 6-35, helix 2 is defined by AA residues 71-98, helix 3 is defined by AA residue 107-127 and helix for is defined as AA residues 155-184. Growth hormone molecules including human growth hormone variants and conjugates have been described in multiple documents including WO2011089250, WO2011089255 and WO2012010516. In one embodiment a growth hormone compound or conjugate according to the invention comprises a GH protein with less than 8 modifications (substitutions, deletions, additions) relative to hGH. In one embodiment a GH protein comprises less than 7 modifications (substitutions, deletions, additions) relative to hGH. In one embodiment a growth hormone protein comprises less than 6 modifications (substitutions, deletions, additions) relative to human growth hormone. In one embodiment a growth hormone protein comprises less than 5 modifications (substitutions, deletions, additions) relative to human growth hormone. In one embodiment a growth hormone protein comprises less than 4 modifications (substitutions, deletions, additions) relative to human growth hormone. In one embodiment a growth hormone protein comprises less than 3 modifications (substitutions, deletions, additions) relative to human growth hormone. In one embodiment a growth hormone protein comprises less than 2 modifications (substitutions, deletions, additions) relative to human growth hormone. In a series of embodiment the growth hormone protein of the growth hormone is at least 95, 96, 97, 98 or 99% identical to human growth hormone identified by SEQ ID NO: 5. In one embodiment the growth hormone protein is a variant that is stabilized towards proteolytic degradation (by specific amino acid substitutions generated by mutation of the coding DNA sequence) Non-limiting examples of growth hormone proteins that are stabilized towards proteolytic degradation may be found in WO2011089250. Protease-stabilized growth hormone protein variants include variants where an additional disulfide bridge is introduced. The additional disulfide bridge preferably connects L3 with helix 2. This may be obtained by introducing two extra cysteine amino acid residues, which in preferred embodiments are substituted for the wild type amino acid residue in positions corresponding to AA84 or AA85 in H2 and AA143 or AA144 in L3 of SEQ ID NO: 5. The growth hormone variant may thus according to the invention preferably comprise a pair of amino acid substitutions corresponding to L73C/S132C, L73C/F139C, R77C/I138C, R77C/F139C, L81C/Q141C, L81C/Y143C, Q84C/Y143C, Q84C/S144C, S85C/Y143C, 585C/5144C, P89C/F146C, F92C/F146C or F92C/T148C in SEQ ID NO:5. In a further embodiment the growth hormone variant comprises a pair of amino acid substitutions corresponding to L81C/Y143C, Q84C/Y143C, S85C/Y143C, 585C/5144C or F92C/T148C in SEQ ID NO: 5. In one embodiment the growth hormone protein is a growth hormone variant, suited for mono-substitution/site specific modification such as alkylation by one chemical moiety to a free cysteine introduced by amino acid substitutions (by mutation of DNA sequence) possibly in addition to any protease stabilizing amino acid changes described above. A non-limiting list of growth hormone variants suitable for alkylation may be found in WO2011089255. The terms “free Cys” or “free cysteine” are used herein to indicate a cysteine amino acid residue which in the reduced form is available for conjugation, hence having a free thiol group (—SH). In general the free Cys is not engaged in a disulfide bond. Usually the free Cys is a variant amino acid introduced to the protein although a natural Cys may serve as the free Cys. The ability to introduce a free Cys by insertion or amino acid substitution in a protein greatly enhances the options for creating new molecules. In a further embodiment the protein is a growth hormone variant including a free cysteine. In a further embodiment the protein is a growth hormone variant including a free cysteine introduced in human growth hormone identified by SEQ ID NO.: 5. In a further embodiment the protein is a growth hormone variant including an additional cysteine introduced by an amino acid substitution selected from the group of: T3C, P5C, S7C, D11C, H18C, Q29C, E30C, E33C, A34C, Y35C, K38C, E39C, Y42C, S43C, D47C, P48C, 555C, 557C, P59C, S62, E65C, Q69C, E88C, Q91C, 595C, A98C, N99C, S100C, L101C, V102C, Y103C, D107C, S108C, D112C, Q122C, G126C, E129C, D130C, G131C, P133C, T135C, G136C, T142C, D147C, N149C, D154C, A155C, L156C, R178C, E186C, G187C and G190C. Such introduced Cys residues are termed free Cys substitutions. In a further embodiment the protein is a growth hormone variant including an additional cysteine selected from the group of: T3C, P5C, S7C, D11C, H18C, Q29C, E30C, E33C, A34C, Y35C, E88C, Q91C, 595C, A98C, N99C, S100C, L101C, V102C, Y103C, D107C, S108C, D112C, Q122C and G126C. In further embodiments the free Cys substitution is located within AA 93-106 in hGH or corresponding residues in hGH variants. In further specified embodiments the free Cys substitution is located within L2, such as within AA 99-106 or AA 99-103 or corresponding residues. In further embodiment the free Cys substitution is selected from the group of: E30C, Y42C, 555C, 557C, S62C, Q69C, 595C, A98C, N99C, L101C, V102C, and S108C. In an embodiment the growth hormone variant include one free Cys substitution. In a further embodiment the free Cys substitution is E30C. In further embodiment the free Cys substitution is Y42C. In a further embodiment the free Cys substitution is 555C. In a further embodiment the free Cys substitution is 557C. In a further embodiment the Cys substitution is 562C. In a further embodiment the free Cys substitution is Q69C. In further embodiment the free Cys substitution is 595C. In a further embodiment the free Cys substitution is A98C. In further embodiment the free Cys substitution is N99C. In a further embodiment the free Cys substitution is S100C. In a further embodiment the free Cys substitution is L101C. In a further embodiment the free Cys substitution is V102C. In a further embodiment the free Cys substitution is S108C. In a further embodiment the protein is a growth hormone variant including a cysteine substitution selected from Y42C and L101C. Effector Protein An effector protein is a polypeptide capable of modifying the properties of the (therapeutic) protein. Examples—but not limited to—of effector proteins are PEG, albumin, XTEN, and Fc-domain, the latter being the key example of the present application. Fc-Domain The fragment crystallizable region (Fc region or Fc-domain) of an antibody is the tail of an antibody. For IgG, IgA and IgD antibodies the Fc region contains two identical polypeptides both comprising the second and third constant domains (CH2 and CH3) of the heavy chain. The Fc regions of IgM and IgE antibodies contain three heavy chain constant domains (CH domains 2-4) in each polypeptide chain. The protein sequences of the Fc-domain are herein referred to as Fc polypeptides and usually comprise at least the CH2 and CH3 domains. The Fc-domain may also be referred to as a dimer as the two Fc polypeptides interacts non-covalent and possibly also covalently as hinge cysteines may form disulfide bond(s). The Fc-domain mediates interaction with cell surface receptors called Fc receptors, as well as some proteins of the complement system. The interaction with the Fc neonatal receptor (FcRn) is of particular interests. The Fc region enables antibodies to interact with the immune system. The Fc region of an antibody is at least partly responsible for the long in-vivo half-life of antibody molecules, which for an IgG is approximately 720 hours in humans. The Fc-domain is thus an attractive protractor for extending the in-vivo half-life of potential therapeutic compounds. According to the present invention it has been found that the use of an Fc-domain as a protractor of growth hormone results in a growth hormone conjugate with attractive functionalities. In one embodiment the isotype of the Fc-domain is IgG, such as subtype IgG1, such as IgG2, such as IgG4. In one embodiment the Fc domain comprises the CH2 and CH3 domains of human IgG1 defined by SEQ ID NO: 1 or IgG4 defined by SEQ ID NO 3. In one embodiment the growth hormone conjugate comprises two identical Fc polypeptides each defined by SEQ ID NO 1 or SEQ ID NO 3. The hinge region is the protein segment between CH1 and CH2 of the constant region of the antibody. In one embodiment the Fc-polypeptide comprises a hinge region including one or more cysteine's. In one embodiment the polypeptides of the Fc domain each comprises the sequence as defined by SEQ ID NO: 2 or 4. In one embodiment the Fc polypeptide comprises a hinge and the CH1 and CH2 domains. In one embodiment, the hinge region is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. In one embodiment the hinge regions of the Fc polypeptides comprise only one cysteine. In one embodiment this cysteine is capable of forming a disulfide bond with the same cysteine of the second Fc polypeptide. Thus in one embodiment the two polypeptides of the Fc-domain holds two cysteines in the hinge region e.g. one in each polypeptide. As seen in the section describing the preparation of proteins conjugates, such a disulfide bond may be reduced and the thiols use for coupling to the linker and there trough to other proteins. The cysteines of the Fc polypeptide are capable of forming a disulfide bond, but may act as a free Cys when reduced. In one embodiment the Fc polypeptide comprise a Cys residue. In particular as shown herein the disulfide bonds in the hinge region which links the two Fc polypeptide may be reduced producing two cysteines that can act like free cysteines. In one embodiment the Fc polypeptide comprise a Cys residue in the hinge sequence. In one embodiment the hinge region of the Fc polypeptide include only native amino acid residues. In one embodiment the hinge region comprise an amino acid insertion or substitution in the hinge region. For heterologous expression a methionine may be encoded by the DNA in the expression vector although not always present in the Fc domain of the conjugate. In on embodiment the Fc polypeptide does not include a methionine at the N-terminal. In one embodiment the hinge sequence is a truncated version of an IgG hinge sequences, such as the IgG1 or IgG4 hinge sequences specifically mentioned herein. In one embodiment the hinge sequence of the Fc hinges is derived from the IgG1 hinge sequence PKSCDKTHTCPPCP (SEQ ID NO: 2). In one embodiment the hinge sequence is selected from the group consisting of: PKSCDKTHTCPPCP, PPCP, PCP and CP. In one embodiment the hinge sequence of the Fc hinges is derived from the IgG4 hinge sequence SKYGPPCPSCP (SEQ ID NO: 4). In one embodiment the hinge sequence is selected from the group consisting of: SKYGPPCPS*CP, PSCP, SCPL and CP. In one embodiment the Fc polypeptide comprise a Cys residue in the hinge sequence. In one embodiment the Fc polypeptide comprise only one Cys residue in the hinge. In one embodiment the constant region may be modified to stabilize the molecule, for example, in an IgG4 hinge region, residue S228 (marked * above, residue numbering according to the EU index) may be substituted by a proline (S228P). In one embodiment the Fc polypeptides includes a proline residue in position 228, or in a position corresponding to 228 in an IgG4 derived hinge sequence. The Fc polypeptides of the Fc-domain may thus be covalently linked by disulfide bridges or alternatively non-covalently bound. In one embodiment the Fc region may be engineered to include modifications within the Fc region, typically to alter one or more of its functional properties, such as serum half-life, complement fixation, Fc-receptor binding, protein stability and/or antigen-dependent cellular cytotoxicity, or lack thereof, among others. In one embodiment the Fc domain comprises and FcRn binding site, thus any amino acid deletions, insertions or substitutions relative to the wt Fc polypeptide should not disrupt or decrease substantially the ability of the Fc domain to interact with the Fc neonatal receptor similar to what is described in WO05001025. Binding assays for such receptor interactions are well known in the art. Furthermore, an Fc-domain of the invention may be chemically modified (e.g., one or more chemical moieties can be attached to the Fc part) to alter its degree of glycosylation, again to alter one or more functional properties of the antibody. A variety of such mutations to the Fc domain have previously been described and the Fc domain according to the present invention may include such mutation as long as the functionality is maintained e.g. the ability to increase the in-vivo half-life of the protein linked thereto. An IgG1 Fc-domain may comprises one or more, and perhaps all of the following amino acid substitutions that will result in decreased affinity to certain Fc receptors selected from L234A, L235E, and G237A, and/or in reduced C1q-mediated complement fixation selected from A330S and P331S, respectively. In order to improve binding affinity to FcRn mutations in the Fc may be included to obtain amino acid substitutions such as M428L and/or N434S in an Fc-domain of the IgG1 isotype. Linker The linker is a chemical moiety used to covalently link the proteins. The linker is a separate moiety e.g. proteins solely linked by disulfide bonds are not consider to comprise a linker according to the present invention. As seen below the linker may include amino acid, amino acid like and non amino acid elements, while the linker is not itself produced by heterologous expression as part of one or more of the proteins of the conjugate. As the linker is reacted with a protein a linker radical is formed. The term “-Linker-” is thus intended to mean the chemical unit of the protein-conjugate which is covalently linked to an amino acid residue of each of the proteins of the protein conjugate. Depending on the attachment point the reactivity of the linker ends will vary. The linker may have various forms depending on the desired product to be obtained. The concept as described herein in an embodiment relates to an ordered conjugation to ensure that different proteins are attached at each end of the linker. A reactive end is a chemical sub-structure that is useful for conjugation of the linker to an amino acid residue of a protein. The reactive end may be suited for linkage to the N-terminal, the C-terminal or internal amino acid residues. Various formats are known in the art including chemical structures that are amino acid residue specific as well as structures that are amino acid residue unspecific. Depending on the target protein it may be desired to target specific amino acid residues to obtain a high yield of the desired product. The reactive end (or group) will differ depending on which amino acid residue it should target. As demonstrated in the Examples conjugation with thiols can be obtained using various cysteine or thiol-reactive ends, while other reactive groups are suitable for conjugation to alternative amino acid residues. The resulting conjugate includes the radical of the reactive end as part of the linker. The resulting radical of the reactive end will be covalently bond to the amino acid residue of attachment. The radical of the reactive end may be referred to as -RR-. The -Linker- moiety of the conjugates may thus be further described by specifying the reactive end radical(s) (-Linker-RR). The N-terminal amino acid may be targeted by an aldehyde or ketone. In one embodiment an N-terminal reactive end comprises —CHO. Lys residues may be target by i.e. a 2,5-dioxopyrrolidin-1-yl. In one embodiment the reactive end comprises a Lys reactive end, such as a dioxopyrrolidin. A Gln residue may be targeted by a two-step process described in WO2005070468 using transglutaminase to create and aldehyde that is reactive with an amine or hydroxyl amine. In one embodiment the reactive end comprises an aldehyde or ketone. A Ser residue may be oxidised using sodium periodate (NaIO4) to an aldehyde (glyoxyl) which can be target by an amine under reductive amination conditions. A Lys residue may likewise be suitable for a reveres transglutaminase reaction as described in WO2009027369. In order to describe the linker in further details R1, R2 etc. may be used to describe the individual reactive ends. In one embodiment the individual reactive ends are selected from C-term, N-term, Gln, Lys, Ser or Cys reactive ends. In one embodiment R1, R2 etc. is an N-terminal reactive end. In one embodiment R1, R2 etc. is a Gln reactive end. In one embodiment R1, R2 etc. is a Lys reactive end. In one embodiment R1, R2 etc. is a Ser reactive end. In one embodiment R1, R2 etc. is a Cys reactive end. In order to obtain regio-selectivity it may be preferred that the reactive ends differ, such as providing linkers having one Lys reactive end and one Cys reactive ends. If two Cys reactive ends are used the reactivity may be controlled as described below for bivalent and trivalent linkers. The linkers according to the invention may comprise one or more thiol or cysteine reactive ends separated by a spacer. The reactive ends of the linker may in an embodiment be referred to as Cys or thiol reactive ends. It is preferred that the reactive end is capable of reacting with the thiol independent on the position of the thiol. In one embodiment the thiol reactive end enables linkage to an N-terminal cysteine. In one embodiment the thiol reactive end enables linkage to a C-terminal cysteine. In one preferred embodiment the thiol reactive end enables linkage to an internal amino acid residues e.g. a free cysteine as described herein. The skilled person is aware of several routes enabling coupling to cysteines. Two key reactive ends are alpha-substituted acetamides (e.g., alpha-halogen acetamides) with a suitable leaving group (LG-acetamide) and alpha-beta-unsaturated carbonyl compounds (such as e.g., maleimides). The reactive end may thus be such as a maleimide or a LG-acetamide, wherein the leaving group is e.g. a sulfonic ester (such as tosylate or mesylate) or a halide (forming an alpha halo-acetamide). The halogen may be C1, Br or I. In an alternative embodiment the leaving (LG) group may be an alternative molecule providing same functionality. A Cys reactive end (or thiol-reactive end) is thus a chemical entity that allows conjugation to cysteine residues in a protein of interest. Examples of Cys reactive end groups are such as a terminal aldehyde, a pyrrolidin-2,5-dione (2,5-Pyrroledione)(also referred to as maleimide) and a leaving group-acetamide (such as a halo-acetamide). In one embodiment the Cys reactive end comprises -pyrrolidin-2,5-dione. In one embodiment the Cys reactive end comprises —NHC(═O)—CH2-pyrrolidin-2,5-dione. In one embodiment the Cys reactive end comprises a leaving group, such as a halogen, exemplified by Bromide, Chloride or Iodide. In one embodiment the Cys reactive end comprises a halo-acetamide. For halo-acetamides, the reactive end —NH—C(═O)—CH2—I is more reactive than —NH—C(═O)—CH2—Br which again is more reactive than —NH—C(═O)—CH2—Cl. The halo-acetamide is reactive towards Cys residues and may therefore be used for coupling of two proteins each including a free Cys either a wt residue or more likely a variant amino acid residue introduced for the purpose of conjugation. As described further herein below, the reactive ends of the linker may comprise leaving group(s) which are different and the linker may thus have the overall structure: LG1-Linker-LG2. The leaving groups are in one embodiment halogens and in particular halogens with different reactivity. When the leaving group(s) are included as part of a halo-acetamide the reactivity is I>Br>>Cl. In one embodiment the invention relates to a linker of the structure: LG1-Linker-LG2, wherein both ends are Cys reactive and the reactivity of LG1 and LG2 are different. The moiety between the reactive ends is here called Spacer. The Spacer may consist of one or more spacer elements as described herein below. The Spacer elements may be linked by peptide bonds. When the Cys reactive end is a halo-acetamide a peptide bond (—C(═O)—NH—/—NH—C(═O)—) is comprised by the reactive end. In one embodiment the linker has the structure: Halo1-CH2—C(═O)—NH-Spacer-NH—C(═O)—CH2-Halo2, Cl—CH2—C(═O)—NH-Spacer-NH—C(═O)—CH2—Br or Br—CH2—C(═O)—NH-Spacer-NH—C(═O)—CH2—Cl Trivalent Linker As described above, the present invention covers linkage of a protein with an Fc-domain. Notably an Fc domain consists of two polypeptides that are usually held together by covalent and non-covalent bonds including also inter-polypeptide disulfide bond(s). Covalent linkage using a traditional bivalent linker the linkage would go from the protein to only one of the Fc polypeptides. Using a trivalent linker according to the present invention a protein conjugate where both of the two Fc polypeptide chains are linked to the protein can be obtained. Clearly, use of such trivalent linkers is not limited to the conjugation of Fc domains. The trivalent linker may have a structure including a central unit referred to as “U”, that is at least a tri-radical The central unit may be any chemical structure that allows for at least three bonds extending from the unit (-U=). The central unit may in one embodiment be and Nitrogen atom (—N═). The central unit may in one embodiment be a tetravalent carbon atom (═C═), in which case the forth “arm” may be —H, —CH3 or any other structure that does not interfere with the linker functionality. The central unit may in one embodiment be a benzene ring. The trivalent linker structure may comprise three linker arms that may be identical or different. The linker arms each comprise a spacer part (S) and a reactive end (R). The overall structure being: wherein U represents a tri-radical (central unit) S1, S2 and S3 represent individual spacers and R1, R2 and R3 represent individual reactive ends (suitable) for conjugation to a protein molecule. In one embodiment R1, R2 and R3 are not identical. In one embodiment R2 and R3 are identical. In one embodiment R2 and R3 are identical but R1 is different. In one embodiment one or more of the reactive ends R1, R2 and R3 are thiol reactive ends. In one embodiment R2 and R3 are thiol reactive ends. In one embodiment at least R2 and R3 are thiol reactive ends. In one embodiment R2 and R3 are thiol reactive ends while R1 is not a thiol reactive end. In one embodiment R1, R2 and R3 are thiol reactive ends. In one embodiment R1, R2 and R3 have different reactivity towards cysteines. In one embodiment R2 and R3 are thiol reactive ends and R1 is not a thiol reactive end. In such embodiments R1 may be a reactive end selected from C-term, N-term, Gln, Lys and Ser reactive ends. In one embodiment the thiol reactive end comprises a maleimide. In one embodiment the thiol reactive end comprise a leaving group (LG), such leaving group may be an inorganic leaving group, such as a halogen exemplified by bromide, chloride or iodide or an organic leaving group exemplified by such as mesylate or tosylate. In one embodiment the leaving group is a halogen such as bromide, chloride or iodide. In one embodiment one or more of the reactive ends is/are halo-acetamide(s). In one embodiment R2 and R3 are thiol reactive ends, such as —NH—C(═O)—CH2-LG, providing a linker with the structure: wherein LG2 and LG3 are leaving groups and R1 is a reactive end and S1, S2 and S3 represent individual spacers as above. In one embodiment the leaving groups 2 (LG2) and 3 (LG3) are identical, whereas in a further embodiment the leaving groups 2 and 3 are different. Different leaving groups may have different reactivity and enable sequential conjugation. In one embodiment R1 comprises a thiol reactive end. In on embodiment R1 is a thiol reactive end. In one embodiment the thiol reactive end is a haloacetamide. In one embodiment R1 comprises a leaving group (LG1), such leaving group may be an inorganic leaving group, such as a halogen exemplified by bromide, chloride or iodide or an organic leaving group such as mesylate or tosylate. In on embodiment the first linker arm has the structure: LG1-CH2—C(═O)—NH—S1. In one embodiment R1 comprises a leaving group (LG), such as a halogen, such as bromide, chloride or iodide. In one embodiment R1 is different from R2 and R3. In one embodiment R1 comprises a different leaving group than R2 and R3. In one embodiment LG1 is different from LG2 and LG3 In order to direct sequential conjugation of the linker arms to the different proteins to be conjugated different reactive ends can be used. In one embodiment where all arms include a thiol reactive end comprising a LG, the LG's may be different to achieve different reactivity towards the proteins. In one embodiment the reactivity's of the thiol reactive ends are different. In one embodiment the reactivity of LG1 is higher than the reactivity of LG2 and LG3. In one embodiment the reactivity of LG2 and LG3 is higher than the reactivity of LG1. The order of reactivity for the haloacetamides is I>Br>>CI. Thus a reactive end of —NH—C(═O)—CH2—Br will be more reactive than —NH—C(═O)—CH2—Cl and —NH—C(═O)—CH2—I will have even higher reactivity towards (reduced) cysteine residues. The linkers according to the invention may thus comprise one or more thiol or Cys reactive group. Key examples of Cys reactive groups and reactive end radicals are: The zigzag line and N—* indicate the attachment to the rest of the linker while the *- to the left indicates attachment to the —S— of the Cys amino acid residue. The linker of the invention may further comprise individual spacer segments that link the reactive end with the central unit. The spacer segments are designated S1, S2 and S3. As mentioned above the linkers allows for symmetrical conjugation of an Fc-domain, which is obtained when e.g. R2-S2 and R3-S3 are identical, while the linkage to a protein of interest via the first arm may be different. In other embodiments the conjugates are none-symmetrical and all of R1-S1, R2-S2 and R3-S3 may be different. The spacers S1, S2 and S3 may comprise different spacer elements. In a situation where protein 2 and 3 are to be in close proximity a short spacer may be used for S2 and S3. A short space could be from 1-10 atoms, such as 2-5 atoms in length counting the number of atom bound in the shortest distance. In one embodiment the short spacer is —(CH2)n—, wherein n is an integer in the range of 1-5. In one embodiment the short spacer is —(CH2)n—, wherein n is an integer in the range of 1-3, such as n=2 and such as n=3 and S2 and S3 are thus—(CH2)2— or —(CH2)3—. In one embodiment the distance from the central unit is increased by extending one or more of the spacers. In one embodiment the linker comprises at least one extended spacer. If the distance to the central unit is only to be increase for one of the proteins of the conjugate only one of the spacer should be extended. In one embodiment the extended spacer is S1. An extended spacer is longer that the short spacers exemplified above for S2 and S3. An extended spacer could be from 10-50 atoms, such as 20-30 atoms in length counting the number of atom bound in the shortest distance. In one embodiment where the central unit comprise a nitrogen (N) one arm of the linker may be linked to the N via an amide bond. In a further embodiment 51 (which connects R1 with the central unit (here N) has a carbonyl (C═O) at the end which may form an amide bond with the nitrogen atom. In further embodiment the spacers may include one or more amino acid like spacer elements. The spacer elements may be linked by amide bond(s). Such spacer element thus holds an N-terminal and a C-terminal as does an amino acid in a polypeptide. Such spacer elements may be amino acid residues or modified amino acid residues or alternative chemical entities capable of being linked by amide bonds. Examples are glycine, alanine, glutamic acid and gamma-Glu (γ-Glu) as shown in a) through d) below. The carbonyl end may form an amide bond with the central nitrogen in one of the linker arms. Alternative amino acid like elements including an additional amino group (instead of the carbonyl-group) as exemplified by e) and f) below may also be used next to the central Nitrogen in one of the linker arms. When coupled hereto and urea/carbamide group is present in the linker structure. The glycine spacer element may be extended by including polyethyleneglycol unites. In a further embodiment, the spacer comprises a polyethylene glycol (PEG) moiety. The PEG moiety being a bi-radical comprising the structure wherein n′ is an integer larger than 1. In one such embodiment n′ is an integer selected from 2-20. In one such embodiment n′ is an integer selected from 2-10 or 2-5. In one embodiment n′ is 2. In one embodiment a PEG moiety may have the structure In one embodiment the PEG moiety is included in an amino acid or amino acid like spacer element as described above. In one embodiment the spacer element has the formula. wherein k is an integer in the range of 1-5, and n is an integer in the range of 1-5. In one specific embodiment k=1 and n=1 providing a spacer element. In one embodiment the space element g) is defined by a k=1 and n=1 providing *—NH—(CH2)2—O—(CH2)2—O—CH2—C(═O)—*(e1) which may be referred to as OEG or a di-radical of 8-amino-3,6-dioxaoctanoic acid. In the structures above the zig-zag lines mark the bond to the spacer, the central unit or the reactive end. In the cases where the spacer links to the reactive end “—NH—” may be considered part of the reactive end. This may be the case in the embodiments described above where the reactive end holds a halo-acetamide. In one embodiment R1-S1- comprise 3-8 spacer elements linked by amid bonds. In one embodiment R1-S1- comprises 4-6 spacer elements linked by amid bonds. In one embodiment the structure of the trivalent linker is wherein leaving groups LG2 and LG3 are identical. In one such embodiment R1 is not a thiol reactive end. In one embodiment the structure of the trivalent liner is wherein leaving group LG2 and LG3 are identical and R1 is a Ser, Lys, Gln, C-term or N-term reactive end. In one alternative embodiment all reactive ends are Cys reactive. In one embodiment R2 and R3 are identical and R1 is a different Cys reactive end. Such different reactive ends will allow selected linkage of the proteins. In one embodiment R2 and R3 comprise a halo-acetamide. In one embodiment LG2 and LG3 are C1 with the linker having the overall structure: In one embodiment LG2 and LG3 are Br with the linker having the overall structure: In one embodiment all the reactive ends are halo-acetamides. In one embodiment R1 comprises a halo-acetamide including LG1. In one embodiment the structure of the trivalent linker is: In one embodiment the structure of the trivalent linker is: In one embodiment the structure of the trivalent linker is: In one embodiment the structure of the trivalent linker is: In one embodiment the structure of the trivalent linker is: In one embodiment the structure of the trivalent linker is: In an embodiment where the 2nd and 3rd arms are identical the linker moiety can be described by the generic structure A-B where A- is the 1st arm and -B comprises the central unit and the 2nd and 3rd arms. If similar structures are used as above A is R1-S1- and B is —N—(S2-R2)2. The overall size of the linker may thus vary, but will usually be relatively small as seen by the examples herein. When measuring the size of the linker only the linkerpart remaining in the conjugate is included meaning that the leaving groups are not include. In one embodiment the size of the linker is below 10 kDa, such as below 5 kDa, such as below 2 kDa, such as below 1 kDa, such as below 500 kDa. In one embodiment the linker is from 250 Da to 20 kDa, such as from 500 Da to 10 kDa. In one embodiment the linker is from 500-2000 Da, such as from 600-1500 da, such as 700-100 da. The length of the full linker of a conjugate may be estimated by counting the number of atoms in the shortest distance between two proteins. In cases where two arms are identical the length is the longest distance e.g. from Protein1 to Protein) in the examples where 2nd and 3rd arms are identical and 1st arm longest. When the central atom is N, this counts one atom while a benzene ring as central unit counts 3 atoms if arms are positioned symmetrically. As leaving groups are not part of the final conjugate such are not counted only the reactive end radical. In one embodiment the linker is 5-200 atoms in length such as 8-150 atoms or 10 to 100 atoms or in length. In a further embodiment the linker is 10-80 atoms in length. In a further embodiment the linker is 10-60 atoms in length. In a further embodiment the linker is 10-40 atoms in length, such as 15-40 or such as 20-35 atoms. Examples of trivalent linkers according to the invention are provided in table 1 here below. TABLE 1 Examples of Trivalent linkers. Structure and Name Protein Conjugates The protein conjugates of the invention will thus comprise 2 or more individual polypeptides (Protein1, Protein2 and/or Protein3) covalently bound to each other via the linker moiety. In one embodiment Protein1, Protein2 and Protein3 are individual polypeptides. In one embodiment the individual polypeptides may be identical or two of Protein1, Protein2 and Protein3 may be identical. The individual polypeptides thus have each an N-terminal and C-terminal amino acid residue. That said the individual polypeptides may additionally be attached to each other or even a further polypeptide/protein via e.g. disulfide bonds. As the proteins are bound to each other via the linker, the linker moiety will be part of all such compound including intermediates also covered by the present invention. In one embodiment the conjugate of the invention have the structure wherein Linker is a chemical moiety S is a sulfur atom and Protein1 is covalently linked to Protein2 and Protein3 via said linker and sulfur atoms. In one embodiment the sulfur atom is part of a thioether e.g. the bonds from the sulfur atoms goes to two individual carbon atoms C—S—C where the carbon atoms may be part of any organic structure. The most common thioether is —CH2—S—CH2— where in the present case one CH2 group stems from the protein linked to the linker and most frequently from the cysteine residue providing the sulfur atom for the conjugation. In one embodiment the sulfur atom of the conjugate is not part of a disulfide bond. In one embodiment where -S2-R2 is identical with -S3-R3, and two copies of Protein2 is to be conjugated with Protein1, the structure of the linker may be described by -A-B= and the resulting conjugate is of the structure Protein1-A-B-(Protein2)2. When B is consider a linker arm, the structure may be Protein1-A-(B-Protein2)2 Further examples of compounds of the invention are described here in below when addressing the method for preparation of the conjugates. Fc Conjugates In one embodiment Protein2 and Protein3 is/are the same Fc polypeptide, which together forms an Fc-domain. As described herein the linkage to an Fc polypeptide can be obtained by reducing a disulfide bridge of the hinge region and linking each of the cysteines to the linker, such as via linker arms 2 and 3. The 3rd linker arm (here linker arm 1) is either prior or later conjugated to a Protein1 of interest. As described in the section on preparation of protein conjugates the conjugates may have the form of: In an embodiment where the reactive ends 2 and 3 are haloacetamides the conjugate has the form: In one embodiment the conjugate has the structure: Protein1-RR1-S1-U-[S2-NH—C(═O)—CH2—S-Fc]2 wherein RR1 represents a reactive end radical, U represents a central unit, S1 and S2 represent individual spacers and Fc is an Fc polypeptide. If the alternative description above is used it follows that the Fc conjugates can be described by: Protein1-A-(B-Fc)2. If the Sulfur atom for linkage to Fc is include the structure is: Protein1-A-(B-S-Fc)2 and including the thioether the structure is Protein1-A-(B-CH2—S-Fc)2 or Protein1-A-(B-CH2-S—CH2-Fc)2 if the —CH2— of the cysteine of the Fc is shown as well. When the linkage is obtained by a thiol and haloacetamide coupling, B thus includes at least the —NH—C(═O)—CH2— element remaining from the thiol and haloacetamide reaction providing: Protein1-A-(B′-NH—C(═O)—CH2—S-Fc)2 It appears that S2 and B′ are then similar in the sense that they symbolize the remaining part of the linker arms. In a preferred embodiment as S2 or B′ is —CH2—CH2— providing a conjugate structure of: Protein1-A-(CH2—CH2—NH—C(═O)—CH2—S-Fc)2 wherein A is the linker unit connecting to Protein1 which, as has been described elsewhere in the application, comprise a central unit (with options for at least three bonds) and a suitable spacer and a reactive end radical providing the linkage to Protein1. Growth Hormone Fc Conjugates In an aspect the present invention relates to growth hormone Fc conjugates, such GH conjugates preferably has increased in vivo half-life (T1/2) compared to wild type human growth hormone. In addition the growth hormone Fc conjugates preferably maintain the therapeutic capabilities of human growth hormone which can be assayed in vitro by testing receptor binding and the activity in a BAF assay as described in Assay 1 and 2. Animal models may be used to further evaluate the therapeutic potential of growth hormone Fc conjugates (Assay 3-5). Examples of growth hormone conjugates according to the invention are included in table 3 here below. TABLE 3 GH-Fc compounds of example 2 # Com- pound Structure 1 2 3 4 5 Pharmaceutical Compositions A protein conjugate according to the invention may be formulated as a pharmaceutical composition. The formulation may further comprise a suitable buffer, a preservative, a tonicity agent, a chelating agent, a stabilizer, and/or a surfactant, as well as various combinations thereof. The use of preservatives, isotonic agents, chelating agents, stabilizers and surfactants in pharmaceutical compositions is well-known to the skilled person. The formulations may be prepared using standard procedures know in the art. Reference may be made to Remington: The Science and Practice of Pharmacy, 19th edition, 1995. In one embodiment of the invention the pharmaceutical composition is a liquid formulation. In one embodiment of the invention the pharmaceutical composition is an aqueous composition, i.e. a composition where the components are dissolved or suspended in water. Such composition is typically a solution or a suspension. If the composition comprises components which cannot be dissolved in water the composition may be an emulsion of two liquids, frequently water and oil or a fatty acid based liquid. In another embodiment the pharmaceutical composition is a freeze-dried composition, whereto the physician or the patient adds solvents and/or diluents prior to use. In one embodiment the composition of the invention has a pH of 5.0-8.5, such as 6.0-8.5, such as 6.0-8.2, such as 6.0-8.0, such as 7.0-8.5, such as 7.0-8.0, such as 7.5-8.0, such as 6.0-7.5, such as 6.2-7.5, such as 6.4-7.2 such as 6.5-7.0, such as 6.6-7.0. The pH may also be 6.6-6.9 or 6.7-6.9. In further embodiments the pH of the composition is 6.6, 6.7, 6.8, 6.9 or 7.0. In a further embodiment of the invention the buffer is selected from the group consisting of sodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, and tris(hydroxymethyl)-aminomethan, bicine, tricine, malic acid, succinate, maleic acid, fumaric acid, tartaric acid, aspartic acid or mixtures thereof. In one embodiment the pharmaceutical composition does not include glycine. In one embodiment composition comprises histidine as buffer. In one embodiment the pharmaceutical composition comprises a surfactant, such as a polyoxypropylene-polyoxyethylene block polymer. In one embodiment the surfactant is selected from non-ionic surfactants, such as poloxamers including Pluronic® F68, poloxamer 188 and 407 and Triton X-100. In one embodiment the surfactant is selected from polyoxyethylene and polyethylene derivatives such as alkylated and alkoxylated derivatives (tweens, e.g. Tween-20, Tween-40, Tween-80 and Brij-35). In one embodiment the surfactant is polysorbate 80. In a further embodiment the composition comprises a pharmaceutically acceptable preservative. In a further embodiment of the invention the preservative is selected from the group consisting of phenol, o-cresol, m-cresol, p-cresol, methyl p-hydroxybenzoate, propyl p-hydroxybenzoate, 2-phenoxyethanol, butyl p-hydroxybenzoate, 2-phenylethanol, benzyl alcohol, chlorobutanol, and thiomerosal, bronopol, benzoic acid, imidurea, chlorohexidine, sodium dehydroacetate, chlorocresol, ethyl p-hydroxybenzoate, benzethonium chloride, chlorphenesine (3-(p-chlorophenoxy)propane-1,2-diol) or mixtures thereof. Methods of Treatment Protein conjugates as described herein may be useful in the treatment of various diseases and disorders depending on the combined therapeutic effect of the proteins of the conjugate. It is will know that growth hormone compounds are suitable for treatment of growth hormone deficiencies. Basically a pharmaceutical composition according to the invention comprising a growth hormone protein conjugated may be for use in treatment of any disease or disorder where the patient will benefit from an increase in circulating growth hormone activity. In current treatments a growth hormone protein is administered. As an alternative growth hormone variants or compounds may be administered to provide growth hormone activity. An aspect of the invention is the growth hormone conjugate for use in a method of treatment. An aspect of the invention relates to the use of the growth hormone conjugate for the manufacture of a medicament for treatment, in particular treatment of growth hormone deficiency in children and/or adults or other diseases or states where the patient benefit from an increased level of growth hormone as described herein. The invention further relates to the aspects of preparation of a pharmaceutical composition according to the invention for use in a method of treatment as well as the pharmaceutical composition for use in a method of treatment comprising a protein conjugate including growth hormone conjugates. In such embodiments, the pharmaceutical composition according to the invention is for use in a method of treatment or prevention of growth hormone deficiency in children and/or adults. Other diseases or disorders where an increased concentration of circulating growth hormone may be helpful may also be treated or prevented using the pharmaceutical composition of the invention. In one embodiment the pharmaceutical compositions of the invention is for use in a method for treating diseases or states where a benefit from an increase in the amount of circulating growth hormone is observed. Such diseases or states include growth hormone deficiency (GHD); Turner Syndrome; Prader-Willi syndrome (PWS); Noonan syndrome; Down syndrome; chronic renal disease, juvenile rheumatoid arthritis; cystic fibrosis, HIV-infection in children receiving HAART treatment (HIV/HALS children); short children born short for gestational age (SGA); short stature in children born with very low birth weight (VLBW) but SGA; skeletal dysplasia; hypochondroplasia; achondroplasia; idiopathic short stature (ISS); GHD in adults; fractures in or of long bones, such as tibia, fibula, femur, humerus, radius, ulna, clavicula, matacarpea, matatarsea, and digit; fractures in or of spongious bones, such as the scull, base of hand, and base of food; patients after tendon or ligament surgery in e.g. hand, knee, or shoulder; patients having or going through distraction oteogenesis; patients after hip or discus replacement, meniscus repair, spinal fusions or prosthesis fixation, such as in the knee, hip, shoulder, elbow, wrist or jaw; patients into which osteosynthesis material, such as nails, screws and plates, have been fixed; patients with non-union or mal-union of fractures; patients after osteatomia, e.g. from tibia or 1st toe; patients after graft implantation; articular cartilage degeneration in knee caused by trauma or arthritis; osteoporosis in patients with Turner syndrome; osteoporosis in men; adult patients in chronic dialysis (APCD); malnutritional associated cardiovascular disease in APCD; reversal of cachexia in APCD; cancer in APCD; chronic abstractive pulmonal disease in APCD; HIV in APCD; elderly with APCD; chronic liver disease in APCD, fatigue syndrome in APCD; Chron's disease; IBD, UC, impaired liver function; males with HIV infections; short bowel syndrome; central obesity; HIV-associated lipodystrophy syndrome (HALS); male infertility; patients after major elective surgery, alcohol/drug detoxification or neurological trauma; aging; frail elderly; osteo-arthritis; traumatically damaged cartilage; erectile dysfunction; fibromyalgia; memory disorders; depression; traumatic brain injury; subarachnoid haemorrhage; very low birth weight; metabolic syndrome; glucocorticoid myopathy; or short stature due to glucocorticoid treatment in children. Growth hormones have also been used for acceleration of the healing of muscle tissue, nervous tissue or wounds; the acceleration or improvement of blood flow to damaged tissue; or the decrease of infection rate in damaged tissue. In one embodiment, the growth hormone conjugates and compositions hereof is for treatment of GHD in children, GHD in adults (AGHD), Turner syndrome (TS), Noonan syndrome, Idiopathic short stature (ISS), Small for gestational age (SGA), Prader-Willi syndrome (PWS), Chronic renal insufficiency (CRI), Skeletal dysplasia, SHOX deficiency, AIDS wasting, HIV associated lipdystrophy (HARS), Short bowel syndrome optionally including, steroid dependent disease, cystic fibrosis and fibromyalgia. In one embodiment the growth hormone conjugate or composition is for use in the manufacture of a pharmaceutical composition as described herein. In one embodiment, the present invention relates to a method of treating diseases or states mentioned above, wherein the activity of the pharmaceutical composition according to the invention is useful for treating said diseases or states. The administering of the pharmaceutical composition e.g. the growth hormone conjugate resulting in a therapeutic benefit associated with an increase in the amount of circulating growth hormone activity in the patient. In an embodiment said method comprises, administering to a patient an effective amount of the pharmaceutical composition comprising a growth hormone conjugate thereby ameliorating the symptoms of said patient. In one embodiment, the present invention relates to a method comprising administration to a patient in need thereof an effective amount of a therapeutically effective amount of a pharmaceutical composition according to the invention. The present invention thus provides a method for treating these diseases or states, the method comprising administering to a patient in need thereof a therapeutically effective amount of a growth hormone variant or compound in a pharmaceutical composition according to the present invention. A “therapeutically effective amount” of a compound of the invention as used herein means an amount sufficient to cure, alleviate or partially arrest the clinical manifestations of a given disease and its complications. An amount adequate to accomplish this is defined as “therapeutically effective amount”. Effective amounts for each purpose will depend on e.g. the severity of the disease or injury as well as the weight, sex, age and general state of the subject. As described herein the growth hormone variant or compound of the pharmaceutical composition may have an extended half-life aimed at increasing the exposure in the patient to the compound after each dosage and the administration regime of the pharmaceutical composition should be adjusted to reach an effective exposure. Method for Preparation of Protein Conjugates An aspect of the invention relates to a method for preparing protein conjugates as described herein. The proteins to be conjugated (protein1, protein2 and optional protein3) and the linker are produced separately and coupled together in a suitable reaction. Preparation of Proteins Depending on protein of interest various sources are available to the skilled person. The protein may be produced by recombinant only by heterologous expression in a suitable host, such as E. coli, yeast or mammalian cell (Molecular Cloning: A Laboratory Manual by Joseph Sambrook, E. F. Fritsch and J. Sambrook (Author). An example of GH preparation is provide in the Examples section herein and variation hereof can be performed as desired by the skilled person. The application holds examples with Fc-domains. Fc-domains may be obtained from full length antibodies isolated from humans and other animals or may be produced recombinant and obtained from transformed mammalian cells or microorganisms. Multiple technologies to obtain Fc-domains are known in the art. An Fc-domain can be produced from a full length antibody by digestion with a proteolytic enzyme such as papain or pepsine. Protein A affinity chromatography and DEAE anion-exchange chromatography can be used to separate the resulting Fab and F(ab′)2 from the Fc-domain. Based on SEC-HPLC analysis, purity of Fc-fragment can be determined. When recombinant methods are used the desired polypeptide can be expressed and the Fc domain subsequently purified. In one embodiment the Fc-domain is a human-derived Fc-domain, such as a human IgG Fc-domain obtained from transformed microorganisms or mammalian cells. In addition, the Fc-fragment of the present invention may be in the form of having native sugar chains, increased sugar chains compared to a native form or decreased sugar chains compared to the native form, or may be in an aglycosylated form. The increase, decrease or removal of sugar chains of the Fc-fragment may be achieved by methods common in the art, such as a chemical method, an enzymatic method and a genetic engineering method using a microorganism such as E. coli. An Fc-fragment from E. coli which will be aglycosylated will have diminished or weak binding to Fc gamma receptors I, IIa, IIb, IIIa, respectively, which has the advantage of low ADCC and CDC. Preferably is an aglycosylated hIgG4 Fc-fragment which naturally does not have binding to Fc gamma receptor III. Sulfur Atoms and Free Cysteine. As described herein above the linker is covalently bond to one or more proteins via a sulfur atom. The sulfur atom(s) is in an embodiment derived from protein thiol(s). The most common source of protein thiol is the amino acid cysteine. Cysteines may be engage in disulfide bonds and it may be preferred that the cysteine supplying the thiol is not usually engaged in disulfide bonds. Alternatively, a disulfide bond may be reduced providing two sulfur atoms available for conjugation. In one embodiment the sulfur atom is derived from a thiol of a cysteine amino acid present in the protein of the conjugate. As the protein conjugate may comprise one or more sulfur atoms, the sulfur atoms may be derived from one or more protein cysteines. A protein cysteine is thus a cysteine residue of a polypeptide of the protein. In one embodiment the cysteine may be a wild type residue, while in other embodiment the cysteine may be a variant cysteine, such as an amino acid substitution of a wild-type residue. As multiple Cys may be engage in the conjugate some —S— may be from a wt Cys while others may be a variant Cys. In one embodiment the invention relates to a conjugate according to any of the previous embodiments, wherein one or more of the protein cysteines are variant amino acid residues. The method described herein is suitable for preparing protein conjugates wherein at least one of the proteins to be conjugated includes a free cysteine. A free cysteine (Cys) is a cysteine residue available for conjugation via a thiol reactive linker. A free Cys is usually a cysteine residue that does not engage in intra protein disulfide bonds. As described herein above for human growth hormone a free Cys maybe generated by recombination introducing an amino acid in a suitable place in a protein of interest. Usually the amino acid insertion will be a substitution of a dispensable amino acid although a Cys could also be introduce as an additional amino acid. Frequently the free cysteine need to be liberated prior to the conjugation reaction as proteins with a free Cys may form mixed disulfide with other sulfur molecules usually small organic molecules present in the cell extract when the protein is produced and purified. Free cysteines may also be generated by reducing an existing disulfide bond which will make available two free cysteines. In one embodiment two equivalent cysteines may be generated by reducing an Fc-domain including at least one disulfide bond between the two polypeptides of the Fc-domain. In one embodiment an Fc-domain with a single disulfide bond between the two Fc polypeptides is prepared. In one embodiment the Fc-domain comprise a single disulfide bond in the hinge region of the Fc-domain. As illustrated by example 2, such a molecule can be linked with two arms of a trivalent linker using the method described herein. The resulting protein conjugation (or conjugate intermediate) will have a symmetric linkage with the Fc-domain and a third arm available for conjugation with a second protein. As described below the order of conjugation may be varied. Preparation of Linker The linkers of the invention may be produced by standard chemical technologies and multiple examples are included herein. Reaction Schemes for Protein Conjugates Depending on proteins used in the conjugation and the individual linker to be use various methods may be applied and it is foreseen that the skilled person is capable of adjusting the methods as set out herein for any specific needs without deviating from the concept of the invention. The proteins to be conjugated and the linker is prepared and purified separately. An aspect of the invention relates to a method for coupling of at least two proteins. A sequential reaction with two proteins is obtained by use of a linker with different reactive ends. In an embodiment where both proteins include a free cysteine such proteins are coupled together using a linker with two thiol reactive ends. The method is further exemplified using a linker with halo-acetamides as reactive ends as describe in relation to the linkers. The invention in an embodiment relates to a method for preparation of a protein conjugate, wherein Protein1-SH and Protein2-SH are coupled together obtaining a protein conjugate of Formula II Protein1-S—CH2—C(═O)—NH-Linker-NH—C(═O)—CH2—S-Protein2  Formula II using a thiol reactive linker: LG1-CH2—C(═O)—NH-Linker-NH—C(═O)—CH2-LG2, wherein LG1 has a higher reactivity than LG2, the method comprising the steps of: a) reacting Protein1-SH with —NH—C(═O)—CH2-LG1 of the linker b) obtaining a conjugate intermediate: Protein1-S—CH2—C(═O)—NH-Linker-NH—C(═O)—CH2-LG2 c) performing a leaving group exchange reaction increasing the reactivity of LG2. d) reacting the intermediate of c) with Protein2-SH e) obtaining the protein conjugate. In case only one Protein2 includes a free cysteine Protein1 may be coupled by an alternative route to provide an intermediate having the structure: Protein1-Linker-NH—C(═O)—CH2-LG. The method steps c), d) and e) may still be used in a slightly modified method where the conjugation of step a) is performed using an intermediate having the structure: Protein1-Linker-NH—C(═O)—CH2-LG In one embodiment the invention relates to method for preparing a protein conjugate wherein Protein1-Linker-NH—C(═O)—CH2-LG2 and Protein2-SH are coupled together obtaining a protein conjugate of Protein1-Linker-NH—C(═O)—CH2—S-Protein2  Formula I: wherein LG2 is a leaving group of low reactivity, the method comprising the steps of b) obtaining an conjugate intermediate: Protein1-Linker-NH—C(═O)—CH2-LG2 c) performing a leaving group exchange reaction increasing the reactivity of LG2. d) reacting the intermediate obtained by c) with Protein2-SH e) obtaining the protein conjugate. The leaving group exchange reaction may be performed as an aqueous Finkelstein halogen exchange reaction whereby the reactivity of the leaving group is increased. In one embodiment LG2 is C1. In such an embodiment —NH—C(═O)—CH2—Cl is transformed to —NH—C(═O)—CH2—I in step c) which is subsequently reacted with Protein2-SH to obtain the protein conjugate. In this way the first intermediate can be prepared with LG2 in a rather in-active form. In one embodiment LG1 is Br and LG2 is C1. As Br-acetamide is more reactive than Cl-acetamide the leaving group will determine the order of conjugation and the subsequent activation changing LG2 from Cl to I will ensure that the linker end of the second intermediate is reactive with Protein2. The invention also relates to coupling of more than two proteins such as three proteins exemplified herein by the GH and Fc conjugations of Example 2. In one embodiment the method is for preparation of a protein conjugate, wherein Protein1-SH and two copies of Protein2-SH are coupled together obtaining a protein conjugate of Formula IV. In one embodiment the invention relates to method for preparing a protein conjugate wherein Protein1-SH and 2×(Protein2-SH) are coupled together obtaining a protein conjugate of using a thiol reactive linker: wherein LG1 has a higher reactivity than LG2, the method comprising the steps of: a) reacting Protein1-SH with —NH—C(═O)—CH2-LG1 (R1) of the linker b) obtaining a conjugate intermediate: Protein1-S—CH2—C(═O)—NH-Linker[-NH—C(═O)—CH2-LG2]2 c) performing a leaving group exchange reaction increasing the reactivity of LG2. d) reacting the intermediate of c) with Protein2-SH e) obtaining the protein conjugate. As above the leaving group exchange reaction transforming serves to activate the 2nd and 3rd reactive end of the linker. In one embodiment LG2 is changed from Cl to I increasing the reactivity. In one embodiment LG1 is changed from Cl to I increasing the reactivity. In one embodiment the reactive ends of both R2 and R3 in the final intermediate comprises —NH—C(═O)—CH2—I. If the intermediate of b) is obtained by alternative means of if a protein linker conjugate including only thiol reactive ends in the second (and/or third arm) the method may be applied starting from step 2. b) obtaining a conjugate intermediate: Protein1-Linker[-NH—C(═O)—CH2-LG2]2 c) performing a leaving group exchange reaction increasing the reactivity of LG2 d) reacting the intermediate of c) with Protein2-SH and Protein3-SH e) obtaining the protein conjugate. In one such embodiment the method is for preparing a protein conjugate, wherein Protein1-linker and Protein2-SH are coupled together obtaining a protein conjugate of Formula III, where two copies of Protein2 is linked via sulfur atoms. The method comprising the steps of: a) obtaining an conjugate intermediate or the structure: wherein LG2 is a leaving group of low reactivity, b) performing a leaving group exchange reaction increasing the reactivity of LG2 c) reacting the intermediate of b) with Protein2-SH d) obtaining the protein conjugate. A protein linker intermediate may be used having the following structure: and as described above the LG may be a halogen that is exchanged for an alternative halogen with higher reactivity and as before Cl may be exchanged for an I that is highly reactive towards Cys residues. Based on the above schemes the reactivity of the reactive ends is important and in one embodiment LG1 has a higher reactivity than LG2. In an embodiment LG1 is Br or I. In one embodiment LG2 is Cl. In one embodiment the LG2 is Cl and is exchange to I. The exchange reaction may be performed as an aqueous halogen exchange, such as an aqueous Finkelstein reaction. The reaction may be performed in an aqueous KI solution. The solution may further comprise ascorbic acid. In one embodiment the reaction is performed in the presence of 0.1-5 M KI and 10-50 mM ascorbic acid. The duration of the various steps of the conjugation method may be adjusted for the individual proteins to be conjugated. The reactions steps with Protein1 may be performed for 1-24 hours, such as overnight. The reactions steps with Protein2 may be performed for 1-24 hours, such as overnight. The methods above may be performed using linkers described herein above, such as two-armed or three-armed linkers as appropriate. In all examples provide herein the reaction between the haloacetamide with the reduced free cysteine results in formation of a thioether (—CH2—S—CH2—) which connects the linker structure with the polypeptide. The —CH2— groups of the thioether may be considered part of the linker and/or protein, but may be included in the structure to illustrate the identity of the linkage. Intermediates Based on the overall set out of the method described above a series of products and intermediates as described are part of the presents invention. It is clear from the above that the protein conjugations can in many situations be performed in the reverse order. In the present overview it is contemplated that Protein1 is the first protein to be conjugated. The two different orders of conjugation are also illustrated in example 3 of the application. Protein Conjugates Protein1-Linker-S-Protein2 Protein1-Linker-CH2—S—CH2-Protein2 Protein1-Linker-NH—C(═O)—CH2—S-Protein2 Protein1-S-Linker-NH—C(═O)—CH2—S-Protein2 Protein1-S—CH2—C(═O)—NH-Linker-NH—C(═O)—CH2—S-Protein2 Protein1-linker intermediates Protein1-Linker-Cl/Br/I Protein1-Linker-NH—C(═O)—CH2—Cl/Br/I Protein1-S-Linker-NH—C(═O)—CH2—Cl/Br/I Protein1-CH2—S—CH2-Linker-NH—C(═O)—CH2—Cl/Br/I Protein1-S—CH2—C(═O)—NH-Linker-NH—C(═O)—CH2—Cl/Br/I Protein2-linker intermediates I/Br/Cl-Linker-Protein2 I/Br/Cl—CH2—C(═O)—NH-Linker-Protein2 I/Br/Cl—CH2—C(═O)—NH-Linker-S-Protein2 I/Br/Cl—CH2—C(═O)—NH-Linker-NH—C(═O)—CH2-Protein2 In one embodiment the intermediate is a structure with two Fc polypeptides individually linked to two arms of the linker, while the 1st arm is used for conjugation with a different protein/peptide. The intermediate according to the invention may thus by a trivalent linker structure including Fc polypeptides linked to arm 2 and 3, while arm 1 is still free. In the alternative the intermediate is a protein conjugate to arm 1 of the trivalent linker which is thus suited for conjugation to the Fc polypeptides via arm 2 and arm 3. The overall structure of the protein conjugate including two Fc polypeptides, independent of the reactive end of the linkers, is In an embodiment the conjugate includes thiol linkages of Fc medicated by a halo-acetamide leaving group providing the structure —NH—C(═O)—CH2— inserted in the protein conjugate: Details with regards to the trivalent linker have been provided elsewhere in the application and can be read into the structures above. While certain features of the invention have been described herein and illustrated in the subsequence examples, many modifications, substitutions, changes, and equivalents will now occur to those of ordinary skill in the art. It is, therefore, to be understood that the appended embodiment and claims are intended to cover all such modifications and changes as fall within the true spirit of the invention. EMBODIMENTS 1. A protein conjugate of the following structure wherein Linker is a chemical moiety S is a sulfur atom and Protein1 is covalently linked to Protein2 and Protein3 via the linker and sulfur atoms. 2. The protein conjugate of embodiment 1, wherein the conjugate has the following structure 3. The protein conjugate of embodiment 1, wherein the conjugate has the following structure wherein U represents a central unit, RR1 a reactive end radical and S1, S2 and S3 represent individual spacers. 4. The conjugate according to any of the previous embodiment, wherein the sulfur atom's (—S—) are part of thioethers (—CH2—S—CH2—). 5. The conjugate according to any of the previous embodiments, wherein U comprises or consists of a nitrogen atom. 6. The conjugate according to any of the previous embodiments, wherein U comprises or consists of a benzene ring structure. 7. The conjugate according to any of the previous embodiments, wherein Protein2 and Protein3 are Fc polypeptides. 8. The conjugate according to any of the previous embodiments, wherein the conjugate has the structure: 9. The conjugate according to any of the previous embodiments, wherein the conjugate has the structure: Protein1-RR1-S1-U-[S2-NH—C(═O)—CH2—S-Fc]2 wherein RR1 a reactive end radical, S1 and S2 represent individual spacers, U represents a central unit and Fc is an Fc polypeptide. 10. The conjugate according to any of the previous embodiments, wherein Protein1 is a growth hormone. 11. The conjugate according to any of the previous embodiments, wherein the conjugate has the structure: wherein GH represents a growth hormone molecule. 12. The conjugate according to any of the previous embodiments, wherein the conjugate has the structure: GH-RR1-S1-U-[S2-NH—C(═O)—CH2—S-Fc]2 wherein GH represents a growth hormone molecule, RR1 a reactive end radical S1 and S2 represent individual spacers U represents a central unit and Fc is an Fc polypeptide. 13. The conjugate according to embodiments 7 to 12, wherein the Fc polypeptides is derived from IgG, such are IgG1, IgG2, IgG3 or IgG4. 14. The conjugate according to any of embodiments 7-13, wherein the Fc polypeptides comprise a hinge region. 15. The conjugate according to any of embodiments 7-14, wherein the hinge region of each Fc polypeptide includes a Cys residue. 16. The conjugate according to any of embodiments 7-15, wherein the hinge region of the Fc polypeptide is selected from the group of sequences consisting of: an IgG1 derived hinge sequence and an IgG4 derived sequence. 17. The conjugate according to embodiment 16, wherein the IgG1 derived hinge sequence is selected from: PKSCDKTHTCPPCP, PPCP, PCP and CP. 18. The conjugate according to embodiment 16, wherein the IgG4 derived hinge sequence is selected from: SKYGPPCPSCP, PSCP, SCPL and CP. 19. The conjugate according to any of the previous embodiments of the following structure 20. The conjugate according to any of the previous embodiments wherein the conjugate has the structure: 21. The conjugate according to any of the previous embodiments, wherein the conjugate has the structure: 22. The conjugate according to any of the previous embodiments, wherein the sulfur atoms (—S—) are derived from protein thiols. 23. The conjugate according to any of the previous embodiments, wherein the sulfur atoms (—S—) are derived from protein cysteines. 24. The conjugate according to any of the previous embodiments, wherein the sulfur atoms (—S—) are derived from free Cys's. 25. The conjugate according to any of the previous embodiments, wherein one or more of the protein cysteine's is/are wild type residue(s). 26. The conjugate according to any of the previous embodiments, wherein one or more of the protein cysteines is/are variant amino acid residue(s). 27. The conjugate according to any of the previous embodiments, wherein the —S— linking Protein2 and Protein3 with the linker are from wild type cysteines. 28. The conjugate according to any of the previous embodiments 19-27, wherein the —S— linking Protein1 with the linker is derived from a variant cysteine. 29. The conjugate according to any of the previous embodiments 19-28, wherein the —S— linking Protein1 is derived from a free Cys. 30. The conjugate according to any of the previous embodiments 19-29, wherein the —S— linking Protein1 is derived from a free cysteine in a growth hormone variant selected from the group consisting of T3C, P5C, S7C, D11C, H18C, Q29C, E30C, E33C, A34C, Y35C, K38C, E39C, Y42C, S43C, D47C, P48C, 555C, 557C, P59C, S62, E65C, Q69C, E88C, Q91C, 595C, A98C, N99C, S100C, L101C, V102C, Y103C, D107C, S108C, D112C, Q122C, G126C, E129C, D130C, G131C, P133C, T135C, G136C, T142C, D147C, N149C, D154C, A155C, L156C, R178C, E186C, G187C and G190C. 31. The conjugate according to any of the previous embodiments 19-30, wherein the —S— linking Protein1 is derived from a free cysteine in a growth hormone variant selected from the group consisting of A98C, N99C, L101C, V102C and S108C. 32. The conjugate according to any of the previous embodiments 19-31, wherein the —S— linking Protein1 is derived from a Cys substitution located within AA 93-106 in a growth hormone variant. 33. A trivalent linker of the structure: wherein U represent a central unit, S1, S2 and S3 represent individual spacers and R1, R2 and R3 individually represent a reactive end. 34. The linker according to embodiment 33, wherein R1, R2 and R3 are not identical. 35. The linker according to any of the previous embodiments 33-34, wherein R2 and R3 are identical. 36. The linker according to any of the previous embodiments 33-35, wherein R2 and R3 are identical but R1 is different. 37. The linker according to any of the previous embodiments 33-36, wherein R2 and R3 are thiol reactive ends. 38. The linker according to any of the previous embodiment 33-37, wherein R2 and R3 each comprise a halogen leaving group, such as Bromide, Chloride or Iodide. 39. The linker according to any of the previous embodiments 33-38, wherein R2 and R3 comprise —NH—C(═O)—CH2-LG, providing a linker of the structure: wherein LG2 and LG3 are halogen leaving groups 40. The linker according to any of the previous embodiments 33-39, wherein the reactive ends R2 and R3 are identical cys reactive ends. 41. The linker according to any of the previous embodiments 33-40, wherein R1 is different from R2 and R3. 42. The linker according to any of the previous embodiments 33-41, wherein R1 has a different reactivity than R2 and R3. 43. The linker according to any of the previous embodiments 33-42, wherein R1 is a thiol reactive end. 44. The linker according to any of the previous embodiments 33-43, wherein R1 is a thiol reactive comprising a leaving group, such as Bromide, Chloride or Iodide. 45. The linker according to any of the previous embodiments 33-44, wherein the first linker arm has the structure LG1-CH2—C(═O)—S1- 46. The linker according to any of the previous embodiments 33-45, wherein R1 comprises Cl as LG1 and R2 and R3 comprises Br as LG2. 47. The linker according to any of the previous embodiments 33-46, wherein R1 comprises Br as LG1 and R2 and R3 comprises Cl as LG2. 48. The linker according to any of the previous embodiments 33-47, wherein S2 and S3 are identical. 49. The linker according to any of the previous embodiments 33-48, wherein the length of the linker is 10 to 60 atoms, such as 12-45, or such as 15-40 atoms. 50. The linker according to any of the previous embodiments 33-49, wherein S2 and S3 is a short spacer, such as —(CH2)2—. 51. The linker according to any of the previous embodiments 33-50, wherein 51 is different from S2 and S3. 52. The linker according to any of the previous embodiments 33-51, wherein 51 is an extended spacer of 10-50 atoms in length. 53. The linker according to any of the previous embodiments 33-52, wherein 51 comprise one or more spacer elements linked by peptide bond(s). 54. The linker according to any of the previous embodiments 33-53, wherein the spacer elements of 51 are selected from the group of: wherein k is an integer in the range of 1-5, and n is an integer in the range of 1-5. 55. The linker according to any of the previous embodiments 33-54, wherein a spacer element is *—NH—(CH2)2—O—(CH2)2—O—CH2—CO—* (OEG or a di-radical of 8-amino-3,6-dioxaoctanoic acid). 56. The linker according to any of the previous embodiments 33-55, wherein the linker is selected from the group consisting of: (S)-4-(2-{2-[((S)-1-{Bis-[2-(2-chloro-acetylamino)-ethyl]-carbamoyl}-3-carboxy-propylcarbamoyl)-methoxy]-ethoxy}-ethylcarbamoyl)-2-(2-bromo-acetylamino)-butyric acid 4S,18S)-4-(bis(2-(2-Chloroacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-bromoacetamido)ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid 2-(2-bromoacetamido)-N,N-bis(2-(2-chloroacetamido)ethyl)acetamide 2-(2-(2-(2-(2-bromoacetamido)ethoxy)ethoxy)acetamido)-N,N-bis(2-(2-chloroacetamido)ethyl)acetamide (13R,18S)-18-(bis(2-(2-Chloroacetamido)ethyl)carbamoyl)-1-bromo-13-carboxy-2,11,16-trioxo-6,9-dioxa-3,12,17-triazahenicosan-21-oic acid (18R,23S)-23-(Bis(2-(2-chloroacetamido)ethyl)carbamoyl)-1-bromo-18-carboxy-2,7,16,21-tetraoxo-11,14-dioxa-3,8,17,22-tetraazahexacosan-26-oic acid (R)-4-{2-[2-({Bis-[2-(2-chloro-acetylamino)-ethyl]-carbamoyl}-methoxy)-ethoxy]-thylcarbamoyl}-2-[(S)-2-(2-{2-[2-(2-bromo-acetylamino)-ethoxy]-ethoxy}-acetylamino)-4-carboxy-butyrylamino]-butyric acid (4S,18S)-4-(bis(2-(2-Bromoacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-chloroacetamido)ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid (4S)-5-[bis[3-[(2-chloroacetyl)amino]propyl]amino]-4-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-oxo-pentanoic acid (2R)-6-[bis[2-[(2-chloroacetyl)amino]ethyl]carbamoylamino]-2-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]hexanoic acid N-[2-[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]ethylcarbamoyl-[2-[(2-chloroacetyl)amino]ethyl]amino]ethyl]-2-chloro-acetamide (2R)-2-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-[2-[2-[2-[[(1S)-3-carboxy-1-[2-[(2-chloroacetyl)-[2-[(2-chloroacetyl)amino]-ethyl]amino]ethylcarbamoyl]propyl]amino]-2-oxo-ethoxy]ethoxy]ethylamino]-5-oxo-pentanoic acid 57. A method for preparation of a protein conjugate, wherein Protein1-SH, Protein2-SH and a thiol reactive linker are coupled together obtaining a protein conjugate of Formula II Protein1-S—CH2—C(═O)—NH-Linker-NH—C(═O)—CH2—S-Protein2  (Formula II) wherein the thiol reactive linker has the structure: LG1-CH2—C(═O)—NH-Linker-NH—C(═O)—CH2-LG2, wherein LG1 has a higher reactivity than LG2, the method comprising the steps of: a) reacting Protein1-SH with —NH—C(═O)—CH2-LG1 of the linker b) obtaining a conjugate intermediate: Protein1-S—CH2—C(═O)—NH-Linker-NH—C(═O)—CH2-LG2 c) performing a leaving group exchange reaction increasing the reactivity of LG2. d) reacting the intermediate of c) with Protein2-SH e) obtaining the protein conjugate. 58. A method for preparing a protein conjugate, wherein Protein1-Linker-NH—C(═O)—CH2-LG and Protein2-SH are coupled together obtaining a protein conjugate of Formula I Protein1-Linker-NH—C(═O)—CH2—S-Protein2  (Formula I) wherein LG is a leaving group of low reactivity, the method comprising the steps of b) obtaining an conjugate intermediate: Protein1-Linker-NH—C(═O)—CH2-LG c) performing a leaving group exchange reaction increasing the reactivity of LG. d) reacting the conjugate intermediate of b) with Protein2-SH e) obtaining the protein conjugate. 59. A method for preparation of a protein conjugate, wherein Protein1-linker and Protein2-SH are coupled together obtaining a protein conjugate of Formula III: the method comprising the steps of: b) obtaining an conjugate intermediate or the structure: wherein LG2 is a leaving group of low reactivity, c) performing a leaving group exchange reaction increasing the reactivity of LG2. d) reacting the intermediate of b) with Protein2-SH e) obtaining the protein conjugate. 60. A method for preparation of a protein conjugate, wherein Protein1-SH, Protein2-SH and a thiol reactive linker are coupled together obtaining a protein conjugate of Formula VI wherein the thiol reactive linker has the structure: wherein LG1 has a higher reactivity than LG2, the method comprising the steps of: a) reacting Protein1-SH with —NH—C(═O)—CH2-LG1 of the linker b) obtaining a conjugate intermediate: Protein1-S—CH2—C(═O)—NH-Linker-[NH—C(═O)—CH2-LG2]2 c) performing a leaving group exchange reaction increasing the reactivity of LG2. d) reacting the conjugate intermediate of c) with Protein2-SH e) obtaining the protein conjugate. 61. The method according to any of the above embodiments 57-60, wherein LG1 is Br. 62. The method according to any of the above embodiments 57-61, wherein LG2 is C1. 63. The method according to any of the above embodiments 57-62, wherein LG1 is C1. 64. The method according to any of the above embodiments 57-63, wherein LG2 is Br. 65. The method according to any of the above embodiments 57-64, wherein the exchange reaction is a Cl to I exchange. 66. The method according to any of the above embodiments 57-65, wherein the exchange reaction is performed in the presence of 0.1-5 M KI and 10-50 mM ascorbic acid. 67. The method according to any of the above embodiments 57-66, wherein protein-SH is reacted with a thiol reactive linker or a conjugate intermediate overnight 68. The method according to any of the above embodiments 57-67, wherein the Protein1-Linker-[NH—C(═O)—CH2—I]2 is reacted with Protein2-SH overnight. 69. The method according to any of the above embodiments 57-8, wherein an Fc domain is conjugated to Protein1 via covalent linkage of both Fc polypeptide chains. 70. The method according to any of the above embodiments 57-69, wherein a step of obtaining Protein-SH by reduction is included. EXAMPLES Abbreviations amu=Atomic mass units Boc=tert-Butyloxycarbonyl O-t-Bu=tert-Butyl ester t-Bu=tert-Butyl CDCl3=Deuterio chloroform CD3OD=Tetradeuterio methanol CV=Column volumes DMSO-d6=Hexadeuterio dimethylsulfoxide DCM=DCM, CH2Cl2, methylenechloride DIC=Diisopropylcarbdiimide DIPEA=diisopropylethylamine DMF=N,N-Dimethylformamide DMSO=Dimethylsulfoxide DTT=Dithiothreitol EDAC=1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride Et2O=Diethyl ether EtOAc=Ethyl acetate FA=Formic acid Fmoc=9H-Fluoren-9-ylmethoxycarbonyl Fmoc-Glu-O-t-Bu=N-Fmoc-glutamic acid-1-t-butyl ester Fmoc-Lys(Mtt)-OH=(S)-6-[(Diphenyl-p-tolyl-methyl)-amino]-2-(9H-fluoren-9-ylmethoxycarbo-nylamino)-hexanoic acid Fmoc-OEG-OH=(2[2-(Fmoc-amino)ethoxy]ethoxy)acetic acid Fmoc-Thx-OH=N-Fmoc-trans-4-aminomethylcyclohexancarboxylic acid H2O=Water hr(s)=Hour(s) Hz=Hertz HOBt=1-Hydroxybenzotriazole HPLC=High pressure liquid chromatography HPLC-MS=High pressure liquid chromatography—mass spectrometry i.v.=Intravenous L=Liter(s) M=Molar mbar=Millibar mg=Milligram(s) min.=Minute(s) mL=Milliliter(s) mM=Millimolar mol=Mole(s) mmol=Millimole(s) m/z=Mass to charge ratio MS=Mass spectrometry MeCN=Acetonitrile MeOH=Methanol μL=Microliters N=Normal nm=Nanometer(s) nmol=Nanomole(s) NaCl=Sodium chloride NaOH=Sodium hydroxide NMR=Nuclear magnetic resonance spectroscopy OEG=(2[2-(Amino)ethoxy]ethoxy)acetyl ppm=Parts per million PyBrOP=Bromo-tris-pyrrolidino-phosphonium hexafluorophosphate p.o.=Per oral RP=Reverse phase rt or RT=Room temperature tr or Rt=Retention time sec=Second(s) s.c.=Subcutaneous TCTU=O-(6-Chloro-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate TEA=Triethylamine TFA=Trifuloroacetic acid THF=Tetrahydrofuran TIS=Triisopropylsilane TSTU=O—(N-Succinimidyl)-1,1,3,3-tetramethyl uranium tetrafluoroborate HATU=1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate TCEP=Tris(2-carboxyethyl)phosphine TPPDS=Bis(p-sulfonatophenyl)phenylphosphine TPPTS=Tris((m-sulfonatophenyl)phenylphosphine Methods Method 1—Method for Preparation and Analysis of a Growth Hormone Protein The gene coding for the growth hormone or growth hormone variant was inserted recombinant into a plasmid vector. A suitable E. coli strain was subsequently transformed using the plasmid vector. hGH or GH variants may be expressed with an N-terminal methionine or as a MEAE fusion from which the MEAE sequence is subsequently cleaved off. Cell stock was prepared in 25% glycerol and stored at −80° C. Glycerol stock strain was inoculated into LB plates and subsequently incubated at 37° C. overnight. The content of each plate was washed with LB medium and diluted into 500 mL LB medium for expression. The cultures were incubated at 37° C. with shaking at 220 rpm until OD600 0.6 was reached. Succeeding induction was performed using 0.2 mM IPTG at 25° C. for 16 hrs. Cells were finally harvested by centrifugation. Cells were subsequently suspended in 10 mM Tris-HCl, pH 9.0 containing 0.05% Tween 20, 2.5 mM EDTA, 10 mM cystamine and 4M urea, and disrupted using a cell disrupter at 30 kPSI. The supernatant was collected by centrifugation and subsequently subjected to chromatographic purification. The purification was performed using ion-exchange chromatography and hydrophobic interaction, followed by removal of the peptide tag using human dipeptidyl peptidase I (hDPPI) expressed from CHO cell. Final purification was achieved by isoprecipitation and ion-exchange chromatography. The purification could also be achieved by using but not limited to ion-exchange chromatography, hydrophobic interaction chromatography, affinity chromatography, size exclusion chromatography and membrane based separation techniques known to a person skilled in the art. Characterization of Growth Hormone Preparation The intact purified protein was analysed using MALDI-MS. The observed mass corresponded to the theoretical mass deduced from the amino acid sequence. The expected linkage disulfide bonds may be demonstrated by peptide mapping using trypsin and AspN digestion followed by MALDI-MS analysis of the digest before and after reduction of the disulfide bonds with DTT. Proteolytic Digestion: 100 μL of test compound solution at 1 mg/mL in ammonium bicarbonate buffer is degraded by enzyme for up till 24 hrs at 37° C. Sub-samples are taken to various time points and the proteolytic reaction is stopped by acidifying the sample by 10 times dilution into 1% TFA. These diluted samples are analysed by reversed phase HPLC to estimate the degree of proteolytic digestion. HPLC Method: 10 μL of the above solution is injected on a reversed phase Vydac C4 2×150 mm column eluted with a linear gradient from 0.1% TFA in water to 100% MeCN containing 0.1% TFA over a period of 30 min at a flow rate of 0.2 mL/min. Detection of peaks is performed at 214 nm UV absorption. Percentage (%) intact compound at time point t=T is calculated from the peak area at time point t=T (AT) and the peak area at t=0 (A0) as (AT/A0)×100%. Percentage (%) intact compound is plotted against time using GraphPad Prims software ver. 5.01. Half-life (T1/2) is calculated as one phase decay also by GraphPad Prism software. Examples of enzymes that may be used are elastase (Sigma from porcine pancrease) and chymotrypsin (Roche sequencing grade). Example of buffer is 50 mM ammonium bicarbonate, pH=8.5. Capillary Electrophoresis: Capillary electrophoresis was carried out using an Agilent Technologies 3DCE system (Agilent Technologies). Data acquisition and signal processing were performed using Agilent Technologies 3DCE ChemStation. The capillary was a 64.5 cm (56.0 cm efficient length) 50 μm i.d. “Extended Light Path Capillary” from Agilent. UV detection was performed at 200 nm (16 nm Bw, Reference 380 nm and 50 nm Bw). The running electrolyte was phosphate buffer 50 mM pH 7 (method A). The capillary was conditioned with 0.1M NaOH for 3 min, then with Milli-Q water for 2 min and with the electrolyte for 3 min. After each run, the capillary was flushed with milli-Q water for 2 min, then with phosphoric acid for 2 min, and with milli-Q water for 2 min. The hydrodynamic injection was done at 50 mbar for 4.0 sec. The voltage was +25 kV. The capillary temperature was 30° C. and the runtime was 10.5 min. Maldi-Tof Mass Spectrometry: Molecular weights were determined using the Autoflex Maldi-Tof instrument (Bruker). Samples were prepared using alfa-cyano-4-hydroxy-cinnamic acid as matrix. RP-HPLC: RP-HPLC analysis was performed on an Agilent 1100 system using a Vydac 218TP54 4.6 mm×250 mm 5 μm C-18 silica column (The Separations Group, Hesperia). Detection was by UV at 214 nm, 254 nm, 280 nm and 301 nm. The column was equilibrated with 0.1% TFA/H2O and the sample was eluted by a suitable gradient of 0 to 90% MeCN against 0.1% TFA/H2O. LC-MS: LC-MS analysis was performed on a PE-Sciex API 100 or 150 mass spectrometer equipped with two Perkin Elmer Series 200 Micropumps, a Perkin Elmer Series 200 auto-sampler, an Applied Biosystems 785A UV detector and a Sedex 75 Evaporative Light scattering detector. A Waters Xterra 3.0 mm×50 mm 5p C-18 silica column was eluted at 1.5 mL/min at room temperature. It was equilibrated with 5% MeCN/0.1% TFA/H2O and eluted for 1.0 min with 5% MeCN/0.1% TFA/H2O and then with a linear gradient to 90% MeCN/0.1% TFA/H2O over 7 min. Detection was by UV detection at 214 nm and Evaporative light Scattering. A fraction of the column elute was introduced into the ionspray interface of a PE-Sciex API 100 mass spectrometer. The mass range 300-2000 amu was scanned every 2 seconds during the run. Quantification of Protein: Protein concentrations were estimated by measuring absorbance at 280 nm using a NanoDrop ND-1000 UV-spectrophotometer. Enzymatic Peptide Mappinq for Determination of Site(s) of Derivatization: Peptide mapping was performed using Asp-N digestion of the reduced and alkylated protein. First the protein was treated with DTT and iodoacetamide according to standard procedures. The alkylated product was purified using HPLC. Subsequently the alkylated purified product was digested overnight with endoprotease Asp-N (Boehringer) at an enzyme:substrate ratio of 1:100. The digest was HPLC separated using a C-18 column and standard TFA/MeCN buffer system. The resulting peptide map was compared to that of un-derivatized hGH and fractions with different retention times were collected and further analysed using Maldi-tof mass spectrometry. SDS Page: SDS poly-acrylamide gel electrophoresis was performed using NuPAGE 4%-12% Bis-Tris gels (Invitrogen NPO321BOX). The gels were silver stained (Invitrogen LC6100) or Coomassie stained (Invitrogen LC6065) and where relevant also stained for PEG with barium iodide as described by M. M. Kurfurst in Anal. Biochem. 200(2), 244-248, (1992). Protein Chromatography: Protein chromatography was performed on an Äkta Explorer chromatographic system and columns from GE Health Care. Anion exchange was done using a Q-Sepharose HP 26/10 column. Starting buffer was 20 mM triethanolamine buffer pH 8.5 and eluting buffer was starting buffer+0.2 M NaCl. The compounds were typically eluted with a gradient of 0-75% eluting buffer over 15 column volumes. De-salting and buffer exchange was performed using a HiPrep 26/10 column. Method 2—Method for Preparation of an Fc-Domain Fc domains may be expressed by technologies known in the art such as by expression in E. coli (WO05047334, WO05047335, WO05047336, WO05047337, and WO05001025) or in mammalian cells such as HEC (Farge, F. et. al, Journal of Chromatography (1976) vol 123, page 247-250). The following overall method has been applied for the present application. An Fc-domain was obtained using a fragment of human IgG4, which was truncated at the N-terminal of the hinge region. The coding region including a Met start codon was inserted in a pET11d derived vector to guide expression of a Fc polypeptide with MPSCPAPEFLGGPSVF . . . N-terminal. The Fc polypeptide was expressed in E. coli. The strain used was (BL21(DE3)_TKO::ybhE as described in WO2010052335 additionally including an ybhE knock-in. The Fc domain was subsequently purified. An initiator ATG (Met-codon) was included in-frame with the truncated hinge allowing expression in E. coli. Due to host enzymes this Methionine was removed and not present in the purified Fc which thus have a proline at the N-terminal. An expression level of above 5 g/L of soluble Fc fragment from the cytoplasm of E. coli was obtained using defined medium. After purification a yield of 1.4 g/L was obtained. An in vitro disulfide bridge formation step was included to ensure correct folding of the Fc domain. E. coli cells were cultivated at 37° C. in defined medium to an optical density (OD600) of about 80 in 20-L fermentor. Then the culture was induced with 0.2 mM IPTG and continue to cultivate at 25° C. for overnight. Finally the cells were harvested by centrifugation. After the homogenization of the cell pellet in buffer containing Tris-HCl 50 mM, NaCl 300 mM, EDTA 5 mM and DTT 1 mM, pH 7.4, the target protein was recovered by treatment with 0.2% PEI (polyethyleneimine) for 30 min followed by centrifugation at 6,000×g. The Fc was purified from the supernatant of the cell lysate by affinity chromatography using MabSelect SuR (GE Healthcare Life Sciences), and then oxidized by adding urea 3.5 M, cystamine 0.01 mM, pH 8.5 at room temperature for overnight. Finally, the formed Fc dimer was further purified by ion-exchange chromatography using Q Sepharose HP (GE Healthcare Life Sciences) at pH 8.5. The final protein is in TEA (Tris-acetate-EDTA) 20 mM, NaCl 500 mM, pH 8.0. Method 4—Method for Preparation of Protein Conjugates—GH First Chemistry The conjugation method can be performed with a variety of suitable proteins comprising suitable attachment points, here exemplified using a GH variant and an Fc domain all including one or more sulfur atom(s) that serves as connector to the linker. The conjugate, GH-A-B-Protein (IX) is prepared as illustrated below: Reaction where linker is first attached to hGH and subsequently to Fc. The cysteine residue in (I) is optionally protected as a mixed disulfide (GH-S-S-R) with R being a small organic moiety. Non limited examples of mixed disulfides may include disulfides between cysteamine (R=—CH2CH2NH2); cysteine (R=—CH2CH(C(═O)OH)NH2); homocysteine (R=—CH2CH2CH(C(═O)OH)NH2); and glutathione (R=—CH2CH(C(═O)NH—CH2C(═O)OH)NH—C(═O)CH2CH2CH(C(═O)OH)NH2). The conjugation process utilise a trivalent linker LG1-A-B-(LG2)2 (III) wherein LG1 and LG2 independently represent inorganic leaving groups such as —Cl, —Br, —I and/or organic leaving groups such as mesylate or tosylate. Conjugation of reduced GH (II) with the linker LG1-A-B-(LG2)n (III) occurs via nucleophilic substitution (II+III→IV). Selectivity for LG1 versus LG2 is obtained via utilization of difference in leaving group ability between LG1 and LG2. In order for LG2 to act as a proper leaving group in the next step, it is changed into iodo (VI) via an aqueous Finkelstein reaction with potassium iodine. This conjugate intermediate (VI) is next treated with a protein of interest (VIII) here a Fc-domain wherein a disulphide bond selectively has been reduced (VII→VIII) using as suitable reducing agent such as dithiothreitol (DTT), TCEP, TPPTS, and TPPDS affording the GH-A-B-Protein conjugate (IX). The steps of the reaction may be described as follows starting from a GH compound (I) having an internal free Cys, a trivalent linker (III) and a Fc domain including a reducible disulfide bond. 1) Optionally liberating a free Cys GH (II) via reduction of mixed disulfide (I) with a suitable selective reducing agent 2) Alkylating a free Cys GH (II) with an trivalent linker (III) affording a Cys conjugated GH protein linker intermediate (IV) 3) Activating leaving groups LG2 in the intermediate (IV) via an aqueous Finkelstein iodine exchange reaction (V) affording activated Cys GH conjugate intermediate (VI) 4) Liberating free cysteines in an Fc-domain (VII) via selective reduction of a disulfide bridge with a suitable selective reducing agent affording (VIII) 5) Coupling of Fc-domain (VIII) with activated Cys GH conjugate intermediate (VI) affording a Cys conjugated GH-Fc conjugate (IX) Method 5—Method for Preparation of Protein Conjugates—Fc First In an alternative the conjugate GH-A-B-Protein (IX) is prepared as illustrated below: Reaction where linker is first attached to Fc subsequently to GH. Wherein the cysteine residue in (I) optionally is protected as a mixed disulfide (GH-S-S-R) with R being a small organic moiety. Non limited examples of mixed disulphides may include disulfides between cysteamine (R=—CH2CH2NH2); cysteine (R=—CH2CH(C(═O)OH)NH2); homocysteine (R=—CH2CH2CH(C(═O)OH)NH2); and glutathione (R=—CH2CH(C(═O)NH—CH2C(═O)OH)NH—C(═O)CH2CH2CH(C(═O)OH)NH2). The conjugation process utilise a trivalent linker LG1-A-B-(LG2)2 (III) wherein LG1 and LG2 independently represent inorganic leaving groups such as —Cl, —Br, —I and/or organic leaving groups such as mesylate or tosylate. Linker (III) is conjugated via nucleophilic substitution to reduced Protein (VIII) e.g. a Fc-domain obtained from (VII) via selective reduction of a disulphide bond (VII→VIII) using (DTT, TCEP, TPPTS, and TPPDS, or other reducing agents) affording LG1-A-B-Protein conjugate (X). Selectivity for LG1 versus LG2 is obtained via utilization of difference in leaving group ability between LG1 and LG2. In order for LG1 in compound (X) to act as a proper leaving group for the next coupling step, it is changed into iodo (XI) via an aqueous Finkelstein reaction with potassium iodine. Compound (XI) is then coupled with reduced GH (II) affording GH-A-B-Protein conjugate (IX). The steps of the reaction may be described as follows starting from a GH compound (I) having an internal free Cys, a trivalent linker (III) and a Fc domain including a reducible disulfide bond. 1) Liberating free cysteines in an Fc-domain (VII) via selective reduction of a disulfide bridge with a suitable selective reducing agent affording (VIII) 2) Alkylation of the Fc-domain (VIII) with a trivalent linker (III) affording an LG1-A-B-Fc conjugate intermediate (X) 3) Optionally liberating free Cys GH (II) via reduction of a mixed disulfide (I) with a suitable selective reducing agent 4) Activating leaving group LG1 of intermediate (X) via an aqueous Finkelstein iodine exchange reaction (V) affording activated conjugate intermediate (XI) 5) Coupling of a free Cys GH (II) with an the activated conjugate intermediate (XI) affording a Cys conjugated GH-Fc-compound (IX) Assays Assay 1—GH Receptor Binding Assay Receptor interaction of GH compounds is analysed using surface plasmon resonance (SPR) analysis. The method is general for the GH compounds. The interaction of hGH and GH compounds with the hGH receptor via site 1 was studied by surface plasmon resonance using a Biacore T100 instrument (GE Healthcare, Sweden). Anti-hGH mAb (Fitzgerald Industries International, USA, #10G05B) was immobilized onto a CM-5 chip according to manufacturer's instruction at a level of typically 5000 RU. hGH or GH compounds are captured at 10-25 μg/mL in running buffer (10 mM HEPES, 0.15 M NaCl, 30 mM EDTA, 0.05% Surfactant P20, pH 7.4), which resulted in 250-400 RU captured ligand. hGHR at a concentration of 0-800 nmol was subsequently injected over the surface at 30 mL/min. A surface with immobilized anti-hGH mAb but without captured hGH was used as reference. Kinetic data is analyzed with Biacore™ Evaluation Software 2.0 with the 1:1 Langmuir binding model. Assay 2—BAF-3GHR Assay to Determine Growth Hormone Activity The biological activity of hGH compounds is measured in a cell based receptor potency proliferation assay, namely a BAF assay. The BAF-3 cells (a murine pro-B lymphoid cell line derived from the bone marrow) was originally IL-3 dependent for growth and survival. IL-3 activates JAK-2 and STAT which are the same mediators GH is activating upon stimulation. After transfection of the human growth hormone receptor the cell line was turn into a growth hormone-dependent cell line. This clone can be used to evaluate the effect of different growth hormone samples on the survival of the BAF-3GHR. The BAF-3GHR cells are grown in starvation medium (culture medium without growth hormone) for 24 hrs at 37° C., 5% C02. The cells are washed and re-suspended in starvation medium and seeded in plates. 10 μL of human growth hormone and the growth hormone compound to be tested is used in different concentrations, and the plates are incubated for 68 hrs at 37° C., 5% C02. AlamarBlue® is added to each well and the cells are then incubated for another 4 hrs. The AlamarBlue® is a redox indicator, and is reduced by reactions innate to cellular metabolism and, therefore, provides an indirect measure of viable cell number. Finally, the metabolic activity of the cells is measure in a fluorescence plate reader. The absorbance in the samples is expressed in % of cells not stimulated with growth hormone compound or control and from the concentration-response curves the activity (amount of a compound that stimulates the cells with 50%) can be calculated. Assay 3: Assay for Evaluating Pharmacokinetics Parameters of Growth Hormone Compounds in Normal Rats) The pharmacokinetic of the compounds of the examples is investigated in male Sprague Dawley rats after intravenous (iv.) single dose administration. Test compounds are diluted to a final concentration of 150 nmol/mL in a dilution buffer consisting of: Glycine 20 mg/mL, mannitol 2 mg/mL, NaHCO32.5 mg/mL, pH adjusted to 8.2. The test compounds are studied in male Sprague Dawley rats weighing approximately 250 g. The test compounds are administered as a single injection either iv. in the tail vein with a 27 G needle at a predetermined dose such as of 15 nmol/rat in volume of 0.1 mL (concentration 150 nmol/mL) or approximately 60 nmol/kg body weight. For each test compound blood sampling is conducted according to the following schedule: Time (h) Animal Predose 0.08 0.5 1 2 4 6 8 18 24 48 72 96 168 240 336 1 X X X X 2 X X X X 3 X X X X 4 X X X X 5 X X X X 6 X X X X 7 X X X X 8 X X X X 9 X X X X 10 X X X X 11 X X X X 12 X X X X At each sampling time 200 μL blood is drawn from the tail vein or the sublingual plexus using a 25 G needle. The blood is sampled into an EDTA coated test tube and stored on ice until centrifugation at 1200×G for 10 min at 4° C. Two times 50 μL plasma is transferred to two separate Micronic tubes and stored at −20° C. until analysis. Test substance concentrations will be determined by Luminescence Oxygen Channeling Immunoassay (LOCI), which is a homogenous bead based assay. LOCI reagents include two latex bead reagents and biotinylated GH binding protein, which is one part of the sandwich. One of the bead reagents is a generic reagent (donor beads) and is coated with streptavidin and contains a photosensitive dye. The second bead reagent (acceptor beads) is coated with an antibody making up the sandwich. During the assay the three reactants combine with analyte to form a bead-aggregate-immune complex. Illumination of the complex releases singlet oxygen from the donor beads which channels into the acceptor beads and triggers chemiluminescence which is measured in the EnVision plate reader. The amount of light generated is proportional to the concentration of hGH derivative. 2 μL 40× in LOCI buffer diluted sample/calibrator/control is applied in 384-well LOCI plates. 15 μL of a mixture of biotinylated GH binding protein and mAb M94169 anti-(hGH) conjugated acceptor-beads is added to each well (21-22° C.). The plates are incubated for 1 hour at 21-22° C. 30 μL streptavidin coated donor-beads (67 μg/mL) is added to each well and all is incubated for 30 minutes at 21-22° C. The plates are read in an Envision plate reader at 21-22° C. with a filter having a bandwidth of 520-645 nmol after excitation by a 680 nmol laser. The total measurement time per well is 210 ms including a 70 ms excitation time. The limit of detection for growth hormone compounds is 50 pM. A non-compartmental pharmacokinetic analysis is performed on mean concentration-time profiles of each test compound using WinNonlin Professional (Pharsight Inc., Mountain View, Calif., USA). The pharmacokinetic parameter estimates of terminal half-life (T1/2) and mean residence time (MRT) are calculated. IGF-1 Plasma concentration-time profiles are generated for each compound. Assay 4: Assay for Evaluating the In Vivo Response of Growth Hormone Compounds in Hypophysectomised Sprague Dawley Rats. The in vivo response is studied in hypophysectomised male Sprague Dawley rats. The hypophysectomised rat is a well-known and recognised animal model of growth hormone deficiency, where no production of growth hormone occurs after the surgical removal of the pituitary gland. This also leads to low circulating levels of insulin-like growth factor-1 (IGF-1) another important clinical feature of growth hormone deficiency in humans. The hypophysectomy is usually performed on 4 week old male rats weighing 90-100 g. The animals entering the study 3-4 weeks after the surgery weighing 100-110 g. Animals with a body weight gain of more than 10% during the 3-4 weeks after surgery are not allowed to enter the study. Hypophysectomy Procedure Anaesthesia and Pre-Operative Analgesia The rats are anaesthetised with fentanyl-fluanisone (Hypnorm 0.315 mg fentanyl and 10 mg fluanisone per mL) and midazolam (Midazolam Accord 5 mg midazolam per ml). The rats are dosed i.p. 2 mL/kg with a mixture of fentanyl-fluanisone and midazolam diluted in sterile water. The resulting mixture contains 0.079 mg fentanyl, 2.5 mg fluanisone and 1.25 mg midazolam per mL. Surgical Procedure The rats are prepared for aseptic surgery. The rats are mounted in the Hoffman-Reiter stereotactic device designed for the hypophysectomy procedure. An 18G needle on a glass syringe is introduced into the right ear of the rat. During a rotating movement the needle passes through the tympanic membrane, middle ear and temporal bone. From this position the pituitary gland is aspirated. The rat is dismounted from the stereotactic device and transferred to a thermo plate for recovery. When the rat recovers it will be transferred to its cage. Post-Operative Analgesia and Care Before recovery the rat is treated with carprofen sc. (Rimadyl 50 mg carprofen per mL) 1 mL/kg with a solution containing 5 mg carprofen per mL diluted in sterile water. Post-operative analgesia is sustained for 2 days after surgery by adding 0.05 mg carprofen per ml to a 5% dextrose solution which is provided to the rat instead of drinking water. After the first 2 days post-surgery the rat will be provided with at 5% dextrose solution as drinking water for up to 10-14 days post-surgery. Hypophysectomised Sprague Dawley rats were randomly allocated to different dosing groups with ten animals in each group. One group received vehicle only and served as a control group. In all test groups each animal received a single sc. dose of 1, 5, 15, 50 and 150 nmol test compound respectively. The body weight was measured daily during the study between 8-10 am. Blood sampling for exposure and IGF-1 measurements were conducted at day 0, 1, 3, 5, 7, 10 and 14 between 8-10 am. At each sampling time 200 μL blood is drawn from the tail vein or the sublingual plexus using a 25 G needle. The blood is sampled into an EDTA coated test tube and stored on ice until centrifugation at 1200×G for 10 min at 4° C. 50 μL plasma is transferred to a Micronic tube and stored at −20° C. until analysis. IGF-1 concentration-time profiles are generated for each compound. Assay 5: Assay for Detecting IGF Response in Rats. The plasma IGF-1 concentrations is determined by a commercial ELISA assay (Commercial assay from Immunodiagnostic Systems Ltd. Octeia Rat/Mouse IGF-1, Cat. no. AC-18F1 IDS Ltd., England). The assay is a sandwich ELISA using a highly IGF-1 specific polyclonal antibody as catcher, and a horseradish peroxidase labelled high affinity monoclonal antibody as detector. The assay lower limit of detection is 63 ng/mL. IGF-1 plasma concentration-time profiles are generated for each compound together with baseline corrected IGF-1 plasma concentration-time profiles. The time and extent the baseline corrected profile is above zero is used as a measure for the compound efficacy. Assay 6: Assay for Evaluating Pharmacokinetics Parameters of Growth Hormone Compounds in Minipigs. The pharmacokinetic of the compounds of the examples is investigated in female Göttingen minipigs after subcutaneous (sc.) single dose administration. Test compounds are diluted to a final concentration of 15 mg/mL in a dilution buffer consisting of: Glycine 20 mg/mL, mannitol 2 mg/mL, NaHCO32.5 mg/mL, pH adjusted to 8.2. The test compounds are studied in female Göttingen minipigs weighing approximately 10-12 kg. The test compounds were administered as a single subcutaneous injection on the right side of the neck, approximately 5-7 cm from the ear and 7-9 cm from the middle of the neck. The injections were given with a stopper on the 21 G needle, allowing 0.5 cm of the needle to be introduced. Each animal received a dose of 20 nmol/kg in a dosing volume of 0.1 mL/kg. For each test compound blood sampling was conducted from each animal according to the following schedule: Predose, 1, 4, 12, 24, 36, 48, 72, 96, 168, 240, 336, 504, 672, 840, and 1008 hours after dosing. Blood samples of 2 mL were collected from unanaesthetised minipigs by use of Vacutainers inserted in V. Jugularis into EDTA tubes. Immediately after blood collection the tubes were inverted gently in order to ensure sufficient mixing. The blood was kept on ice for max. 10 min before centrifugation at 1500 g for ten min at 4° C. Two hundred μL plasma was pipetted into Micronic tubes for compound concentration determination, and 200 μL plasma was be pipetted into Micronic tubes for IGF-1 determination. The plasma samples were stored at −20° C. until analysis. Test substance concentrations were determined by Luminescence Oxygen Channeling Immunoassay (LOCI), which is a homogenous bead based assay. LOCI reagents include two latex bead reagents and biotinylated GH binding protein, which is one part of the sandwich. One of the bead reagents is a generic reagent (donor beads) and is coated with streptavidin and contains a photosensitive dye. The second bead reagent (acceptor beads) is coated with an antibody making up the sandwich. During the assay the three reactants combine with analyte to form a bead-aggregate-immune complex. Illumination of the complex releases singlet oxygen from the donor beads which channels into the acceptor beads and triggers chemiluminescence which is measured in the EnVision plate reader. The amount of light generated is proportional to the concentration of GH derivative. 2 μL 40× in LOCI buffer diluted sample/calibrator/control is applied in 384-well LOCI plates. 15 μL of a mixture of biotinylated GH binding protein and mAb M94169 anti-(hGH) conjugated acceptor-beads is added to each well (21-22° C.). The plates are incubated for 1 hr at 21-22° C. 30 μL streptavidin coated donor-beads (67 μg/mL) is added to each well and all is incubated for 30 min at 21-22° C. The plates are read in an Envision plate reader at 21-22° C. with a filter having a bandwidth of 520-645 nmol after excitation by a 680 nmol laser. The total measurement time per well is 210 ms including a 70 ms excitation time. The limit of detection for growth hormone compounds is 50 pM. A non-compartmental pharmacokinetic analysis was performed on mean concentration-time profiles of each test compound using WinNonlin Professional (Pharsight Inc., Mountain View, Calif., USA). The pharmacokinetic parameter estimates of terminal half-life (T1/2) and mean residence time (MRT) were calculated. Example 1 Trivalent Linker 1 (4S,18S)-4-(bis(2-(2-Chloroacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-bromoacetamido)ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid Synthetic Protocol: The solution of 2-(1-hydroxy-3-methylbutylidene)-5,5-dimethylcyclohexane-1,3-dione (2) (37.7 g, 168 mmol) in DCM (200 mL) was added dropwise to a solution of diethylenetriamine (1) (8.64 mL, 80.0 mmol) in DCM (130 mL). The reaction mixture was stirred overnight, then solvent was evaporated giving 2,2′-(((azanediylbis(ethane-2,1-diyl))bis(azanediyl))bis(3-methylbutan-1-yl-1-ylidene))bis(5,5-dimethylcyclohexane-1,3-dione) (3) as pale yellow oil. Yield: 41.2 g (100%). 1H NMR spectrum (300 MHz, CDCl3, δH): 3.57 (q, 4H); 3.06-2.89 (m, 8H); 2.36 (bs, 8H); 2.05-1.89 (m, 2H); 1.09-0.94 (m, 26H). The solution of the above amine (3) (33.4 g, 64.8 mmol) in DMF (320 mL) was added to a solution of 2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 5-tert-butyl ester (Fmoc-Glu(OtBu)-OH, 63.4 g, 149 mmol), HATU (56.6 g, 149 mmol), DIPEA (40.0 mL, 227 mmol) in DMF (530 mL). The reaction mixture was stirred at room temperature overnight. Then EtOAc (1.6 L) and water (1.6 L) were added. Separated organic layer was washed with aqueous solution of 10% K2CO3 (2×1.6 L), dried over anhydrous Na2SO4, filtered and evaporated in vacuo. The residue was purified by flash column chromatography (Silicagel 60, 0.063-0.040 mm; eluent: DCM/MeOH 50:1-40:1) to give tert-butyl 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoate (4) as pale yellow viscous oil. Yield: 59.2 g (99%). 1H NMR spectrum (300 MHz, CDCl3, δH): 7.77 (d, J=7.5 Hz, 2H); 7.59 (m, 2H); 7.40 (t, J=7.5 Hz, 2 Hz); 7.36-7.26 (m, 2H); 5.65 (d, J=9.2 Hz, 1H); 4.72-4.59 (m, 1H); 4.46-4.27 (m, 2H); 4.24-4.16 (m, 1H); 4.12-3.99 (m, 1H); 3.94-3.53 (m, 6H); 3.48-3.33 (m, 1H); 2.97 (bs, 4H); 2.46-2.26 (m, 10H); 2.08-1.83 (m, 4H); 1.79-1.64 (m, 1H); 1.43 (s, 9H); 1.07-0.90 (m, 24H). tert-Butyl 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoate (4) (59.2 g, 64.8 mmol) dissolved in DCM (50 mL) was added to TFA/water mixture (95:5, 400 mL) and stirred for 2 hrs. Then solvent was evaporated and residue was co-evaporated with toluene for three times. The residue was dissolved in DCM (800 mL) and washed with water (3×800 mL). The solvent was evaporated under reduced pressure. The residue was purified by column chromatography (Silicagel 60, 0.063-0.040 mm; eluent: DCM/MeOH 60:1-10:1) to give 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)-amino)ethyl)amino)-5-oxopentanoic acid (5) as white powder. Yield: 42.1 g (76%). 1H NMR spectrum (300 MHz, AcOD-d4, 80° C., δH): 7.79 (d, J=7.3 Hz, 2H); 7.64 (d, J=7.5 Hz, 2H); 7.39 (t, J=7.4 Hz, 2H); 7.36-7.26 (m, 2H); 4.83 (bs, 1H); 4.48-4.30 (m, 2H); 4.27-4.19 (m, 1H); 4.19-3.62 (m, 7H); 3.61-3.46 (m, 1H); 3.28-2.90 (m, 4H); 2.55 (t, J=6.7 Hz, 2H); 2.43 (s, 8H); 2.01-1.81 (m, 4H); 1.07-0.88 (m, 24H). Wang resin 0.63 mmol/g (25.7 g, 16.2 mmol) was left to swell in THF (250 mL) for 20 min. A solution of the 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoic acid (5) (42.0 g, 48.5 mmol) in THF (250 mL) was added to resin and then DIC (7.60 mL, 48.5 mmol) and 4-dimethylaminopyridine (DMAP, 200 mg, 1.62 mmol). The mixture was shaken for 18 hrs. Resin was filtered and washed with DCM (6×250 mL). Resin was treated by solution of acetic anhydride (40 mL), pyridine (40 mL) in DMF (360 mL) for 15 min and washed with DCM (6×250 mL) to give compound (6) as yellow solid. Yield: 36.0 g. Loading: 51% (0.321 mmol/g). The above compound (6) (6.27 g, 2.01 mmol) was left to swell in DCM (50 mL) for 20 min. Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×30 mL). Resin was washed with DMF (3×30 mL), 2-propanol (3×30 mL) and DCM (3×30 mL). A solution of {2-[2-(9H-fluoren-9-ylmethoxycarbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 1.25 g, 3.24 mmol), O-(6-chloro-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TCTU, 1.15 g, 3.24 mmol) and DIPEA (1.13 mL, 6.48 mmol) in DMF (35 mL) was added to resin and the mixture was shaken for 3 hrs. Resin was filtered and washed with DMF (3×30 mL), DCM (3×30 mL) and DMF (3×30 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×30 mL). Resin was washed with DMF (3×30 mL), 2-propanol (3×30 mL) and DCM (3×30 mL). Solution of (S)-2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 1-tert-butyl ester (Fmoc-LGlu-OtBu, 1.38 g, 3.24 mmol), O-(6-chloro-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TCTU, 1.15 g, 3.24 mmol) and DIPEA (1.13 mL, 6.48 mmol) in DMF (35 mL) was added to resin and mixture was shaken for 2 hrs. Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×30 mL). Resin was washed with DMF (3×30 mL), 2-propanol (3×30 mL) and DCM (3×30 mL). A solution of {2-[2-(9H-fluoren-9-ylmethoxycarbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 1.25 g, 3.24 mmol), O-(6-chloro-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TCTU, 1.15 g, 3.24 mmol) and DIPEA (1.13 mL, 6.48 mmol) in DMF (35 mL) was added to resin and the mixture was shaken for 3.5 hrs. Resin was filtered and washed with DMF (3×30 mL), DCM (3×30 mL) and DMF (3×30 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×30 mL). Resin was washed with DMF (3×30 mL), 2-propanol (3×30 mL) and DCM (3×30 mL). A solution of 1-(chloro-diphenyl-methyl)-4-methyl-benzene (MttCl, 2.02 g, 6.90 mmol) and DIPEA (2.55 mL, 14.6 mmol) in dry DCM (50 mL) was added to resin and mixture was shaken for 2 hrs. Resin was filtered and washed with DCM (4×30 mL) and DMF (4×30 mL). IvDde group was removed by treatment with 2% hydrazine monohydrate in DMF (3×30 mL, 3×3 min). Resin was washed with DMF (8×30 mL). A solution of chloroacetic acid (0.92 g, 9.74 mmol), bromo-tris-pyrrolidino-phosphonium hexafluorophosphate (PyBrOP, 4.54 g, 9.74 mmol) and DIPEA (3.39 mL, 19.5 mmol) in DMF (60 mL) was added to resin and mixture was shaken for 3 hrs. Resin was filtered and washed with DMF (4×30 mL), DCM (4×30 mL), DMF (4×30 mL), DCM (10×30 mL). Mtt group was removed by treatment with 80% 1,1,1,3,3,3-hexafluoro-2-propanol in DCM (4×30 mL, 2×10 min, 2×30 min). Resin was washed with DCM (5×30 mL) and DMF (4×30 mL). A solution of bromoacetic acid (4.50 g, 32.4 mmol) and DIC (4.27 mL, 27.6 mmol) in DMF (50 mL) was added to resin and mixture was shaken for 25 min. Resin was filtered and washed with DMF (4×30 mL), MeCN (2×30 mL) and DCM (10×30 mL). The product was cleaved from resin by treatment with cleavage cocktail of TFA/TIS/H2O (95:2.5:2.5, 50 mL) for 2 hrs. Resin was filtered and washed with TFA/DCM mixture (1:1, 50 mL) and DCM (10×50 mL). Solutions were combined and solvents were evaporated to dryness. The solvent was co-evaporated with toluene for three times. The residue was purified by Column X-Bridge3 C18, OBD, 5 μm, 50×250 mm (Mobile Phase: A=0.05% TFA/H2O, B=0.05% TFA/MeCN, gradient: 5% to 35%) to give (4S,18S)-4-(bis(2-(2-chloroacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-bromoacetamido)ethoxy)-ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid (7) as white solid. Yield: 694 mg (37%). 1H NMR spectrum (300 MHz, AcOD-d4, 80° C., δH): 5.13 (dd, J=9.3 Hz, J=4.2 Hz, 1H); 4.68 (dd, J=8.3 Hz, J=5.3 Hz, 1H); 4.20-4.06 (m, 8H); 3.95 (s, 2H); 3.91-3.41 (m, 24H); 2.61-2.08 (m, 7H); 2.08-1.88 (m, 1H). LC-MS purity: 100%. LC-MS Rt (Kinetex 4.6 mm×50 mm, MeCN/water 5:95 to 100:0+0.1% FA): Rt=4.74 min. LC-MS m/z: 926.6 (M+H)+. UPLC purity: 97.5% (214 nm). UPLC Rt (Acquity UPLC BEHC 18, 1.7 μm, 2.1×150 mm; MeCN/water 5:95 to 95:5+0.05% TFA): Rt=1.64 min. Trivalent Linker 2 2-(2-Bromoacetamido)-N,N-bis(2-(2-chloroacetamido)ethyl)acetamide Synthetic Protocol: Solution of 2-(1-hydroxy-3-methylbutylidene)-5,5-dimethylcyclohexane-1,3-dione (44.9 g, 200 mmol) in MeOH (400 mL) was added to diethylenetriamine (1) (DETA, 10.3 g, 100 mmol) in DCM (1.50 L) within 40 min. The reaction mixture was stirred overnight. The solvents were removed under reduced pressure and crude product was purified by flash column chromatography (Silicagel 60, 0.040-0.063 mm; eluent: DCM/MeOH 25/1) giving pure compound (2) as yellowish waxy solid. Yield: 41.0 g (80%). 1H NMR spectrum (300 MHz, CDCl3, δH): 13.83 (bs, 2H); 3.57 (q, J=5.7 Hz, 4H); 3.10-2.91 (m, 8H); 2.36 (bs, 8H); 1.97 (sep, J=6.8 Hz, 2H); 1.05-0.96 (m, 24H). To a solution of the above compound (2) (3.09 g, 6.00 mmol) was added a mixture of (tert-butoxycarbonyl)glycine (BocGlyOH, 2.10 g, 12.0 mmol), HATU (4.56 g, 12.0 mmol), and DIPEA (4.19 mL, 3.10 g, 24.0 mmol) in the mixture of DCM (200 mL) and DMF (40 mL). The reaction mixture was allowed to stirred for 2 hrs. Then 1M aqueous solution of potassium carbonate (200 mL) was added. The organic phase was separated and washed with 1M solution of hydrochloric acid (200 mL) and dried over anhydrous Na2SO4. The solvent was removed under reduced pressure. The residue was purified by flash column chromatography (Silicagel 60, 0.040-0.063 mm; eluent: EtOAc) giving pure compound (3) as brownish viscous oil. Yield: 3.98 g (99%). RF (SiO2, EtOAc): 0.50. 1H NMR spectrum (300 MHz, CDCl3, δH): 14.03 (bs, 1H); 13.87 (bs, 1H); 5.43 (bs, 1H); 4.01 (d, J=4.7 Hz, 2H); 3.79-3.56 (m, 8H); 3.06-2.89 (m, 4H); 2.48-2.27 (m, 8H); 1.95 (sep, J=6.8 Hz, 2H); 1.45 (s, 9H); 1.05-0.95 (m, 24H). The above compound (3) (3.98 g, 5.91 mmol) was dissolved in DCM (5 mL) and TFA (30 mL) was added. After 2 hrs the volatiles were removed under reduced pressure and saturated aqueous solution of potassium carbonate was added (60 mL). The product was extracted with EtOAc (3×40 mL). The organic phase was dried over anhydrous Na2SO4 and the solvent was removed under reduced pressure giving pure compound (4) as off-white solid foam. Yield: 3.38 g (100%). 1H NMR spectrum (300 MHz, CDCl3, δH): 14.01 (s, 1H); 13.88 (s, 1H); 3.80-3.70 (m, 2H); 3.68-3.58 (m, 6H); 3.52 (s, 2H); 3.06-2.93 (m, 4H); 2.45-2.29 (m, 8H), 1.95 (sep, J=6.8 Hz, 2H); 1.06-0.95 (m, 24H). 2-Chlorotrityl resin 100-200 mesh 1.8 mmol/g (4) (2 g, 7.43 mmol) was left to swell in dry DCM (100 mL) for 20 min. A solution of above compound (4) (2.83 g, 4.95 mmol) and DIPEA (3.28 mL, 18.8 mmol) in dry DCM (60 mL) was added to resin and the mixture was shaken overnight. Resin was filtered and treated with a solution of DIPEA (1.73 mL, 9.90 mmol) in MeOH/DCM mixture (4:1, 100 mL, 2×5 min). Then resin was washed with DMF (3×90 mL), 2-propanol (2×90 mL) and DCM (3×90 mL). The protecting groups were removed by treatment with hydrazine monohydrate (2% solution in DMF, 3×90 mL, 3×5 min). Then resin was washed with DMF (3×90 mL), 2-propanol (2×90 mL) and DCM (3×90 mL). Solution of chloroacetic acid (3.74 g, 39.6 mmol), 2,4,6-collidine (7.83 mL, 59.4 mmol) and DIC (6.13 mL, 39.6 mmol) in DMF (70 mL) was added to resin and mixture was shaken for 45 min. Resin was filtered and washed with DMF (4×90 mL), 2-propanol (2×90 mL) and DCM (8×90 mL). The product was cleaved from resin by treatment with cleavage cocktail (50% TFA in DCM, 80 mL) for 1 hour. Resin was filtered off and washed with DCM (3×40 mL). Solutions were combined and solvents were evaporated to dryness giving the desired compound (5) as thick brownish oil. Yield: 2.01 g (95%). 1H NMR spectrum (300 MHz, AcOD-d4, δH): 4.23 (s, 2H); 4.19 (s, 2H); 4.16 (s, 2H); 3.71-3.51 (m, 8H). Mixture of sodium bicarbonate (1.58 g, 18.8 mmol) and bromoacetic anhydride (1.71 g, 6.59 mmol) in MeCN (20 mL) was added to a solution of above compound (5) (2.01 g, 4.71 mmol) in MeCN (20 mL). After 90 min the reaction mixture was filtered through sintered glass and the solvent was removed under reduced pressure. The residue was purified by HPLC (Column X-Bridge4 C18, OBD, 5 μm, 50×250 mm, MeCN/H2O 5:95 to 45:55+0.05% TFA). Resulting solution was freeze-dried to give the title compound as colourless viscous oil. Addition of MeCN (8 mL) led to the formation of colourless crystals and the solvent was removed under reduced pressure affording 2-(2-bromoacetamido)-N,N-bis(2-(2-chloroacetamido)ethyl)acetamide (6) as colourless solid. Yield: 900 mg (44%). 1H NMR spectrum (300 MHz, AcOD-d4, δH): 4.27 (s, 2H); 4.19 (s, 2H); 4.13 (s, 2H); 4.02 (s, 2H); 3.70-3.50 (m, 8H). LC-MS purity: 100%. LC-MS Rt (Kinetex 4.6 mm×50 mm, MeCN/water 5:95 to 100:0+0.1% FA): 2.90 min. LC-MS m/z: 433.0 (M+H)+. Trivalent Linker 3 2-(2-(2-(2-(2-Bromoacetamido)ethoxy)ethoxy)acetamido)-N,N-bis(2-(2-chloroacetamido)ethyl)acetamide Synthetic Protocol: TFA (30 mL) was added to a solution of tert-butyl (2-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-2-oxoethyl)carbamate (1) (3.98 g, 5.91 mmol, prepared as described in Example 2, compound (3)) in DCM (5 mL). After 2 hrs the volatiles were removed under reduced pressure and saturated aqueous solution of potassium carbonate was added (60 mL). The product was extracted with EtOAc (3×40 mL). The organic phase was dried over anhydrous Na2SO4 and the solvent was removed under reduced pressure giving pure compound (2) as off-white solid foam. Yield: 3.38 g (100%). 1H NMR spectrum (300 MHz, CDCl3, δH): 14.01 (s, 1H); 13.88 (s, 1H); 3.80-3.70 (m, 2H); 3.68-3.58 (m, 6H); 3.52 (s, 2H); 3.06-2.93 (m, 4H); 2.45-2.29 (m, 8H); 1.95 (sep, J=6.8 Hz, 2H); 1.06-0.95 (m, 24H). To a solution of the above compound (2) (2.84 g, 4.97 mmol) was added a mixture of 2,2-dimethyl-4-oxo-3,8,11-trioxa-5-azatridecan-13-oic acid (Boc-OEG-OH, 1.31 g, 4.97 mmol), HATU (1.89 g, 4.97 mmol), and DIPEA (1.74 mL, 9.94 mmol) in DCM (200 mL) and DMF (30 mL). The reaction mixture was allowed to stir overnight. Then 1M aqueous solution of potassium carbonate (200 mL) was added. The organic phase was separated and washed with 1M solution of hydrochloric acid (200 mL) and dried over anhydrous Na2SO4. The solvent was removed under reduced pressure. The residue was purified by flash column chromatography (Silicagel 60, 0.040-0.063 mm; eluent: DCM/MeOH 20:1) giving pure compound (3) as brownish viscous oil. Yield: 3.19 g (78%). RF (SiO2 DCM/MeOH 20:1): 0.15. 1H NMR spectrum (300 MHz, CDCl3, δH): 14.06 (bs, 1H); 13.91 (bs, 1H); 7.66 (bs, 1H); 5.45 (bs, 1H); 4.19 (d, J=4.7 Hz, 2H); 4.05 (s, 2H); 3.79-3.62 (m, 12H); 3.57 (t, J=5.1 Hz, 2H); 3.38-3.29 (m, 2H); 3.04-2.92 (m, 4H); 2.45-2.27 (m, 8H); 1.93 (sep, J=6.8 Hz, 2H); 1.43 (s, 9H); 1.05-0.94 (m, 24H). TFA (30 mL) was added to a solution the above compound (3) (3.19 g, 3.89 mmol) in DCM (5 mL). After 2 hrs the volatiles were removed under reduced pressure and 1M aqueous solution of sodium hydroxide was added (60 mL). The product was extracted with ethyl acetate (7×30 mL). The organic phase was dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure giving pure compound (4) as off-white solid foam. Yield: 2.97 g (83%). 1H NMR spectrum (300 MHz, CDCl3, δH): 13.95 (bs, 1H); 13.79 (bs, 1H); 7.69 (bs, 1H); 4.17-4.10 (m, 2H); 4.03 (s, 2H); 3.74-3.56 (m, 12H); 3.52 (t, J=5.1 Hz, 2H); 3.03-2.83 (in, 6H); 2.32 (s, 8H); 2.13 (bs, 2H); 1.93 (sep, J=6.8 Hz, 2H); 1.43 (s, 9H); 1.03-0.90 (m, 24H). 2-Chlorotrityl resin 100-200 mesh 1.8 mmol/g (2.97 g, 5.35 mmol) was left to swell in dry DCM (100 mL) for 20 min. A solution of above compound (4) (2.92 g, 3.57 mmol) and DIPEA (2.36 mL, 13.6 mmol) in dry DCM (40 mL) was added to resin and the mixture was shaken overnight. Resin was filtered and treated with a solution of DIPEA (1.38 mL, 10.7 mmol) in MeOH/DCM mixture (4:1, 2×5 min, 70 mL). Then resin was washed with DMF (3×70 mL), 2-propanol (2×70 mL) and DCM (3×70 mL). The protecting groups were removed by treatment with hydrazine monohydrate (2% solution in DMF, 3×6 min, 3×70 mL). Then resin was washed with DMF (3×70 mL), 2-propanol (2×70 mL) and DCM (3×70 mL). Solution of chloroacetic acid (2.70 g, 28.6 mmol), 2,4,6-collidine (5.63 mL, 42.8 mmol) and DIC (4.42 mL, 28.6 mmol) in DMF (70 mL) was added to resin and mixture was shaken for 45 min. Resin was filtered and washed with DMF (4×70 mL), 2-propanol (2×70 mL) and DCM (8×70 mL). The product was cleaved from resin by treatment with cleavage cocktail (50% TFA in DCM, 80 mL) for 1 hour. Resin was filtered off and washed with DCM (3×40 mL). Solutions were combined and solvents were evaporated to dryness giving the desired compound (5) as thick brownish oil. Yield: 1.84 g (90%). 1H NMR spectrum (300 MHz, AcOD-d4, δH): 4.32 (s, 2H); 4.20 (s, 2H); 4.18 (s, 2H); 4.16 (s, 2H); 3.84 (t, J=4.8 Hz, 2H); 3.81-3.72 (m, 4H); 3.68-3.51 (m, 8H); 3.38 (t, J=4.8 Hz, 2H). Mixture of sodium bicarbonate (0.81 g, 9.63 mmol) and bromoacetic anhydride (1.67 g, 6.42 mmol) in MeCN (20 mL) was added to a solution of the above compound (5) (1.84 g, 3.21 mmol) in MeCN (20 mL). After 90 min the reaction mixture was filtered through sintered glass and the solvent was removed under reduced pressure. The residue was purified by HPLC (Column labio DeltaPak C18, 15 μm, 50×500 mm, MeCN/water 5:95 to 45:55+0.05% TFA). Resulting solution was freeze-dried to give 2-(2-(2-(2-(2-bromoacetamido)ethoxy)ethoxy)acetamido)-N,N-bis(2-(2-chloroacetamido)ethyl)acetamide (6) as colourless viscous oil. Yield: 516 mg (33%). 1H NMR spectrum (300 MHz, AcOD-d4, δH): 4.29 (s, 2H); 4.19 (s, 2H); 4.16 (s, 2H); 4.14 (s, 2H); 3.98 (s, 2H); 3.75-3.49 (m, 16H). LC-MS purity: 100%. LC-MS Rt (Kinetex 4.6 mm×50 mm, MeCN/water 5:95 to 100:0+0.1% FA): 3.02 min. LC-MS m/z: 480.2 (M+H)+. Trivalent Linker 4 (R)-4-{2-[2-({Bis-[2-(2-chloro-acetylamino)-ethyl]-carbamoyl}-methoxy)-ethoxy]-thylcarbamoyl}-2-[(S)-2-(2-{2-[2-(2-bromo-acetylamino)-ethoxy]-ethoxy}-acetylamino)-4-carboxy-butyrylamino]-butyric acid Synthetic Protocol: 2-Chlorotrityl resin 100-200 mesh 1.8 mmol/g (1, 41.7 g, 75.1 mmol) was left to swell in dry DCM (350 mL) for 20 min. A solution of {2-[2-(9H-fluoren-9-ylmethoxycarbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 19.3 g, 50.1 mmol) and DIPEA (33.1 mL, 190 mmol) in dry DCM (250 mL) was added to resin and the mixture was shaken overnight. The resin was filtered and treated with a solution of DIPEA (17.4 mL, 100 mmol) in MeOH/DCM mixture (4:1, 5 min, 200 mL). Then resin was washed with DCM (2×250 mL) and DMF (2×250 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×15 min, 2×250 mL). The resin was washed with DMF (2×250 mL), 2-propanol (2×250 mL), DCM (2×250 mL) and DMF (2×250 mL). Solution of 2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 1-tert-butyl ester (Fmoc-Glu-OtBu, 42.6 g, 100 mmol), O-(6-chloro-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TCTU, 35.6 g, 100 mmol) and DIPEA (31.4 mL, 180 mmol) in DMF (200 mL) was added to resin and mixture was shaken for 4 hours. Resin was filtered and washed with DMF (2×250 mL) and DCM (10×250 mL). The product was cleaved from resin by treatment with 2,2,2-trifluoroethanol (350 mL) overnight. Resin was filtered off and washed with DCM (2×200 mL), solvent was evaporated and crude product was purified by flash column chromatography (Silicagel 60, 0.040-063 mm; eluent: DCM/MeOH 95:5 to 85:15) giving (R)-4-[2-(2-carboxymethoxy-ethoxy)-ethylcarbamoyl]-2-(9H-fluoren-9-ylmethoxycarbonylamino)-butyric acid tert-butyl ester (2) as yellowish waxy solid. Yield: 18.6 g (65%). 1H NMR spectrum (300 MHz, CDCl3, δH): 7.77 (d, J=7.5 Hz, 2H); 7.65-7.50 (m, 2H); 7.47-7.37 (m, 2H); 7.37-6.72 (m, 2H); 6.84-6.72 (m, 1H); 5.92-5.81 (m, 1H); 4.58-4.30 (m, 2H); 4.30-3.95 (m, 4H); 3.79-3.51 (m, 6H); 3.43 (q, J=4.6 Hz, 2H); 2.39-1.90 (m, 4H); 1.54-1.39 (m, 9H). LC-MS purity: 100%. LC-MS Rt (Kinetex C18, 4.6 mm×50 mm, MeCN/water 20:80 to 100:0+0.1% FA): 3.65 min. LC-MS m/z: 570.6 (M+H)+. Diethylenetriamine (3) (1.62 mL, 15.0 mmol) was added to a solution of 2-(1-hydroxy-3-methyl-butylidene)-5,5-dimethyl-cyclohexane-1,3-dione (4) (6.73 g, 30.0 mmol) in DCM (125 mL). The resulting solution was stirred overnight, then the solvent was evaporated and residue was dried in vacuo affording 2,2′-(((azanediylbis(ethane-2,1-diyl))bis(azanediyl))bis(3-methylbutan-1-yl-1-ylidene))bis(5,5-dimethylcyclohexane-1,3-dione) (5) as yellow oil. Yield: 7.74 g (100%). 1H NMR spectrum (300 MHz, CDCl3, δH): 13.81 (bs, 2H); 3.64-3.44 (m, 4H); 3.12-2.83 (m, 8H); 2.34 (s, 8H); 2.06-1.86 (m, 2H); 1.05-0.87 (m, 24H). The compound (2) (18.5 g, 32.5 mmol) was dissolved in DCM (220 mL) followed by addition of HATU (12.4 g, 32.5 mmol), DIPEA (8.30 mL, 47.6 mmol) and solution of the amine (5) (7.04 g, 13.6 mmol) in DCM (150 mL). The resulting solution was stirred overnight and then the solvent was evaporated. The residue was dissolved in ethyl acetate (250 mL) and washed with water (4×250 mL). Organic layer was dried over anhydrous sodium sulphate, filtered and evaporated. The residue was purified by flash column chromatography (Silicagel 60, 0.040-063 mm; eluent: DCM/MeOH 97:3 to 96:4) giving (R)-4-(2-{2-[(bis-{2-[1-(4,4-dimethyl-2,6-dioxo-cyclohexylidene)-3-methyl-butylamino]-ethyl}-carbamoyl)-methoxy]-ethoxy}-ethylcarbamoyl)-2-(9H-fluoren-9-ylmethoxycarbonylamino)-butyric acid tert-butyl ester (6) as white solid. Yield: 11.6 g (80%). 1H NMR spectrum (300 MHz, CDCl3, δH): 14.02-13.78 (m, 2H); 7.77 (d, J=7.2 Hz, 2H); 7.60 (d, J=6.8 Hz, 2H); 7.40 (t, J=7.1 Hz, 2H); 7.35-7.24 (m, 2H); 6.76 (bs, 1H); 5.85 (d, J=8.5 Hz, 1H); 4.50-4.13 (m, 6H); 3.79-3.35 (m, 16H); 3.06-2.85 (m, 4H); 2.45-2.12 (m, 11H); 2.09-1.82 (m, 3H); 1.47 (s, 9H); 1.07-0.85 (m, 24H). LC-MS purity: 100%. LC-MS Rt (Kinetex C18, 4.6 mm×50 mm, MeCN/water 70:30 to 100:0+0.1% FA): 2.12 min. LC-MS m/z: 1068.3 (M+H)+. The above prepared compound (6) (11.6 g, 10.9 mmol) was dissolved in mixture of TFA/H2O (95:5, 150 mL) and left to stay for 2.5 hrs. Then solvent was evaporated. The residue was dissolved in DCM (30 mL) and diethyl ether (200 mL) was added. The mixture was stirred overnight; then diethyl ether was decanted. The residue was treated with diethyl ether (200 mL) and then decanted again. The procedure was repeated once more. The residue was dried in vacuo to yield (R)-4-(2-{2-[(bis-{2-[1-(4,4-dimethyl-2,6-dioxo-cyclohexylidene)-3-methyl-butylamino]-ethyl}-carbamoyl)-methoxy]-ethoxy}-ethylcarbamoyl)-2-(9H-fluoren-9-ylmethoxycarbonylamino)-butyric acid (7) as pale yellow solid. Yield: 10.7 g (96%). 1H NMR spectrum (300 MHz, CDCl3, δH): 13.90-13.65 (m, 2H); 7.77 (d, J=7.5 Hz, 2H); 7.67-7.53 (m, 2H); 7.40 (t, J=7.2 Hz, 2H); 7.35-7.23 (m, 3H); 6.01 (d, J=7.5 Hz, 1H); 4.49-4.34 (m, 3H); 4.31-4.15 (m, 3H); 3.86-3.30 (m, 16H); 3.14-2.84 (m, 4H); 2.55-2.31 (m, 10H); 2.30-2.05 (m, 2H); 2.03-1.78 (m, 2H); 1.11-0.88 (m, 24H). LC-MS purity: 100%. LC-MS Rt (Kinetex C18, 4.6 mm×50 mm, MeCN/water 35:65 to 100:0+0.1% FA): 3.68 min. LC-MS m/z: 1012.2 (M+H)+. Wang resin 0.68 mmol/g (1) (4.97 g, 3.38 mmol) was swollen in THF (110 mL) for 60 min. A solution of compound (7) (10.3 g, 10.1 mmol), DIC (1.57 mL, 10.1 mmol) and 4-dimethylamino-pyridine (0.04 g, 0.34 mmol) in THF (80 mL) was added to resin and the mixture was shaken overnight. Resin was filtered, washed with DCM (2×100 mL) and treated with a solution of acetic anhydride (5.00 mL, 50.1 mmol) and pyridine (5.00 mL, 61.6 mmol) in DCM (70 mL) for 10 minutes. Resin was washed with DCM (6×100 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×15 min, 2×80 mL). Resin was washed with DMF (2×100 mL), 2-propanol (2×100 mL), DCM (2×100 mL) and DMF (2×100 mL). A solution of (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(tert-butoxy)-5-oxopentanoic acid (Fmoc-Glu(OtBu)-OH, 4.31 g, 10.1 mmol), 5-chloro-1-((dimethylamino)(dimethyliminio)methyl)-1H-benzo[d][1,2,3]triazole 3-oxide tetrafluoroborate (TCTU, 3.60 g, 10.1 mmol) and DIPEA (3.18 mL, 18.2 mmol) in DMF (70 mL) was added to resin and the mixture was shaken for 3 hours. Resin was filtered and washed with DMF (2×100 mL), DCM (2×100 mL) and DMF (2×100 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×15 min, 2×80 mL). Resin was washed with DMF (2×100 mL), 2-propanol (2×100 mL), DCM (2×100 mL) and DMF (2×100 mL). A solution of {2-[2-(9H-fluoren-9-ylmethoxycarbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 3.90 g, 10.1 mmol), 5-chloro-1-((dimethylamino)(dimethyliminio)methyl)-1H-benzo[d][1,2,3]triazole 3-oxide tetrafluoroborate (TCTU, 3.60 g, 10.1 mmol) and DIPEA (3.18 mL, 18.2 mmol) in DMF (70 mL) was added to resin and the mixture was shaken overnight. Resin was filtered and washed with DMF (2×100 mL), DCM (2×100 mL) and DMF (2×100 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×15 min, 2×80 mL). Resin was washed with DMF (2×100 mL), 2-propanol (2×100 mL), DCM (2×100 mL) and DMF (2×100 mL). A solution of 1-(chloro-diphenyl-methyl)-4-methyl-benzene (MttCl, 2.97 g, 10.1 mmol) and DIPEA (3.53 mL, 20.3 mmol) in dry DCM (70 mL) was added to resin and mixture was shaken for 4.5 hrs. Resin was filtered and washed with DMF (2×100 mL), DCM (3×100 mL) and DMF (3×100 mL). The IvDde group was removed by treatment with 2% hydrazine monohydrate in DMF (3×3 min, 3×80 mL). Resin was washed with DMF (6×100 mL). A solution of chloroacetic acid (1.91 g, 20.3 mmol), DIC (3.14 mL, 20.3 mmol) and 2,4,6-trimethylpyridine (5.35 mL, 40.5 mmol) in DMF (80 mL) was added to resin and mixture was shaken for 2.5 hrs. Resin was filtered and washed with DMF (3×100 mL) and DCM (3×100 mL). The Mtt group was removed by treatment with 80% 1,1,1,3,3,3-hexafluoro-2-propanol in DCM (2×10 min, 3×30 min, 5×75 mL). Resin was washed with DCM (3×100 mL) and DMF (3×100 mL). A solution of bromoacetic acid (9.39 g, 67.6 mmol) and DIC (8.90 mL, 57.5 mmol) in DMF (80 mL) was added to resin and mixture was shaken for 20 min. Resin was filtered and washed with DMF (3×100 mL) and DCM (10×100 mL). The product was cleaved from resin by treatment with mixture of TFA and water (98:2, 100 mL) for 1 hr. Resin was filtered and washed with DCM (2×80 mL). Solutions were combined and solvents were evaporated to dryness. The residue was purified by HPLC (Column DeltaPak C18, 15 um; 50×500 mm; MeCN/water 5:95 to 60:40+0.05% TFA) and freeze-dried to give the title compound (9) as a white solid. Yield: 425 mg (14%). 1H NMR spectrum (300 MHz, AcOD-d4, δH): 4.76 (dd, J=7.8 and 5.9 Hz, 1H); 4.57 (dd, J=8.9 and 4.9 Hz, 1H); 4.36 (s, 2H); 4.20 (s, 2H); 4.14 (s, 4H); 3.98 (s, 2H); 3.81-3.41 (m, 24H); 2.56 (t, J=8.0 Hz, 2H); 2.47 (t, J=7.2 Hz, 2H); 2.37-2.20 (m, 2H); 2.20-2.05 (m, 2H). LC-MS purity: 100%. LC-MS Rt (Kinetex C18, 4.6 mm×50 mm, MeCN/water 5:95 to 100:0+0.1% FA): 3.58 min. LC-MS m/z: 925.6 (M+H)+. Trivalent Linker 5 (13R,18S)-18-(bis(2-(2-Chloroacetamido)ethyl)carbamoyl)-1-bromo-13-carboxy-2,11,16-trioxo-6,9-dioxa-3,12,17-triazahenicosan-21-oic acid Synthetic Protocol: Preparation of Wang resin-bound 1 was described in protocol REaD-24247 (Batch No. 218-004-1). Wang resin-bound (1, 2.85 g, 0.92 mmol) was left to swell in DCM (20 mL) for 20 min. Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×20 mL). Resin was washed with DMF (3×20 mL), 2-propanol (3×20 mL) and DCM (3×20 mL). Solution of (S)-2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 1-tert-butyl ester (Fmoc-LGlu-OtBu, 1.17 g, 2.75 mmol), TCTU (0.98 g, 2.75 mmol) and DIPEA (0.96 mL, 5.49 mmol) in DMF (20 mL) was added to resin and mixture was shaken for 2 hrs. Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×20 mL). Resin was washed with DMF (3×20 mL), 2-propanol (3×20 mL) and DCM (3×20 mL). A solution of {2-[2-(9H-fluoren-9-ylmethoxycarbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 1.06 g, 2.75 mmol), TCTU (0.98 g, 2.75 mmol) and DIPEA (0.96 mL, 5.49 mmol) in DMF (20 mL) was added to resin and the mixture was shaken for 3.5 hrs. Resin was filtered and washed with DMF (3×20 mL), DCM (3×20 mL) and DMF (3×20 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×20 mL). Resin was washed with DMF (3×20 mL), 2-propanol (3×20 mL) and DCM (3×20 mL). A solution of 1-(chloro-diphenyl-methyl)-4-methyl-benzene (MttCl, 1.20 g, 4.10 mmol) and DIPEA (1.44 mL, 8.27 mmol) in dry DCM (40 mL) was added to resin and mixture was shaken for 1 hour. Resin was filtered and washed with DCM (4×20 mL) and DMF (4×20 mL). IvDde group was removed by treatment with 2% hydrazine monohydrate in DMF (3×3 min, 3×20 mL). Resin was washed with DMF (8×20 mL). A solution of chloroacetic acid (0.52 g, 5.49 mmol), PyBroP (2.56 g, 5.49 mmol) and DIPEA (1.91 mL, 11.0 mmol) in DMF (25 mL) was added to resin and mixture was shaken for 3 hrs. Resin was filtered and washed with DMF (4×20 mL), DCM (4×20 mL), DMF (4×20 mL), DCM (10×20 mL). Mtt group was removed by treatment with 80% 1,1,1,3,3,3-hexafluoro-2-propanol in DCM (2×10 min, 2×30 min, 4×25 mL). Resin was washed with DCM (5×20 mL) and DMF (4×20 mL). A solution of bromoacetic acid (2.54 g, 18.3 mmol) and DIC (2.41 mL, 15.6 mmol) in DMF (30 mL) was added to resin and mixture was shaken for 25 minutes. Resin was filtered and washed with DMF (4×20 mL), MeCN (2×20 mL) and DCM (10×20 mL). The product was cleaved from resin by treatment with cleavage cocktail of TFA/TIS/H2O (95:2.5:2.5, 30 mL) for 1 hour. Resin was filtered and washed with DCM (10×30 mL). Solutions were combined and solvents were evaporated to dryness. The solvent was co-evaporated with toluene for three times. The residue was purified by HPLC (Column X-Bridge3 C18, OBD, 5 μm, 50×250 mm, MeCN/water 5% to 35%+0.05% TFA) to give (13R,18S)-18-(bis(2-(2-chloroacetamido)ethyl)carbamoyl)-1-bromo-13-carboxy-2,11,16-trioxo-6,9-dioxa-3,12,17-triazahenicosan-21-oic acid (2) as white solid. Yield: 333 mg (47%). 1H NMR spectrum (300 MHz, AcOD-d4, 80° C., δH): 5.08 (dd, J=9.5 Hz, J=4.2 Hz, 1H); 4.71 (dd, J=8.1 Hz, J=5.1 Hz, 1H); 4.19-4.09 (m, 6H); 3.96 (s, 2H); 3.90-3.44 (m, 16H); 2.59-2.07 (m, 7H); 2.01-1.86 (m, 1H). LC-MS purity: 100%. LC-MS Rt (Kinetex 4.6 mm×50 mm, MeCN/H2O 5:95 to 100:0+0.1% FA): 4.56 min. LC-MS m/z: 781.5 (M+H)+. UPLC purity: 99% (220 nm). UPLC Rt (Acquity UPLC BEHC 18, 1.7 μm, 2.1×150 mm; MeCN/H2O 5:95 to 100:0+0.1% TFA): 1.57 min. Trivalent Linker 6 (18R,23S)-23-(Bis(2-(2-chloroacetamido)ethyl)carbamoyl)-1-bromo-18-carboxy-2,7,16,21-tetraoxo-11,14-dioxa-3,8,17,22-tetraazahexacosan-26-oic acid Reaction Scheme: Synthetic Protocol: Preparation of Compound (1). The solution of 2-(1-hydroxy-3-methylbutylidene)-5,5-dimethylcyclohexane-1,3-dione (37.7 g, 168 mmol) in DCM (200 mL) was added dropwise to a solution of diethylenetriamine (8.64 mL, 80.0 mmol) in DCM (130 mL). The reaction mixture was stirred overnight, then solvent was evaporated affording 2,2′-(((azanediylbis(ethane-2,1-diyl))bis(azanediyl))bis(3-methylbutan-1-yl-1-ylidene))bis(5,5-dimethylcyclohexane-1,3-dione) as pale yellow oil. Yield: 41.2 g (100%). 1H NMR spectrum (300 MHz, CDCl3, δH): 3.57 (q, 4H); 3.06-2.89 (m, 8H); 2.36 (bs, 8H); 2.05-1.89 (m, 2H); 1.09-0.94 (m, 26H). The solution of the above amine (33.4 g, 64.8 mmol) in DMF (320 mL) was added to a solution of 2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 5-tert-butyl ester (Fmoc-Glu(OtBu)-OH, 63.4 g, 149 mmol), HATU (56.6 g, 149 mmol), DIPEA (40.0 mL, 227 mmol) in DMF (530 mL). The reaction mixture was stirred at room temperature overnight. Then ethyl acetate (1.6 L) and water (1.6 L) were added. Separated organic layer was washed with aqueous solution of 10% K2CO3 (2×1.6 L), dried over anhydrous Na2SO4, filtered and evaporated in vacuo. The residue was purified by flash column chromatography (Silicagel 60, 0.063-0.040 mm; eluent: DCM/MeOH 50:1-40:1) to give tert-butyl 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoate as pale yellow viscous oil. Yield: 59.2 g (99%). 1H NMR spectrum (300 MHz, CDCl3, δH): 7.77 (d, J=7.5 Hz, 2H); 7.59 (m, 2H); 7.40 (t, J=7.5 Hz, 2 Hz); 7.36-7.26 (m, 2H); 5.65 (d, J=9.2 Hz, 1H); 4.72-4.59 (m, 1 H); 4.46-4.27 (m, 2H); 4.24-4.16 (m, 1H); 4.12-3.99 (m, 1H); 3.94-3.53 (m, 6H); 3.48-3.33 (m, 1H); 2.97 (bs, 4H); 2.46-2.26 (m, 10H); 2.08-1.83 (m, 4H); 1.79-1.64 (m, 1H); 1.43 (s, 9H); 1.07-0.90 (m, 24H). tert-Butyl 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoate (59.2 g, 64.8 mmol) dissolved in DCM (50 mL) was added to TFA/H2O mixture (95:5, 400 mL) and stirred for 2 hrs. Then solvent was evaporated and the residue was co-evaporated with toluene for three times. The residue was dissolved in DCM (800 mL) and washed with water (3×800 mL). The solvent was evaporated under reduced pressure. The residue was purified by column chromatography (Silicagel 60, 0.063-0.040 mm; eluent: DCM/methanol 60:1-10:1) affording 4-((((9H-fluoren-9-yl)methoxy)-carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)-amino)ethyl)amino)-5-oxopentanoic acid as white powder. Yield: 42.1 g (76%). 1H NMR spectrum (300 MHz, AcOD-d4, 80° C., δH): 7.79 (d, J=7.3 Hz, 2H); 7.64 (d, J=7.5 Hz, 2H); 7.39 (t, J=7.4 Hz, 2H); 7.36-7.26 (m, 2H); 4.83 (bs, 1H); 4.48-4.30 (m, 2H); 4.27-4.19 (m, 1H); 4.19-3.62 (m, 7H); 3.61-3.46 (m, 1H); 3.28-2.90 (m, 4H); 2.55 (t, J=6.7 Hz, 2H); 2.43 (s, 8H); 2.01-1.81 (m, 4H); 1.07-0.88 (m, 24H). Wang resin 0.63 mmol/g (25.7 g, 16.2 mmol) was left to swell in tetrahydrofuran (250 mL) for 20 minutes. A solution of the 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoic acid (42.0 g, 48.5 mmol) in THF (250 mL) was added to resin and then DIC (7.60 mL, 48.5 mmol) and 4-dimethylaminopyridine (DMAP, 200 mg, 1.62 mmol). The mixture was shaken for 18 hrs. The resin was filtered off and washed with DCM (6×250 mL). Resin was treated with a solution of acetic anhydride (40 mL), pyridine (40 mL) in DMF (360 mL) for 15 min. and washed with DCM (6×250 mL) affording compound (1) as yellow solid. Yield: 36.0 g. Loading: 51% (0.321 mmol/g). Wang resin-bound compound (1) (2.85 g, 0.92 mmol) was swollen in DCM (20 mL) for 20 min. The Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×40 mL). Resin was washed with DMF (3×70 mL), 2-propanol (3×70 mL) and DCM (3×70 mL). Solution of (S)-2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 1-tert-butyl ester (Fmoc-LGlu-OtBu, 1.17 g, 2.75 mmol), TCTU (0.98 g, 2.75 mmol) and DIPEA (0.96 mL, 5.49 mmol) in DMF (60 mL) was added to resin and mixture was shaken for 3 hrs. Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×40 mL). Resin was washed with DMF (3×70 mL), 2-propanol (3×70 mL) and DCM (3×70 mL). A solution of {2-[2-(9H-fluoren-9-ylmethoxy-carbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 1.06 g, 2.75 mmol), TCTU (0.98 g, 2.75 mmol) and DIPEA (0.96 mL, 5.49 mmol) in DMF (50 mL) was added to resin and the mixture was shaken for 2 hrs. Resin was filtered and washed with DMF (3×70 mL), DCM (3×70 mL) and DMF (3×70 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×40 mL). Resin was washed with DMF (3×70 mL), 2-propanol (3×70 mL) and DCM (3×70 mL). A solution of 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)butanoic acid (Fmoc-GABA, 0.88 g, 2.70 mmol), TCTU (0.96 g, 2.70 mmol) and DIPEA (0.94 mL, 5.40 mmol) in DMF (50 mL) was added to resin and the mixture was shaken for 2 hrs. Resin was filtered and washed with DMF (3×70 mL), DCM (3×70 mL) and DMF (3×70 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×40 mL). Resin was washed with DMF (3×70 mL), 2-propanol (3×70 mL) and DCM (3×70 mL). A solution of 1-(chloro-diphenyl-methyl)-4-methyl-benzene (MttCl, 0.79 g, 2.70 mmol) and DIPEA (0.94 mL, 5.4 mmol) in dry DCM (60 mL) was added to resin and mixture was shaken for 3 hrs. The resin was filtered off and washed with DCM (3×70 mL) and DMF (3×70 mL). The IvDde group was removed by treatment with 2% hydrazine monohydrate in DMF (3×3 min, 3×30 mL). The resin was washed with DMF (5×40 mL). A solution of chloroacetic acid (0.51 g, 5.40 mmol), DIC (0.85 mL, 5.40 mmol) and 2,4,6-trimethylpyridine (1.43 mL, 10.8 mmol) in DMF (50 mL) was added to the resin and the mixture was shaken for 2.5 hrs. The resin was filtered off and washed with DMF (3×70 mL) and DCM (3×70 mL). The Mtt group was removed by treatment with 80% 1,1,1,3,3,3-hexafluoro-2-propanol in DCM (2×10 min, 2×30 min, 4×50 mL). The resin was washed with DCM (5×50 mL) and DMF (4×50 mL). A solution of bromoacetic acid (0.38 g, 2.70 mmol) and DIC (0.42 mL, 2.70 mmol) in DMF (50 mL) was added to resin and mixture was shaken for 2.5 hrs. Resin was filtered and washed with DMF (3×70 mL) and DCM (3×70 mL). The product was cleaved from resin by treatment with cleavage cocktail of TFA/TIS/H2O (95:2.5:2.5, 26 mL) for 1 hr. The resin was filtered off and washed with DCM (6×40 mL). Solutions were combined and solvents were evaporated to dryness. The residue was purified by HPLC (Column Gemini C18, 5 μm; 50×250 mm; MeCN/H2O 5:95 to 40:60 during 180 min. and 5:95 to 35:65 during 60 min.+0.05% TFA) and freeze-dried to give title compound (2) as white solid. Yield: 336 mg (43%). 1H NMR spectrum (300 MHz, AcOD-d4, δH): 5.07 (dd, J=9.9 and 2.9 Hz, 1H); 4.72 (dd, J=8.7 and 4.7 Hz, 1H); 4.22-4.12 (m, 6H); 3.96 (s, 2H); 3.93-3.37 (m, 16H); 3.33 (t, J=6.8 Hz, 3H); 2.59-2.29 (m, 7H); 2.10 (d, J=4.9 Hz, 2H); 1.97-1.80 (m, 3H). LC-MS purity: 100%. LC-MS Rt (Kinetex 4.6 mm×50 mm, MeCN/H2O 5:95 to 100:0+0.1% FA): 2.86 min. LC-MS m/z: 866.5 (M+H)+. UPLC purity: 98.7% (214 nm). UPLC Rt (Acquity UPLC BEHC 18, 1.7 μm, 2.1×150 mm; MeCN/H2O 5:95 to 100:0+0.05% TFA): 2.74 min. Trivalent Linker 6 (18R,23S)-23-(Bis(2-(2-chloroacetamido)ethyl)carbamoyl)-1-bromo-18-carboxy-2,7,16,21-tetraoxo-11,14-dioxa-3,8,17,22-tetraazahexacosan-26-oic acid Synthetic Protocol: Preparation of Compound (1). The solution of 2-(1-hydroxy-3-methylbutylidene)-5,5-dimethylcyclohexane-1,3-dione (37.7 g, 168 mmol) in DCM (200 mL) was added dropwise to a solution of diethylenetriamine (8.64 mL, 80.0 mmol) in DCM (130 mL). The reaction mixture was stirred overnight, then solvent was evaporated affording 2,2′-(((azanediylbis(ethane-2,1-diyl))bis(azanediyl))bis(3-methylbutan-1-yl-1-ylidene))bis(5,5-dimethylcyclohexane-1,3-dione) as pale yellow oil. Yield: 41.2 g (100%). 1H NMR spectrum (300 MHz, CDCl3, δH): 3.57 (q, 4H); 3.06-2.89 (m, 8H); 2.36 (bs, 8H); 2.05-1.89 (m, 2H); 1.09-0.94 (m, 26H). The solution of the above amine (33.4 g, 64.8 mmol) in DMF (320 mL) was added to a solution of 2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 5-tert-butyl ester (Fmoc-Glu(OtBu)-OH, 63.4 g, 149 mmol), HATU (56.6 g, 149 mmol), DIPEA (40.0 mL, 227 mmol) in DMF (530 mL). The reaction mixture was stirred at room temperature overnight. Then ethyl acetate (1.6 L) and water (1.6 L) were added. Separated organic layer was washed with aqueous solution of 10% K2CO3 (2×1.6 L), dried over anhydrous Na2SO4, filtered and evaporated in vacuo. The residue was purified by flash column chromatography (Silicagel 60, 0.063-0.040 mm; eluent: DCM/MeOH 50:1-40:1) to give tert-butyl 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoate as pale yellow viscous oil. Yield: 59.2 g (99%). 1H NMR spectrum (300 MHz, CDCl3, δH): 7.77 (d, J=7.5 Hz, 2H); 7.59 (m, 2H); 7.40 (t, J=7.5 Hz, 2 Hz); 7.36-7.26 (m, 2H); 5.65 (d, J=9.2 Hz, 1H); 4.72-4.59 (m, 1H); 4.46-4.27 (m, 2H); 4.24-4.16 (m, 1H); 4.12-3.99 (m, 1H); 3.94-3.53 (m, 6H); 3.48-3.33 (m, 1H); 2.97 (bs, 4H); 2.46-2.26 (m, 10H); 2.08-1.83 (m, 4H); 1.79-1.64 (m, 1H); 1.43 (s, 9H); 1.07-0.90 (m, 24H). tert-Butyl 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoate (59.2 g, 64.8 mmol) dissolved in DCM (50 mL) was added to TFA/H2O mixture (95:5, 400 mL) and stirred for 2 hrs. Then solvent was evaporated and the residue was co-evaporated with toluene for three times. The residue was dissolved in DCM (800 mL) and washed with water (3×800 mL). The solvent was evaporated under reduced pressure. The residue was purified by column chromatography (Silicagel 60, 0.063-0.040 mm; eluent: DCM/methanol 60:1-10:1) affording 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoic acid as white powder. Yield: 42.1 g (76%). 1H NMR spectrum (300 MHz, AcOD-d4, 80° C., δH): 7.79 (d, J=7.3 Hz, 2H); 7.64 (d, J=7.5 Hz, 2H); 7.39 (t, J=7.4 Hz, 2H); 7.36-7.26 (m, 2H); 4.83 (bs, 1H); 4.48-4.30 (m, 2H); 4.27-4.19 (m, 1H); 4.19-3.62 (m, 7H); 3.61-3.46 (m, 1H); 3.28-2.90 (m, 4H); 2.55 (t, J=6.7 Hz, 2H); 2.43 (s, 8H); 2.01-1.81 (m, 4H); 1.07-0.88 (m, 24H). Wang resin 0.63 mmol/g (25.7 g, 16.2 mmol) was left to swell in tetrahydrofuran (250 mL) for 20 minutes. A solution of the 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(bis(2-((1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-3-methylbutyl)amino)ethyl)amino)-5-oxopentanoic acid (42.0 g, 48.5 mmol) in THF (250 mL) was added to resin and then DIC (7.60 mL, 48.5 mmol) and 4-dimethylaminopyridine (DMAP, 200 mg, 1.62 mmol). The mixture was shaken for 18 hrs. The resin was filtered off and washed with DCM (6×250 mL). Resin was treated with a solution of acetic anhydride (40 mL), pyridine (40 mL) in DMF (360 mL) for 15 min. and washed with DCM (6×250 mL) affording compound (1) as yellow solid. Yield: 36.0 g. Loading: 51% (0.321 mmol/g). Wang resin-bound compound (1) (2.85 g, 0.92 mmol) was swollen in DCM (20 mL) for 20 min. The Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×40 mL). Resin was washed with DMF (3×70 mL), 2-propanol (3×70 mL) and DCM (3×70 mL). Solution of (S)-2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 1-tert-butyl ester (Fmoc-LGlu-OtBu, 1.17 g, 2.75 mmol), O-(6-chloro-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TCTU, 0.98 g, 2.75 mmol) and DIPEA (0.96 mL, 5.49 mmol) in DMF (60 mL) was added to resin and mixture was shaken for 3 hrs. Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×40 mL). Resin was washed with DMF (3×70 mL), 2-propanol (3×70 mL) and DCM (3×70 mL). A solution of {2-[2-(9H-fluoren-9-ylmethoxy-carbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 1.06 g, 2.75 mmol), O-(6-chloro-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TCTU, 0.98 g, 2.75 mmol) and DIPEA (0.96 mL, 5.49 mmol) in DMF (50 mL) was added to resin and the mixture was shaken for 2 hours. Resin was filtered and washed with DMF (3×70 mL), DCM (3×70 mL) and DMF (3×70 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×40 mL). Resin was washed with DMF (3×70 mL), 2-propanol (3×70 mL) and DCM (3×70 mL). A solution of 4-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)butanoic acid (Fmoc-GABA, 0.88 g, 2.70 mmol), O-(6-chloro-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TCTU, 0.96 g, 2.70 mmol) and DIPEA (0.94 mL, 5.40 mmol) in DMF (50 mL) was added to resin and the mixture was shaken for 2 hours. Resin was filtered and washed with DMF (3×70 mL), DCM (3×70 mL) and DMF (3×70 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (1×5 min, 1×30 min, 2×40 mL). Resin was washed with DMF (3×70 mL), 2-propanol (3×70 mL) and DCM (3×70 mL). A solution of 1-(chloro-diphenyl-methyl)-4-methyl-benzene (MttCl, 0.79 g, 2.70 mmol) and DIPEA (0.94 mL, 5.4 mmol) in dry DCM (60 mL) was added to resin and mixture was shaken for 3 hrs. The resin was filtered off and washed with DCM (3×70 mL) and DMF (3×70 mL). The IvDde group was removed by treatment with 2% hydrazine monohydrate in DMF (3×3 min, 3×30 mL). The resin was washed with DMF (5×40 mL). A solution of chloroacetic acid (0.51 g, 5.40 mmol), DIC (0.85 mL, 5.40 mmol) and 2,4,6-trimethylpyridine (1.43 mL, 10.8 mmol) in DMF (50 mL) was added to the resin and the mixture was shaken for 2.5 hrs. The resin was filtered off and washed with DMF (3×70 mL) and DCM (3×70 mL). The Mtt group was removed by treatment with 80% 1,1,1,3,3,3-hexafluoro-2-propanol in DCM (2×10 min, 2×30 min, 4×50 mL). The resin was washed with DCM (5×50 mL) and DMF (4×50 mL). A solution of bromoacetic acid (0.38 g, 2.70 mmol) and DIC (0.42 mL, 2.70 mmol) in DMF (50 mL) was added to resin and mixture was shaken for 2.5 hrs. Resin was filtered and washed with DMF (3×70 mL) and DCM (3×70 mL). The product was cleaved from resin by treatment with cleavage cocktail of TFA/TIS/H2O (95:2.5:2.5, 26 mL) for 1 hr. The resin was filtered off and washed with DCM (6×40 mL). Solutions were combined and solvents were evaporated to dryness. The residue was purified by HPLC (Column Gemini C18, 5 um; 50×250 mm; MeCN/H2O 5:95 to 40:60 during 180 min. and 5:95 to 35:65 during 60 min.+0.05% TFA) and freeze-dried to give title compound (2) as white solid. Yield: 336 mg (43%). 1H NMR spectrum (300 MHz, AcOD-d4, δH): 5.07 (dd, J=9.9 and 2.9 Hz, 1H); 4.72 (dd, J=8.7 and 4.7 Hz, 1H); 4.22-4.12 (m, 6H); 3.96 (s, 2H); 3.93-3.37 (m, 16H); 3.33 (t, J=6.8 Hz, 3H); 2.59-2.29 (m, 7H); 2.10 (d, J=4.9 Hz, 2H); 1.97-1.80 (m, 3H). LC-MS purity: 100%. LC-MS Rt (Kinetex 4.6 mm×50 mm, MeCN/H2O 5:95 to 100:0+0.1% FA): 2.86 min. LC-MS m/z: 866.5 (M+H)+. UPLC purity: 98.7% (214 nm). UPLC Rt (Acquity UPLC BEHC 18, 1.7 μm, 2.1×150 mm; MeCN/H2O 5:95 to 100:0+0.05% TFA): 2.74 min. Trivalent Linker 7 (S)-4-(2-{2-[((S)-1-{Bis-[2-(2-chloro-acetylamino)-ethyl]-carbamoyl}-3-carboxy-propylcarbamoyl)-methoxy]-ethoxy}-ethylcarbamoyl)-2-(2-bromo-acetylamino)-butyric acid Synthetic Protocol: Wang resin 0.63 mmol/g (1, 5.08 g, 3.20 mmol) was left to swell in THF (60 mL) for 45 minutes. A solution of (S)-4-(bis-{2-[1-(4,4-dimethyl-2,6-dioxo-cyclohexylidene)-ethylamino]-ethyl}-carbamoyl)-4-(9H-fluoren-9-ylmethoxycarbonylamino)-butyric acid (7.52 g, 9.61 mmol), DIC (1.49 mL, 9.61 mmol) and 4-dimethylaminopyridine (DMAP, 0.04 g, 0.32 mmol) in THF (50 mL) was added to resin and the mixture was shaken overnight. Resin was filtered and washed with DMF (2×50 mL), DCM (2×50 mL) and DMF (2×50 mL). The Fmoc group was removed by treatment with 20% piperidine in DMF (2×50 mL, 1×5 min, 1×20 min). Resin was washed with DMF (2×50 mL), 2-propanol (2×50 mL), DCM (2×50 mL) and DMF (2×50 mL). A solution of {2-[2-(9H-fluoren-9-ylmethoxycarbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 2.47 g, 6.41 mmol), TCTU (2.28, 6.41 mmol) and DIPEA (2.01 mL, 11.5 mmol) in DMF (50 mL) was added to resin and mixture was shaken for 2 hrs. Resin was filtered and washed with DMF (2×50 mL), DCM (2×50 mL) and DMF (2×50 mL). The Fmoc group was removed by treatment with 20% piperidine in DMF (2×50 mL, 1×5 min, 1×20 min). Resin was washed with DMF (2×50 mL), 2-propanol (2×50 mL), DCM (2×50 mL) and DMF (2×50 mL). A solution of (S)-2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 1-tert-butyl ester (Fmoc-Glu-OtBu, 2.04 g, 4.80 mmol), TCTU (1.71, 4.80 mmol) and DIPEA (1.67 mL, 9.61 mmol) in DMF (50 mL) was added to resin and mixture was shaken for 1.5 hrs. Resin was filtered and washed with DMF (2×50 mL), DCM (2×50 mL) and DMF (2×50 mL). The Fmoc group was removed by treatment with 20% piperidine in DMF (2×50 mL, 1×5 min, 1×20 min). Resin was washed with DMF (2×50 mL), 2-propanol (2×50 mL), DCM (2×50 mL) and DMF (2×50 mL). A solution of 1-(chloro-diphenyl-methyl)-4-methyl-benzene (MttCl, 1.13 g, 3.84 mmol) and DIPEA (1.67 mL, 9.61 mmol) in dry DCM (50 mL) was added to resin and mixture was shaken for 2 hrs. Resin was filtered and washed with DCM (4×50 mL) and DMF (4×50 mL). Dde group was removed by treatment with 2% hydrazine in DMF (3×50 mL, 3×3 min). Resin was washed with DMF (8×50 mL). A solution of chloroacetic acid (0.91 g, 9.61 mmol), PyBrOP (4.48 g, 9.61 mmol) and DMF (3.35 mL, 19.2 mmol) in DMF (50 mL) was added to resin and mixture was shaken overnight. Ninhydrin test was still positive, therefore recoupling was made. A solution of chloroacetic acid (0.91 g, 9.61 mmol), PyBrOP (4.48 g, 9.61 mmol) and DIPEA (3.35 mL, 19.2 mmol) in DMF (50 mL) was added to resin and mixture was shaken for 3 hrs. Resin was filtered and washed with DMF (4×50 mL) and DCM (4×50 mL). Mtt group was removed by treatment with 80% 1,1,1,3,3,3-hexafluoro-2-propanol in DCM (7×50 mL, 2×10 min, 5×30 min). Resin was washed with DCM (5×50 mL) and DMF (4×50 mL). A solution of bromoacetic acid (8.90 g, 64.1 mmol) and DIC (8.43 mL, 54.5 mmol) in DMF (50 mL) was added to resin and mixture was shaken for 20 minutes. Resin was filtered and washed with DMF (4×30 mL) and DCM (10×30 mL). The product was cleaved from resin by treatment with TFA (50 mL) for 1.5 hrs. Resin was filtered off and washed with TFA (1×50 mL) and DCM (2×50 mL). Solutions were combined and solvents were evaporated. The residue was co-evaporated with toluene twice and purified by preparative LC/MS (SunFire Prep C18 OBD 5m, 19×100 mm, gradient 5-100% MeCN/H2O in 0.1% FA). Fractions containing pure product were combined and freeze-dried yielding the title compound as beige solid. Yield: 150 mg (30%). 1H NMR spectrum (300 MHz, AcOD-d4, δH): 5.19-5.05 (m, 1H); 4.72-4.56 (m, 1H); 4.27-4.07 (m, 6H); 4.06-3.37 (m, 18H); 2.60-2.37 (m, 4H); 2.36-2.22 (m, 1H); 2.21-1.87 (m, 3H). LC-MS purity: 100%. LC-MS Rt (Kinetex 4.6 mm×50 mm, MeCN/water 05:95 to 100:0+0.1% FA): 2.94 min. LC-MS m/z: 780.4 (M+H)+. Trivalent Linker 8 (4S,18S)-4-(bis(2-(2-Bromoacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-chloroacetamido)ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid Synthetic Protocol: IvDde group from one portion of resin (1) (8.25 g, 2.71 mmol, preparation as described in Example 5) was removed by treatment with 2% hydrazine monohydrate in DMF (3×3 min, 3×50 mL). Resin was washed with DMF (6×50 mL). A solution of bromoacetic acid (2.26 g, 16.3 mmol), DIC (2.52 mL, 16.3 mmol) and 2,4,6-collidine (4.30 mL, 32.5 mmol) in DMF (50 mL) was added to resin and mixture was shaken for 2.5 hrs. Resin was filtered and washed with DMF (3×50 mL), DCM (3×50 mL), DMF (3×50 mL) and DCM (4×50 mL). Mtt group was removed by treatment with 80% 1,1,1,3,3,3-hexafluoro-2-propanol in DCM (2×10 min, 2×30 min, 4×50 mL). Resin was washed with DCM (5×50 mL) and DMF (4×50 mL). A solution of chloroacetic acid (5.12 g, 54.2 mmol) and DIC (7.13 mL, 46.1 mmol) in DMF (50 mL) was added to resin and mixture was shaken for 30 min. Resin was filtered and washed with DMF (4×50 mL) and DCM (10×50 mL). The product was cleaved from resin by treatment with cleavage cocktail of TFA/TIS/H2O (95:2.5:2.5, 50 mL) for 2 hrs. Resin was filtered and washed with TFA/DCM mixture (1:1, 50 mL) and DCM (10×30 mL). Solutions were combined and solvents were evaporated to dryness. The residue was purified by HPLC (column X-Bridge3 C18, OBD, 5 μm, 50×250 mm MeCN/H2O 3:35 to 35:40+0.05% TFA) to give (4S,18S)-4-(bis(2-(2-bromoacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-chloroacetamido)ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid (2) as white solid. Yield: 213 mg (8%). 1H NMR spectrum (300 MHz, AcOD-d4, 80° C., δH): 5.12 (dd, J=9.5 and 3.9 Hz, 1H); 4.69 (dd, J=8.0 and 5.4 Hz, 1H); 4.19-4.10 (m, 6H); 3.94 (s, 2H); 3.90-3.43 (m, 24H); 2.55 (t, J=7.0 Hz, 2H); 2.48 (m, 2H); 2.41-1.99 (m, 4H). LC-MS purity: 100%. LC-MS Rt (Kinetex 4.6 mm×50 mm, MeCN/H2O 5:95 to 100:0+0.1% FA): 3.06 min. LC-MS m/z: 970.5 (M+H)+. UPLC purity: 98.3% (214 nm). UPLC Rt (Acquity UPLC BEHC 18, 1.7 μm, 2.1×150 mm; MeCN/H2O 5:95 to 95:5+0.05% TFA): 1.66 min. Trivalent Linker 9 (4S)-5-[bis[3-[(2-chloroacetyl)amino]propyl]amino]-4-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-oxo-pentanoic acid Trivalent linker 9 was prepared in a similar way as described in Example 1 for Trivalent linker 1 substituting 2,2′-(((azanediylbis(ethane-2,1-diyl))bis(azanediyl))bis(3-methylbutan-1-yl-1-ylidene))bis(5,5-dimethylcyclohexane-1,3-dione) with 2,2′-(((azanediylbis(propane-3,1-diyl))bis(azanediyl))bis(3-methylbutan-1-yl-1-ylidene))bis(5,5-dimethylcyclohexane-1,3-dione). Crude (4S)-5-[bis[3-[(2-chloroacetyl)amino]propyl]amino]-4-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-oxo-pentanoic acid was purified by preparative LC-MS (Column Labio C18, 50×500 mm, MeCN/water+0.05% TFA, gradient 10:40 during 120 min to give pure (4S)-5-[bis[3-[(2-chloroacetyl)amino]propyl]amino]-4-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-oxo-pentanoic acid as colorless oil. Yield: 0.25 g (56%). LC-MS purity: 100% (ELSD). LC-MS Rt (Kinetex C18, 4.6 mm×50 mm, MeCN/H2O 5:95 to 100:0+0.1% FA): 3.01 min. LC-MS m/z: 680.2 (M+H)+. 1H NMR spectrum (300 MHz, CDCl3, δH): 7.71-7.60 (m, 2H); 7.49-7.41 (m, 1H); 7.35-7.29 (m, 1H); 5.17-5.07 (m, 1H); 4.43 (bs, 1H); 4.12-3.98 (m, 6H); 3.90 (s, 2H); 3.76-3.57 (m, 8H); 3.50-3.14 (m, 8H); 2.50-2.42 (m, 2H); 2.10-1.70 (m, 6H). Trivalent Linker 10 N-[2-[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]ethylcarbamoyl-[2-[(2-chloroacetyl)amino]ethyl]amino]ethyl]-2-chloro-acetamide Synthetic Protocol: Wang-OH resin 0.53 mmol/g (1, 3.85 g, 2.04 mmol) was left to swell in dry DCM (40 mL) for 30 min. A solution of 4-nitrophenyl carbonochloridate (2, 0.27 g, 1.36 mmol) and pyridine (0.12 mL, 1.50 mmol) in dry DCM (40 mL) was added to resin (1) and mixture was shaken overnight. Resin (3) was washed with ice water (1×30 mL), ice water/1,4-dioxane mixture (1:1, 1×30 mL), DMF (3×30 mL) and DCM (3×30 mL). A solution of 2,2′-(ethane-1,2-diylbis(oxy))bis(ethan-1-amine) (4) (0.59 mL, 4.08 mmol) in DMF (30 mL) was added to resin (3) and mixture was shaken overnight. Resin (5) was washed with DMF (4×30 mL), DCM (3×30 mL), DMF (2×30 mL) and DCM (2×30 mL). A solution of 4-nitrophenyl carbonochloridate (2) (1.10 g, 5.44 mmol) and N,N-diisopropylethylamine (1.88 mL, 6.80 mmol) in DMF/tetrahydrofuran mixture (1:1, 30 mL) was added to resin (5) and mixture was shaken for 2 hrs. Resin (6) was washed with DMF (4×30 mL), DCM (4×30 mL) and DMF (4×30 mL). A solution of 2,2′-(((azanediylbis(ethane-2,1-diyl))bis(azanediyl))bis(3-methylbutan-1-yl-1-ylidene))bis(5,5-dimethylcyclohexane-1,3-dione) (7) (1.40 g, 2.72 mmol) and N,N-diisopropylethylamine (0.47 mL, 2.72 mmol) in DMF (30 mL) was added to resin (6) and mixture was shaken for 2 hrs. Resin (8) was washed with DMF (4×30 mL), DCM (4×30 mL) and DMF (4×30 mL). The ivDde-protecting groups were removed by treatment with 2% solution of hydrazine monohydrate in DMF (3×5 min, 3×30 mL). Resin was washed with DMF (3×30 mL), DCM (3×30 mL) and DMF (3×30 mL). Solution of chloroacetic anhydride (0.93 g, 5.44 mmol) and N,N-diisopropylethylamine (1.90 mL, 10.9 mmol) in DMF (30 mL) was added to resin and the mixture was shaken for 1 hr. Resin was filtered and washed with DMF (3×30 mL), DCM (3×30 mL), DMF (3×30 mL) and DCM (10×30 mL). The product (9) was cleaved from resin by treatment with 95% trifluoroacetic acid in water (30 mL) for 3 hrs. Resin was filtered off and washed with DCM (3×10 mL). Solutions were combined and solvents were evaporated to dryness. The residue was dissolved in 60% aqueous solution of MeCN (50 mL) and freeze-dried to give the desired compound (9) as a thick yellow oil. Yield: 240 mg (42%). LC-MS purity: 93% (ELSD). LC-MS Rt (Kinetex C18, 4.6 mm×50 mm, MeCN/H2O 5:95 to 100:0+0.1% FA): 2.42 min. LC-MS m/z: 430.29 (M+H)+. A stirred solution of the above compound (9) (0.24 g, 0.56 mmol) in MeCN (20 mL) was cooled at 0° C. and bromoacetic anhydride (0.22 g, 0.84 mmol) and sodium bicarbonate (0.18 g, 2.09 mmol) were added. The reaction mixture was stirred for 1 hr at 0° C. Freeze-drying of the reaction mixture gave the title crude compound (10) as colorless oil. The crude compound (10) was twice purified by preparative LC/MS (SunFire Prep C18 OBD, 5 μm, 19×100 mm, MeCN/H2O 5:95 to 100:0+0.1% FA) to give title compound N-[2-[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]ethylcarbamoyl-[2-[(2-chloroacetyl)amino]ethyl]amino]ethyl]-2-chloro-acetamide as colorless oil. Yield: 35.0 mg (12%). 1H NMR spectrum (300 MHz, CDCl3, δH): 7.45 (bs, 2H); 7.27 (bs, 1H); 6.06-5.93 (m, 1H); 4.06 (s, 4H); 3.90 (s, 2H); 3.64 (s, 4H); 3.62-3.55 (m, 4H); 3.53-3.38 (m, 12H). LC-MS purity: 100% (ELSD). LC-MS Rt (Kinetex C18, 4.6 mm×50 mm, MeCN/H2O 5:95 to 100:0+0.1% FA): 2.91 min. LC-MS m/z: 552.3 (M+H)+. Trivalent Linker 11 (2R)-6-[bis[2-[(2-chloroacetyl)amino]ethyl]carbamoylamino]-2-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]hexanoic acid Trivalent linker 11 was prepared in a similar way as described for Trivalent linker 10. Trivalent Linker 12 (2R)-2-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-[2-[2-[2-[[(1S)-3-carboxy-1-[2-[(2-chloroacetyl)-[2-[(2-chloroacetyl)amino]ethyl]amino]ethylcarbamoyl]propyl]amino]-2-oxo-ethoxy]ethoxy]ethylamino]-5-oxo-pentanoic acid Synthetic Protocol: Wang resin-bound compound (1) (1.42 g, 0.70 mmol) was left to swell in DCM (20 mL) for 20 min. Fmoc group was removed by treatment with 20% piperidine in DMF (2×5 min, 1×20 min, 3×10 mL). Resin was washed with DMF (3×15 mL), 2-propanol (3×15 mL) and DCM (3×15 mL). A solution of {2-[2-(9H-fluoren-9-ylmethoxycarbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 0.77 g, 2.00 mmol), 5-chloro-1-((dimethylamino)(dimethyliminio)methyl)-1H-benzo[d][1,2,3]triazole 3-oxide tetrafluoroborate (TCTU, 0.71 g, 2.00 mmol) and N,N-diisopropylethylamine (0.63 mL, 3.60 mmol) in DMF (10 mL) was added to resin and the mixture was shaken for 2 hrs. Resin was filtered and washed with DMF (3×15 mL), DCM (3×15 mL) and DMF (3×15 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (2×5 min, 1×20 min, 3×10 mL). Resin was washed with DMF (3×15 mL), 2-propanol (3×15 mL) and DCM (3×15 mL). Solution of (S)-2-(9H-fluoren-9-ylmethoxycarbonylamino)-pentanedioic acid 1-tert-butyl ester (Fmoc-L-Glu-OtBu, 0.85 g, 2.00 mmol), 5-chloro-1-((dimethylamino)(dimethyliminio)methyl)-1H-benzo[d][1,2,3]triazole 3-oxide tetrafluoroborate (TCTU, 0.71 g, 2.00 mmol) and N,N-diisopropylethylamine (0.63 mL, 3.60 mmol) in DMF (10 mL) was added to resin and mixture was shaken for 2 hrs. Resin was filtered and washed with DMF (3×15 mL), DCM (3×15 mL) and DMF (3×15 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (2×5 min, 1×20 min, 3×10 mL). Resin was washed with DMF (3×15 mL), 2-propanol (3×15 mL) and DCM (3×15 mL). A solution of {2-[2-(9H-fluoren-9-ylmethoxycarbonylamino)-ethoxy]-ethoxy}-acetic acid (Fmoc-OEG-OH, 0.77 g, 2.00 mmol), 5-chloro-1-((dimethylamino)(dimethyliminio)methyl)-1H-benzo[d][1,2,3]triazole 3-oxide tetrafluoroborate (TCTU, 0.71 g, 2.00 mmol) and N,N-diisopropylethylamine (0.63 mL, 3.60 mmol) in DMF (10 mL) was added to resin and the mixture was shaken for 2 hrs. Resin was filtered and washed with DMF (3×15 mL), DCM (3×15 mL) and DMF (3×15 mL). Fmoc group was removed by treatment with 20% piperidine in DMF (2×5 min, 1×20 min, 3×10 mL). Resin was washed with DMF (3×15 mL), 2-propanol (3×15 mL) and DCM (3×15 mL). A solution of di-tert-butyl dicarbonate (0.44 g, 2.00 mmol) and N,N-diisopropylethylamine (0.35 mL, 2.00 mmol) dissolved in DCM (10 mL) was added to resin and the mixture was shaken for 2 hrs. Resin was washed with DMF (3×15 mL), DCM (3×15 mL) and DMF (3×15 mL). Resin was treated with hydrazine monohydrate solution in DMF (2% v/v solution, 3×10 min, 3×15 mL). Resin (2) was washed with DMF (3×15 mL), DCM (3×15 mL) and DMF (3×15 mL). Resin (2) was treated with solution of acetic acid (0.12 mL, 2.00 mmol) and N,N-diisopropylethylamine (0.34 mL, 2.00 mmol) in DMF (10 mL) for 16 hrs. Resin was washed with DMF (3×15 mL), N,N-diisopropylethylamine (0.34 mL, 2.00 mmol) in DMF (10 mL), DCM (2×15 mL) and DMF (3×15 mL). Chloroacetic acid (0.53 g, 5.60 mmol), N,N′-diisopropylcarbodiimide (0.86 mL, 5.60 mmol) and 2,4,6-collidine (0.74 mL, 5.60 mmol) was added and the mixture was shaken for 2 hrs. Resin was washed with DMF (3×15 mL), DCM (6×15 mL). The product was cleaved from resin by treatment with trifluoroacetic acid/water mixture (98:2, 20 mL) for 2 hrs. The solution was concentrated in vacuo and the residue of trifluoroacetic acid was removed by co-evaporation with toluene. A solution of bromoacetic acid (276 mg, 2.00 mmol) and N,N′-dicyclohexylcarbodiimide (DCC, 0.21 mL, 1.00 mmol) was dissolved in MeCN (5 mL), stirred for 15 min and filtered. This solution was added to the crude product cleaved from the resin cooled to 0° C. and sodium hydrogencarbonate (0.24 mmol, 2.80 mmol) was added. The mixture was stirred and let worm to 25° C. for 3 hrs. The mixture was filtered, evaporated and crude product was purified by preparative HPLC (Gemini NX C18, 5 μm, 50×250 mm, MeCN/H2O 5:95 to 45:55 during 180 min and 45:55 to 50:50 during 10 min+0.05% TFA) to give title compound (2R)-2-[[2-[2-[2-[(2-bromoacetyl)amino]ethoxy]ethoxy]acetyl]amino]-5-[2-[2-[2-[[(1S)-3-carboxy-1-[2-[(2-chloroacetyl)-[2-[(2-chloroacetyl)amino]ethyl]amino]ethylcarbamoyl]propyl]amino]-2-oxo-ethoxy]ethoxy]ethylamino]-5-oxo-pentanoic acid Yield: 33 mg (5%). 1H NMR spectrum (300 MHz, AcOD-d4, δH): 4.74-4.55 (m, 2H); 4.33 (s, 1H); 4.31 (s, 1H); 4.23-4.10 (m, 6H); 3.98 (s, 2H); 3.83-3.42 (m, 24H); 2.59-2.41 (m, 4H); 2.38-2.08 (m, 4H). LC-MS purity: 100% (ELSD). LC-MS Rt (Kinetex C18, 4.6 mm×50 mm, MeCN/water 5:95 to 100:0+0.1% TFA): 3.04 min. LC-MS m/z: 927.0 (M+H)+. Example 2 GH-Fc Conjugate 1 GH-trivalent linker 1 intermediate [(bis(2-(2-Chloroacetamido)ethylamine)))-Glu-OEG-γGlu-OEG]-carbonylmethylene-S101-hGH[L101C] Step 1 Preparation of hGH[L101C]: hGH[L101C] as obtained above had part of its free cysteine blocked with glutathione and cystamine. Deblocking was performed enzymatically using glutaredoxin II (Grx2) in an equilibrium buffer containing GSH and GSSG. Deblocked hGH[L101C] was separated from low molecular weight GSH/GSSG by buffer exchanged on a Sephadex G25 column. hGH[101C] used: 100 mg (20.2 mL, Mw=22190.93 Fc used: 200 mg (100 mL in 50 mM ammoniumbicarbonate pH 7.8). Concentration 2.03 mg/mL Procedure (step 1): To a vial containing hGH[L101C] (50 mg, 4.95 mg/mL, 225 μM, in 20 mM triethylamine, 100 mM NaCl, pH 8) was added 3 eq. bis(p-sulfonatophenyl)phenylphosphine dihydrate dipotassium (TPPDS, 3.6 mg) at room temperature. After 1 hr incubation, 3 eq. of (4S,18S)-4-(bis(2-(2-chloroacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-bromoacetamido)-ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid (from Example 1, 6.3 mg) along with NaCl (177 mg giving 0.3 M NaCl final concentration) were added and the resulting reaction mixture allowed to incubate at room temperature for 18 hrs, whereupon it was buffer exchanged into 20 mM HEPES, 10 mM EDTA, pH 7.5 and up-concentrated to 22 mg/mL by ultrafiltration (2.2 mL). Yield=50 mg (99%). LC-MS (electrospray): Found m/z=22959,43; Calculated m/z=22959,48 Purity on HPLC: 93% at 214 nm. System: Agilent 1200 series HPLC Column: Zorbax 300SB-C3, 4.6×50 mm, 3.5μ Detector: Agilent Technologies LC/MSD TOF (G1969A) Detector setup: DAD: 280 nm, (G1315A) Scanning range: m/z min. 100, m/z max. 3000 Linear reflector mode Positive mode Conditions: Step gradient: 5% to 90% B Run-time: 12 minutes: 0-1 min 5% B, 1-8 min 5-90% B, 8-9 min 90% B, 9-9.1 min 90-5% B 9.1-12 min 5% B Flow rate: 1.00 mL/min fixed Column temperature: 40° C. Eluents: Solvent A: 99.9% H2O, 0.1% Formic Acid Solvent B: 99.9% MeCN, 0.1% Formic Acid Reaction overview of step 2 including 3 separate steps describe below [(bis(2-(2-Fc-S3-acetamido)ethylamine)))-Glu-OEG-γGlu-OEG]-carbonylmethylene-S101-hGH[L101C] Procedure for step 2: 1. Chloro to Iodo exchange (Finkelstein reaction): The above compound [(bis(2-(2-Chloroacetamido)ethylamine)))-Glu-OEG-γGlu-OEG]-carbonylmethylene-S101-hGH[L101C] from step 1. (2.2 mL, 22 mg/mL, 956 μM) was diluted with 2.2 mL of an aq. 5M KI, 50 mM ascorbic acid solution and incubated at room temperature for 18 hrs in the dark. Finally, the reaction mixture was buffer exchanged in 20 mM HEPES, 10 mM EDTA, pH 7.5 buffer (2.3 mL, 21.7 mg/mL, 945 μM) and used directly in step 3. below. 2. Fc-fragment disulphide bridge reduction: To the Fc-fragment obtained as described above (50 mL, 2.03 mg/mL, 41 μM in 50 mM ammonium bicarbonate, pH 7.8) was added 5 eq. dithiothreitol (DTT, 52 μL of a 195 mM solution in 20 mM HEPES, 10 mM EDTA, pH 7.5) and incubated for 2 hrs at room temperature whereupon the reaction mixture was buffer exchanged and up-concentrated to 4.3 mL (23.6 mg/mL, 475 μM as dimer) by ultrafiltration and used directly in step 3. below. 3. hGH[L101C]-Fc conjugate formation: The hGH[L101C] compound from step 1. (50 mg, 21.7 mg/mL, 945 μM) was mixed with reduced Fc-fragment from step 2. (100 mg, 23.6 mg/mL, 475 μM) obtaining a molar ratio between hGH[L101C] and Fc of 1.1 to 1. The reaction mixture (6.6 mL) was allowed to incubate in the dark for 18 hrs whereupon the desired conjugate was purified from the reaction mixture on a Capto Adhere 16/10 column operated in HIC mode (CV=20 mL; A: 20 mM TEA, pH 7.5; B: 40 mM MES, 40 mM formic acid, pH 3.5; application buffer: 20 mM TEA, 200 mM NaCl, pH 7.5; segment gradient: segment 1: 0-30% B, 1 CV; segment 2: 30-70% B, 15 CV; segment C: 70-100% B, 1 CV; flow 3 mL/min). Fractions containing product were buffer exchanged in PBS giving 50 mg of the desired conjugate (25 mL, 2.0 mg/mL, 27.5 μM). Yield=50 mg (34%). LC-MS (electrospray): Found m/z=72682.09; Calculated m/z=72682.13 Purity on HPLC: 96% at 214 nm. GH-Fc Conjugate 2 [(bis(2-(2-Fc-S3-acetamido)ethylamine)))-Gly]-carbonylmethylene-S101-hGH[L101C] The compound was prepared using the method as described in Example 2 conjugate 1 except that the linker (4S,18S)-4-(bis(2-(2-chloroacetamido)-ethyl)carbamoyl)-18-(2-(2-(2-(2-bromoacetamido)-ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid (Example 1, linker 1) was substituted with 2-(2-bromoacetamido)-N,N-bis(2-(2-chloroacetamido) ethyl)acetamide (Example 1, linker 2) 100 mg in HEPES/EDTA Purification: The above reaction mixture was buffer changed to loading buffer (TRIS+salt) and loaded on a G25 column: Column: 50/30 Sephadex G25 fine Buffer A: 10 mM Ammonium bicarbonate Flow: 10 mL/min Temp: RT Fractions: 40 mL per fraction Fraction A4+A5 were pooled and applied onto a Capto Adhere column: Column: Capto Adhere 16/10 Buffer A: 20 mM TEA pH 7.5 Buffer A2: 40 mM TEA+0,2M NaCl pH 7.5 Buffer B: 40 mM MES+40 mM Formic acid pH 3.5 Gradient 1: 0-30% Buffer B over 1 CV Gradient 2: 30-70% Buffer B over 15 CV Gradient 3: 70-100% Buffer B over 1 CV Flow: 3 mL/min Temp: RT Fractions: 1 mL per fraction in peak fractionation Fractions A6-A10 were pooled and buffer changed to PBS buffer by UF (Amicon ultra 15K). Concentration=>7.22 mg/mL=>101 mg in total Yield: 101 mg (31%) LC-MS (electrospray): Found m/z=72193.48; Calculated m/z=72190.6444 Purity on HPLC: ˜100% at 214 nm. GH-FC Conjugate 3 [(bis(2-(2-Fc-S3-acetamido)ethylamine)))-Gly-OEG]-carbonylmethylene-S101-hGH[L101C] The compound was prepared using the method as described in Example 2 conjugate 1 except that the linker (4S,18S)-4-(bis(2-(2-chloroacetamido)-ethyl)carbamoyl)-18-(2-(2-(2-(2-bromoacetamido)-ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid (Example 1) was substituted with 2-(2-(2-(2-(2-bromoacetamido)ethoxy)ethoxy)acetamido)-N,N-bis(2-(2-chloroacetamido)-ethyl)acetamide (Example 1, linker 3) 100 mg in HEPES/EDTA hGH[L101C] used: 100 mg (20.2 mL) as described above. Mw=22190.93 Fc used: 200 mg (100 mL in 50 mM ammoniumbicarbonate pH 7.8). Concentration 2.03 mg/mL Purification: The above reaction mixture was buffer changed to loading buffer (TRIS+salt) and loaded on a G25 column: Column: 50/30 Sephadex G25 fine Buffer A: 10 mM Ammonium bicarbonate Flow: 10 mL/min Temp: RT Fractions: 40 mL per fraction Fractions A4+A5 were pooled and applied onto a Capto Adhere column: Column: Capto Adhere 16/10 Buffer A: 20 mM TEA pH 7.5 Buffer A2: 40 mM TEA+0.2M NaCl pH 7.5 Buffer B: 40 mM MES+40 mM Formic acid pH 3.5 Gradient 1: 0-30% Buffer B over 1 CV Gradient 2: 30-65% Buffer B over 15 CV Gradient 3: 65-100% Buffer B over 1 CV Flow: 3 mL/min Temp: RT Fractions: 3 mL per fraction in peak fractionation Fractions A4-A9 were pooled and buffer changed to PBS buffer by UF (Amicon ultra 15K) affording the desired hGH-linker-Fc conjugate [(bis(2-(2-Fc-S3-acetamido)ethylamine)))-Gly-OEG]-carbonylmethylene-S101-hGH[L101C]. Yield: 91 mg (28%) LC-MS (electrospray): Found m/z=72338.69; Calculated m/z=72335.8008 Purity on HPLC: 81% at 214 nm. GH-FC Conjugate 4 In a similar way as described above for conjugates 1-3 a conjugate 4 was prepared using trivalent linker 4 of Example 1. GH-FC Conjugate 5 In a similar way as described above for conjugates 1-3 a conjugate was prepared using trivalent linker 6 of Example 1. GH-Fc Conjugate 1 by Alternative Method Fc-Linker Intermediate Step 1 Procedure Step 1: To the Fc-fragment obtained as described above (12 mL, 2.03 mg/mL, 41 μM in 50 mM ammonium bicarbonate, pH 7.8) was up-concentrated with a vivaspin UF device (CU 30 kDa, PES membrane) to 1.9 mL (22 mg, 11.6 mg/mL, 232 μM) was added 220 μL of a 10 mM TCEP solution in PBS (4.3 eq) and incubated for 1 hrs at room temperature. The reaction mixture was buffer exchanged into 50 mM phosphate buffer, 400 mM NaCl, 10 mM EDTA to get 19.25 mg of reduced Fc (7.7 mg/mL, 154 μM as dimer) to which 375 uL of a 5 mM freshly prepared solution of (4S,18S)-4-(bis(2-(2-Bromoacetamido)ethyl)carbamoyl)-18-(2-(2-(2-(2-chloroacetamido)ethoxy)ethoxy)acetamido)-6,15-dioxo-8,11-dioxa-5,14-diazanonadecanedioic acid in the same buffer was added (2.5 eq) and allowed to incubate in the dark for 18 hrs at rt whereupon the reaction mixture was buffer exchanged by gel filtration into PBS to furnish the target Fc-linker conjugate quantitatively. Yield=50 mg (95%). LC-MS (electrospray): Found m/z=50605.58; Calculated m/z=50605,68 Purity on HPLC: 92% at 214 nm. System: Agilent 1200 series HPLC Column: Zorbax 300SB-C3, 4.6×50 mm, 3.5μ Detector: Agilent Technologies LC/MSD TOF (G1969A) Detector setup: DAD: 280 nm, (G1315A) Scanning range: m/z min. 100, m/z max. 3000 Linear reflector mode Positive mode Conditions: Step gradient: 5% to 90% B Run-time: 12 minutes: 0-1 min 5% B, 1-8 min 5-90% B, 8-9 min 90% B, 9-9.1 min 90-5% B 9.1-12 min 5% B Flow rate: 1.00 mL/min fixed Column temperature: 40° C. Eluents: Solvent A: 99.9% H2O, 0.1% Formic Acid Solvent B: 99.9% MeCN, 0.1% Formic Acid Reaction Overview of Step 2 Including 3 Separate Steps as Described Below. Procedure (Step 2): 1. Chloro to Iodo exchange (Finkelstein reaction): The above compound Fc-linker conjugate from step 1. (2.5 mL, 7.7 mg/mL, 152 μM) was buffer exchange by gel filtration in a Zeba spin column (10 mL size, Pierce) into 50 mM phosphate buffer, 5 M KI, 50 mM ascorbic acid, 100 mM NaCl, pH 6 and incubated at rt in the dark for 18 hrs. Finally, the reaction mixture was up-concentrated with a vivaspin UF device (CU 30 kDa, PES membrane) to 1.3 mL and buffer exchange by gel filtration in a Zeba spin column (10 mL size, Pierce) into 50 mM PB, 1 M NaCl, 10 mM EDTA, pH 7.6 and used immediately in step 3. 2. hGH[L101C] decappinq of cysteine: To a vial containing hGH[L101C] (24.75 mg, 4.95 mg/mL, 225 μM, in 20 mM triethylamine, 100 mM NaCl, pH 8) was added 3 eq. bis(p-sulfonatophenyl)phenylphosphine dihydrate dipotassium (TPPDS, 3.6 mg) and incubated at room temperature for 18 hrs. Following, the reaction mixture was buffer exchanged into 50 mM PB, 1 M NaCl, 10 mM EDTA, pH 7.6 and up-concentrated to 17 mg/mL by ultrafiltration (1.45 mL) and used immediately in step 3. 3. hGH[L101C]-Fc conjugate formation: Fc-activated linker conjugate from step 1 (1.3 mL, 14.8 mg/mL, 300 μM) and the free Cys hGH[L101C] compound from step 2 (0.61 mL, 17 mg/mL, 770 μM) were mixed together, obtaining a molar ratio between Fc and hGH[101] of 1 to 1.2. The reaction mixture (1.91 mL) was allowed to incubate in the dark for 18 hrs whereupon the desired conjugate was purified from the reaction mixture on a Capto Adhere 16/10 column operated in HIC mode (CV=20 mL; A: 20 mM TEA, pH 7.5; B: 40 mM MES, 40 mM formic acid, pH 3.5; application buffer: 20 mM TEA, 200 mM NaCl, pH 7.5; segment gradient: segment 1: 0-30% B, 1 CV; segment 2: 30-70% B, 15 CV; segment C: 70-100% B, 1 CV; flow 3 mL/min). Fractions containing product were buffer exchanged in PBS giving 8.6 mg of the desired conjugate (4.3 mL, 2.0 mg/mL, 27.5 μM). Yield=8.6 mg (30%). LC-MS (electrospray): Found m/z=72682.09; Calculated m/z=72682.13 Purity on HPLC: 94% at 214 nm. Example 3 Evaluation of GH Compounds The GH compounds produced according to above example 2 were evaluated as described in Assay 2, 3 and 5. All compounds were administered intravenously and the mean residence time (MRT) calculated. IGF-1 Plasma concentration-time profiles were generated for each compound. IGF-1 AUC GH Compound RoA MRT (h) hr * ng/mL WT i.v. 0.15 — 1 i.v. 16.9 26273 2 i.v. 23.4 37897 3 i.v. 16.6 39956 4 i.v. 23.0 37689 5 i.v. 19.7 24211 15763857 novo nordisk a/s USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open Apr 5th, 2022 04:33PM Apr 5th, 2022 04:33PM Novo Nordisk Health Care Pharmaceuticals & Biotechnology
nyse:nvo Novo Nordisk Apr 5th, 2022 12:00AM Dec 28th, 2017 12:00AM https://www.uspto.gov?id=US11291777-20220405 Needle cannula, an injection needle assembly for an injection device and an injection device comprising such assembly A needle cannula for an injection device, extends along a central axis and comprises: a) a skin piercing distal end (133b), b) a proximal end (133a) configured for penetrating a seal structure (102b) of a medicament container (102), and c) a sidewall extending between the skin piercing distal end (133b) and the proximal end (133a), wherein the proximal end (133a) is defined by an angled surface (134) pointing proximally, the angled surface (134) defining a leading portion (134.1) and a heel portion (134.2), and wherein the angled surface (134) comprises a surface portion (134a, 134b) at the heel portion (134.2) forming an angle (α1) between 50 to 75 degrees with respect to the central axis, and further comprises a surface portion (134c) at the leading portion (134.1) forming an angle (α2) between 60 to 85 degrees with respect to the central axis. An injection needle assembly for an injection device and an injection device comprising such assembly are further described. 11291777 1. An elongated tubular needle cannula for an injection device, the needle cannula extending along a central axis and comprising: a skin piercing distal end, a proximal end arranged axially opposite the skin piercing distal end, the proximal end being configured for penetrating a seal structure of a medicament container, a sidewall extending between the skin piercing distal end and the proximal end, and a lumen extending through the needle cannula along said central axis, said lumen has a first opening arranged axially at the skin piercing distal end and a second opening arranged axially at the proximal end, wherein the proximal end being defined by an angled surface pointing proximally, the angled surface intersecting with the sidewall at a proximal-most point defining a leading portion and intersecting with the sidewall at an oppositely arranged trailing portion defining a heel portion, wherein the angled surface pointing proximally comprises a surface portion at the heel portion forming an angle (α1) between 50 to 75 degrees with respect to the central axis, and further comprises a surface portion at the leading portion forming an angle (α2) between 60 to 85 degrees with respect to the central axis, wherein said angled surface intersects with the sidewall to define a radially inwards facing circumferential edge and a radially outwards facing circumferential edge, and wherein the radially inwards facing circumferential edge and/or the radially outwards facing circumferential edge is/are blunted along an annular section symmetrically disposed around the heel portion, said annular section having an annular width within 100 to 220 degrees. 2. The elongated tubular needle cannula as defined in claim 1, wherein said surface portion at the heel portion defines at least 20% of said angled surface, and wherein said surface portion at the leading portion defines at least 30% of said angled surface. 3. The elongated tubular needle cannula as defined in claim 1, wherein edges arranged at the leading portion are sharpened. 4. The elongated tubular needle cannula as defined in claim 1, wherein the surface portion at the leading portion forms an angle (α2) with respect to the central axis which is 5 to 10 degrees greater than the angle (α1) of surface portion at the heel portion with respect to the central axis. 5. The elongated tubular needle cannula as defined in claim 1, wherein said surface portion at the heel portion forms an angle (α1) between 55 to 70 degrees with respect to the central axis. 6. The elongated tubular needle cannula as defined in claim 1, wherein the needle cannula is configured for subcutaneous injection, and wherein the needle cannula is a 26 G needle or smaller. 7. The elongated tubular needle cannula as defined in claim 6, wherein the needle cannula is a 27 G needle or smaller. 8. The elongated tubular needle cannula as defined in claim 6, wherein the needle cannula is a 28 G needle or smaller. 9. The elongated tubular needle cannula as defined in claim 6, wherein the needle cannula is a 29 G needle or smaller. 10. The elongated tubular needle cannula as defined in claim 6, wherein the needle cannula is a 30 G needle or smaller. 11. The elongated tubular needle cannula as defined in claim 1, wherein the sidewall of the needle cannula forms an elongated tubular enclosure that does not include opening(s) other than said first opening and said second opening. 12. The elongated tubular needle cannula as defined in claim 1, wherein said annular section has an annular width within 140 to 190 degrees. 13. An injection needle assembly for an injection device, the injection needle assembly comprising: a needle hub, an elongated tubular needle cannula as defined in claim 1, and a first needle cover, wherein the needle hub holds the needle cannula between the skin piercing distal end and the proximal end such that a distal portion of the needle cannula extends distally from the needle hub and such that a proximal portion of the needle cannula extends proximally from the needle hub towards the proximal end of the needle cannula, and wherein the first needle cover is mounted on the needle hub and forms an axially extending elongated flexible enclosure which sealingly accommodates the proximal portion of the needle cannula, wherein the first needle cover has a needle hub end mounted relative to the needle hub and a free end extending beyond the proximal end of the needle cannula, the first needle cover being configured to axially collapse and become penetrated by the proximal portion of the needle cannula when a distally directed penetration force is applied on the free end of the first needle cover urging the free end of the first needle cover towards the needle hub. 14. The injection needle assembly as defined in claim 13, wherein the first needle cover is made from silicone rubber. 15. The injection needle assembly as defined in claim 13, further comprises a second needle cover, wherein the second needle cover is mounted on the needle hub and forms an axially extending elongated flexible enclosure which sealingly accommodates the distal portion of the needle cannula, wherein the second needle cover has a needle hub end mounted relative to the needle hub and a free end extending beyond the distal end of the needle cannula, the second needle cover being configured to axially collapse and become penetrated by the distal portion of the needle cannula when a proximally directed penetration force is applied on the free end of the second needle cover urging the free end of the second needle cover towards the needle hub. 16. An injection device, comprising: a housing defining a distal drug expelling end and an opposite proximal end, an injection needle assembly as defined in claim 13, the injection needle assembly arranged relative to the housing with the proximal end of the needle cannula pointing towards the proximal end of the housing, a medicament container containing a medicament, the medicament container comprising a cylindrical body extending along a longitudinal axis arranged coaxially with the central axis of the needle cannula, the medicament container further comprising a seal structure adapted to become penetrated by the proximal portion of the needle cannula enabling expelling of medicament from the medicament container, wherein the medicament container is arranged axially movable relative to the injection needle assembly for moving the medicament container from a first state where the first needle cover and the seal structure are not penetrated by the needle cannula and into a second state where the proximal portion of the needle cannula has penetrated the first needle cover and the seal structure, and an expelling assembly comprising a strained spring configured to act on the medicament container, wherein the expelling assembly is activatable to enable the strained spring to move the medicament container from the first state to the second state. 17. The injection device as defined in claim 16, wherein, upon activation of the expelling assembly, the proximal portion of the needle cannula penetrates the seal structure of the medicament container by a relative axial movement with no relative rotation between the needle cannula and the seal structure. 18. The injection device as defined in claim 16, wherein the injection needle assembly comprises a second needle cover mounted on the needle hub and forming an axially extending elongated flexible enclosure which sealingly accommodates the distal portion of the needle cannula, wherein the second needle cover has a needle hub end mounted relative to the needle hub and a free end extending beyond the distal end of the needle cannula, the second needle cover being configured to axially collapse and become penetrated by the distal portion of the needle cannula when a proximally directed penetration force is applied on the free end of the second needle cover urging the free end of the second needle cover towards the needle hub, wherein the injection device comprises a needle shield arranged to shield the second needle cover, and wherein the needle shield and the injection needle assembly is arranged to axially move relative to each other for causing the distal portion of the needle cannula to penetrate the second needle cover and enable the distal portion of the needle cannula to extend beyond a distal end portion of the needle shield. 19. An injection device as defined in claim 16, wherein the seal structure of the medicament container is provided as, or comprises, a non-slit medicament container septum. 20. An elongated tubular needle cannula for an injection device, the needle cannula extending along a central axis and comprising: a skin piercing distal end, a proximal end arranged axially opposite the skin piercing distal end, the proximal end being configured for penetrating a seal structure of a medicament container, a sidewall extending between the skin piercing distal end and the proximal end, and a lumen extending through the needle cannula along said central axis, said lumen has a first opening arranged axially at the skin piercing distal end and a second opening arranged axially at the proximal end, wherein the proximal end being defined by an angled surface pointing proximally, the angled surface intersecting with the sidewall at a proximal-most point defining a leading portion and intersecting with the sidewall at an oppositely arranged trailing portion defining a heel portion, wherein the angled surface pointing proximally comprises a surface portion at the heel portion forming an angle (α1) between 50 to 75 degrees with respect to the central axis, and further comprises a surface portion at the leading portion forming an angle (α2) between 60 to 85 degrees with respect to the central axis, wherein the surface portion at the leading portion forms an angle (α2) with respect to the central axis which is 5 to 10 degrees greater than the angle (α1) of surface portion at the heel portion with respect to the central axis. 21. The elongated tubular needle cannula as defined in claim 20, wherein said surface portion at the heel portion defines at least 20% of said angled surface, and wherein said surface portion at the leading portion defines at least 30% of said angled surface. 22. The elongated tubular needle cannula as defined in claim 20, wherein said angled surface intersects with the sidewall to define a radially inwards facing circumferential edge and a radially outwards facing circumferential edge, and wherein the radially inwards facing circumferential edge and/or the radially outwards facing circumferential edge is/are blunted along an annular section symmetrically disposed around the heel portion, said annular section having an annular width within 100 to 220 degrees. 23. The elongated tubular needle cannula as defined in claim 22, wherein said annular section has an annular width within 140 to 190 degrees. 24. The elongated tubular needle cannula as defined in claim 20, wherein edges arranged at the leading portion are sharpened. 25. The elongated tubular needle cannula as defined in claim 20, wherein said surface portion at the heel portion forms an angle (α1) between 55 to 70 degrees with respect to the central axis. 26. The elongated tubular needle cannula as defined in claim 20, wherein the needle cannula is configured for subcutaneous injection, and wherein the needle cannula is a 26 G needle or smaller. 27. The elongated tubular needle cannula as defined in claim 20, wherein the sidewall of the needle cannula forms an elongated tubular enclosure that does not include opening(s) other than said first opening and said second opening. 28. The elongated tubular needle cannula as defined in claim 26, wherein the needle cannula is a 28 G needle or smaller. 29. The elongated tubular needle cannula as defined in claim 26, wherein the needle cannula is a 29 G needle or smaller. 30. The elongated tubular needle cannula as defined in claim 26, wherein the needle cannula is a 30 G needle or smaller. 31. The elongated tubular needle cannula as defined in claim 26, wherein the needle cannula is a 27 G needle or smaller. 32. An injection needle assembly for an injection device, the injection needle assembly comprising: a needle hub, an elongated tubular needle cannula as defined in claim 20, and a first needle cover, wherein the needle hub holds the needle cannula between the skin piercing distal end and the proximal end such that a distal portion of the needle cannula extends distally from the needle hub and such that a proximal portion of the needle cannula extends proximally from the needle hub towards the proximal end of the needle cannula, and wherein the first needle cover is mounted on the needle hub and forms an axially extending elongated flexible enclosure which sealingly accommodates the proximal portion of the needle cannula, wherein the first needle cover has a needle hub end mounted relative to the needle hub and a free end extending beyond the proximal end of the needle cannula, the first needle cover being configured to axially collapse and become penetrated by the proximal portion of the needle cannula when a distally directed penetration force is applied on the free end of the first needle cover urging the free end of the first needle cover towards the needle hub. 33. The injection needle assembly as defined in claim 32, wherein the first needle cover is made from silicone rubber. 34. The injection needle assembly as defined in claim 32, further comprises a second needle cover, wherein the second needle cover is mounted on the needle hub and forms an axially extending elongated flexible enclosure which sealingly accommodates the distal portion of the needle cannula, wherein the second needle cover has a needle hub end mounted relative to the needle hub and a free end extending beyond the distal end of the needle cannula, the second needle cover being configured to axially collapse and become penetrated by the distal portion of the needle cannula when a proximally directed penetration force is applied on the free end of the second needle cover urging the free end of the second needle cover towards the needle hub. 35. An injection device, comprising: a housing defining a distal drug expelling end and an opposite proximal end, an injection needle assembly as defined in claim 32, the injection needle assembly arranged relative to the housing with the proximal end of the needle cannula pointing towards the proximal end of the housing, a medicament container containing a medicament, the medicament container comprising a cylindrical body extending along a longitudinal axis arranged coaxially with the central axis of the needle cannula, the medicament container further comprising a seal structure adapted to become penetrated by the proximal portion of the needle cannula enabling expelling of medicament from the medicament container, wherein the medicament container is arranged axially movable relative to the injection needle assembly for moving the medicament container from a first state where the first needle cover and the seal structure are not penetrated by the needle cannula and into a second state where the proximal portion of the needle cannula has penetrated the first needle cover and the seal structure, and an expelling assembly comprising a strained spring configured to act on the medicament container, wherein the expelling assembly is activatable to enable the strained spring to move the medicament container from the first state to the second state. 36. The injection device as defined in claim 35, wherein, upon activation of the expelling assembly, the proximal portion of the needle cannula penetrates the seal structure of the medicament container by a relative axial movement with no relative rotation between the needle cannula and the seal structure. 37. The injection device as defined in claim 35, wherein the injection needle assembly comprises a second needle cover mounted on the needle hub and forming an axially extending elongated flexible enclosure which sealingly accommodates the distal portion of the needle cannula, wherein the second needle cover has a needle hub end mounted relative to the needle hub and a free end extending beyond the distal end of the needle cannula, the second needle cover being configured to axially collapse and become penetrated by the distal portion of the needle cannula when a proximally directed penetration force is applied on the free end of the second needle cover urging the free end of the second needle cover towards the needle hub, wherein the injection device comprises a needle shield arranged to shield the second needle cover, and wherein the needle shield and the injection needle assembly is arranged to axially move relative to each other for causing the distal portion of the needle cannula to penetrate the second needle cover and enable the distal portion of the needle cannula to extend beyond a distal end portion of the needle shield. 38. An injection device as defined in claim 35, wherein the seal structure of the medicament container is provided as, or comprises, a non-slit medicament container septum. 38 CROSS-REFERENCE TO RELATED APPLICATIONS This application is a 35 U.S.C. § 371 National Stage application of International Application PCT/EP2017/084696 (published as WO 2018/122295), filed Dec. 28, 2017, which claims priority to European Patent Application 16207643.4, filed Dec. 30, 2016, the contents of all above-named applications are incorporated herein by reference. FIELD OF THE INVENTION The present invention relates to a needle cannula, an injection needle assembly and an injection device for injecting a medicament comprising such an injection needle assembly. BACKGROUND OF THE INVENTION Within development of medical devices it is of primary focus to develop devices that are safe. Yet it is also of particular concern that the medical devices are as simple and user-friendly as possible. Within medical injectors, such as auto-injectors, it is an aim that it involves no or little needle handling. Before use the needle must be kept sterile. In some injection devices, the needle is protected by a needle cover, for example a resilient pierceable needle cover, which maintains a sterile barrier around the needle itself. Such needle cover is often assembled together with the needle to form a needle assembly. WO 2015/197866 A1 and WO 2016/116614 A1 disclose medicament injectors which incorporate a pair of flexible pierceable needle covers that prior to use maintains a front needle and a rear needle in a sterile state. By using a flexible pierceable needle cover as sterility barrier the needle can penetrate the sterility barrier thereby obviating the need for removing the needle cover prior to commencing the actual injection procedure. This eases the needle handling significantly. When using septum equipped cartridges and separate needles which, upon initial use, penetrates the septum to establish fluid communication, there is a risk that coring of the septum will occur. U.S. Pat. No. 2,716,983 discloses various piercing needles where the needle tip is provided with a design to inhibit coring. Further, U.S. Pat. No. 5,716,348 discloses a needle with a design where the cylindrical wall has a laterally facing opening spaced from a rearward opening, and wherein a restriction is arranged between the rearward opening and the laterally facing opening. U.S. Pat. No. 5,709,668 discloses an automatic injector employing a non-coring needle having side port geometry to minimize or eliminate coring of a rubber seal or septum when impaled by the internal needle tip of the cannula. Having regard to the above-identified prior art devices, it is an object of the present invention to provide an improved needle cannula, to provide an injection needle assembly including such needle cannula and an injection device comprising such injection needle assembly, wherein the design of the needle cannula minimizes the risk of coring. BRIEF DESCRIPTION OF THE INVENTION In a first aspect the present invention relates to a needle cannula for an injection device, wherein an elongated tubular needle cannula extends along a central axis and comprises: a skin piercing distal end, a proximal end arranged axially opposite the skin piercing distal end, the proximal end being configured for penetrating a seal structure of a medicament container, a sidewall extending between the skin piercing distal end and the proximal end, and a lumen extending through the needle cannula along said central axis, said lumen has a first opening arranged axially at the skin piercing distal end and a second opening arranged axially at the proximal end, wherein the proximal end being defined by an angled surface pointing generally proximally, the angled surface intersecting with the sidewall at a proximal-most point defining a leading portion and intersecting with the sidewall at an oppositely arranged trailing portion defining a heel portion. The angled surface pointing generally proximally comprises a first surface portion at the heel portion where the first surface portion includes surface areas that form an angle (α1) between 50 to 75 degrees with respect to the central axis. The angled surface pointing generally proximally further comprises a second surface portion at the leading portion where the second surface portion includes surface areas forming an angle (α2) between 60 to 85 degrees with respect to the central axis. In accordance with the first aspect, the needle cannula enables penetration of a cooperating sealing element in a way wherein a slightly angled bevel surface at the leading portion of the needle cannula enables the leading portion of the needle cannula to provide a well-defined contact point for the needle cannula to establish a connection with the cooperating sealing element. At the same time, as the first surface portion at the heel portion includes surface areas that form an angle (α1) that is smaller than the angled bevel surface at the leading portion of the needle cannula, the leading portion extends quite far relative to the heel portion. This enables the leading portion to puncture or start rupturing the sealing element before the heel portion is brought to force its way through the sealing element. Hence, a particular safe penetration is enabled providing no or only little risk of coring of the sealing element during penetration. In some embodiments, the surface areas of the first surface portion forming an angle (α1) between 50 to 75 degrees with respect to the central axis constitute at least 20% of the total area of said angled surface. Also, in some embodiments, the surface areas of the second surface portion forming an angle (α2) between 60 to 85 degrees with respect to the central axis constitute at least 30% of the total area of said angled surface. In further embodiments said angled surface intersects with the sidewall to define a radially inwards facing circumferential edge and a radially outwards facing circumferential edge. The radially inwards facing circumferential edge and/or the radially outwards facing circumferential edge may in some embodiments be formed with blunted non-incising edges along an annular section symmetrically disposed around the heel portion. In some embodiments, the annular section having such blunt edges may have an annular width within 100 to 220 degrees, i.e. within 50 to 110 degrees in either circumferential direction from the heel portion. In other embodiments, the annular section may have an annular width within 140 to 190 degrees. The edges arranged at the leading portion may be formed so that these edges are sharpened. In exemplary embodiments, the surface portion at the leading portion forms an angle (α2) with respect to the central axis which is 5 to 10 degrees greater than the angle (α1) of the surface portion at the heel portion. In further exemplary embodiments, said surface portion at the heel portion forms an angle (α1) between 55 to 70 degrees with respect to the central axis. The needle cannula may in some embodiments be configured for subcutaneous injection, with the size of the needle cannula being a 26 G needle or smaller, preferably a 27 G needle, more preferably a 28 G needle, more preferably a 29 G needle and most preferably a 30 G needle. In still further embodiments, the sidewall of the needle cannula forms an elongated tubular enclosure that does not include opening(s) other than said first opening and said second opening. In a second aspect the present invention relates to a needle cannula for an injection device, wherein an elongated tubular needle cannula extends along a central axis and comprises: a skin piercing distal end, a proximal end arranged opposite the skin piercing distal end, the proximal end being configured for penetrating a seal structure of a medicament container, a sidewall extending between the skin piercing distal end and the proximal end, and a lumen extending through the needle cannula along said central axis, said lumen has a first opening arranged axially at the skin piercing distal end and a second opening arranged axially at the proximal end, wherein the proximal end is defined by an angled surface pointing proximally, wherein at least 60% of the surface portions of the angled surface form an angle (α) between 70 degrees to 85 degrees with respect to the central axis. In accordance with the second aspect, the needle cannula enables penetration of a cooperating sealing element in a way wherein a slightly angled bevel surface of the needle cannula enables a leading portion of the needle cannula to provide a well-defined contact point for the needle cannula to establish a connection with the cooperating sealing element. Hence, a particular safe penetration is enabled providing no or only little risk of coring of the sealing element during penetration. In some embodiments at least 60% of the surface portions of the angled surface form an angle (α) between 76 degrees to 84 degrees with respect to the central axis. In further embodiments the angled surface intersects with the sidewall at a proximal-most point defining a leading portion and intersects with the sidewall at an oppositely arranged trailing portion defining a heal portion, wherein the angled surface of the heal portion forms an angle (α) between 70 degrees to 85 degrees with respect to the central axis, preferably between 76 degrees to 84 degrees with respect to the central axis. In particular embodiments, the heal portion is formed with edges that are blunted. In further embodiments, all circumferential edges at the proximal end of the needle cannula are formed with blunted edges. In further exemplary embodiments, the entire angled surface of the proximal end of the needle cannula forms an angle (α) with respect to the central axis between 70 degrees to 85 degrees, preferably between 76 degrees to 84 degrees with respect to the central axis. The said angled surface may in some embodiments be provided as a single planar angled cut tip. The needle cannula may in some embodiments be configured for subcutaneous injection, with the size of the needle cannula being a 26 G needle or smaller, preferably a 27 G needle, more preferably a 28 G needle, more preferably a 29 G needle and most preferably a 30 G needle. In still further embodiments, the sidewall of the needle cannula forms an elongated tubular enclosure that does not include opening(s) other than said first opening and said second opening. In a third aspect the invention relates to an injection needle assembly incorporating a needle cannula according to any one of the first aspect and the second aspect. The injection needle assembly may be formed to comprise: a needle hub, an elongated tubular needle cannula in accordance with any of the first or the second aspect, and a first needle cover, wherein the needle hub holds the needle cannula between the skin piercing distal end and the proximal end such that a distal portion of the needle cannula extends distally from the needle hub and such that a proximal portion of the needle cannula extends proximally from the needle hub towards the proximal end of the needle cannula, and wherein the first needle cover is mounted on the needle hub and forms an axially extending elongated flexible enclosure which sealingly accommodates the proximal portion of the needle cannula, wherein the first needle cover has a needle hub end mounted relative to the needle hub and a free end extending beyond the proximal end of the needle cannula, the first needle cover being configured to axially collapse and become penetrated by the proximal portion of the needle cannula when a distally directed penetration force is applied on the free end of the first needle cover urging the free end of the first needle cover towards the needle hub. In further embodiments, the injection needle assembly may further comprise a second needle cover, wherein the second needle cover is mounted on the needle hub and forms an axially extending elongated flexible enclosure which sealingly accommodates the distal portion of the needle cannula, wherein the second needle cover has a needle hub end mounted relative to the needle hub and a free end extending beyond the distal end of the needle cannula, the second needle cover being configured to axially collapse and become penetrated by the distal portion of the needle cannula when a proximally directed penetration force is applied on the free end of the second needle cover urging the free end of the second needle cover towards the needle hub. In particular embodiments, the first needle cover and/or the second needle cover may be made from silicone rubber. In a fourth aspect the invention relates to an injection device incorporating a needle assembly according to the third aspect. The injection device may be formed to comprise: a housing defining a distal drug expelling end and an opposite proximal end, an injection needle assembly in accordance with the third aspect, the injection needle assembly arranged relative to the housing with the proximal end of the needle cannula pointing towards the proximal end of the housing, a cartridge containing a medicament, the cartridge comprising a cylindrical body extending along a longitudinal axis arranged coaxially with the central axis of the needle cannula, the cartridge further comprising a seal structure adapted to become penetrated by the proximal portion of the needle cannula enabling expelling of medicament from the cartridge, wherein the cartridge is arranged axially movable relative to the injection needle assembly for moving the cartridge from a first state where the first needle cover and the seal structure are not penetrated by the needle cannula and into a second state where the proximal portion of the needle cannula has penetrated the first needle cover and the seal structure, and an expelling assembly comprising a strained spring configured to act on the cartridge, wherein the expelling assembly is activatable to enable the strained spring to move the cartridge from the first state to the second state. The injection device may be so configured that, upon activation of the expelling assembly, the proximal end of the needle cannula penetrates the seal structure of the medicament container by a relative axial movement with no relative rotation between the needle cannula and the seal structure. In some embodiments, the seal structure of the medicament container and the first needle cover abuts each other during penetration by the proximal end of the needle cannula. In further embodiments the injection needle assembly comprises a second needle cover as described in connection with the third aspect. Such injection device may be formed to comprise a needle shield arranged to shield the second needle cover, and wherein the needle shield and the injection needle assembly is arranged to axially move relative to each other for causing the distal portion of the needle cannula to penetrate the second needle cover and enable the distal portion of the needle cannula to extend beyond a distal end portion of the needle shield. In further embodiments, the seal structure comprises a cartridge septum arranged in a plane substantially orthogonal to the longitudinal axis. In still other embodiments, the seal structure of the cartridge is provided as, or comprises, a non-slit cartridge septum. In certain embodiments the injection device is formed as an auto-injector that is activated or triggered by relative movement between the needle shield and the injection needle assembly. The auto-injector may be configured so that a front part of the needle is inserted manually into an injection site by holding the needle shield against an injection site and applying a manual force for moving the needle forward relative to the needle shield such as to cause the front needle to firstly penetrate the second needle cover covering the front part of the needle and subsequently insert the front needle into the injection site. The needle shield may thus be configured to act on the second needle cover for causing the second needle cover to become penetrated by distal portion of the needle cannula. In particular embodiments, the needle shield engages, as it is being moved axially relative to the needle cover, the second needle cover and thus forces the free end of the second needle cover to move relative to the needle cannula which in turn causes the needle cover to become penetrated by the distal portion of the needle cannula. In certain embodiments, the auto-injector is configured for being triggered upon the distal portion of the needle cannula reaching a pre-defined penetration depth. Such triggering is facilitated by the user manually pushing the auto-injector against the injection site. It should be emphasized that the term “comprises/comprising/comprised of” when used in this specification is taken to specify the presence of stated features, integers, steps or components but does not preclude the presence or addition of one or more other features, integers, steps, components or groups thereof. As used herein, the term “medicament” is meant to encompass any flowable drug capable of being passed through a delivery means such as a hollow needle or cannula in a controlled manner. Examples of flowable drugs are a liquid, a solution, a gel or a fine suspension. Also lyophilized drugs which prior to administration are dissolved into a liquid form are encompassed by the above definition. Representative medicaments includes for example pharmaceuticals, peptides, proteins (e.g. insulin, insulin analogues and C-peptide), hormones, biologically derived or active agents, hormonal and gene based agents, nutritional formulas and other substances in both solid (dispensed) or liquid form. BRIEF DESCRIPTION OF THE DRAWINGS In the following, the invention will be described in greater detail with reference to embodiments shown by the enclosed figures. It should be emphasized that the embodiments shown are used for example purposes only and should not be used to limit the scope of the invention. FIG. 1 shows a sectional side view of one example of an injection device suitable for incorporation of a needle assembly in accordance with the present invention, the needle assembly being in an initial shielded state, FIG. 2 shows a sectional side view of the injection device of FIG. 1 in a state where the distal end of the needle fully protrudes from a needle shield, FIG. 3 shows a sectional side view of the injection device of FIG. 1 in a state where a cartridge has been connected to the proximal end of the needle for fluid delivery and wherein expelling has been initiated, FIG. 4 shows a sectional side view of the injection device of FIG. 1 in a state where the needle shield has returned to its original position to put the needle into a shielded state again, FIG. 5 shows a cross sectional side view of the front portion of a first embodiment of an injection device incorporating a needle assembly according to the invention, FIG. 6 shows a magnified side view of the proximal portion of such a needle cannula according to the invention, FIG. 7 shows a cross sectional side view of the proximal portion of a needle cannula according to the invention with the proximal needle cover of the injection needle assembly visible, FIGS. 8a, 8b and 8c schematically show manufacturing steps for providing a further exemplary needle cannula according to the invention, and FIG. 9 shows further details of the proximal end of a needle cannula provided by the manufacturing steps shown in FIGS. 8a through 8c. DETAILED DESCRIPTION OF THE EMBODIMENTS FIGS. 1 to 4 illustrate operational states for an example injection device which is suitable for use in connection with the present invention. The injection device is shown in four different states of operation in order to explain the basic function of the device. The shown device is generally similar to the device disclosed in WO 2015/197866 A1 in connection with FIGS. 1c, 2c, 3c and 5c of said document. For a detailed description of the disclosed device reference is made to the WO document. It is to be noted that the shown injection device forms a suitable but non-limiting example and that the needle cannula and injection needle assembly of the present invention can be used together with other types of injection devices. All the details of the shown injection device will not be described in detail herein since these details have already been described in the above WO document. FIGS. 1 to 4 show an injection device 1 with a medicament containing cartridge 2, an injection needle provided as a needle cannula 3 having a proximal end 3a and a distal end 3b, a needle hub 4, a needle shield 5, a housing 6 and an expelling assembly 7. The details of the expelling assembly will only be described in general terms in this specification since the needle assembly of the present invention will work with many different types of expelling assemblies. In the shown embodiment, in the shielded state as shown in FIG. 1, the distal end of the needle shield 5 is arranged distally to the distal end 3b of the needle cannula. In this way, the needle is completely shielded by the needle shield. It can also be seen that in the current embodiment, the needle shield 5 is a single element which completely encases the needle assembly. As can be seen from FIG. 1, the needle 3 is arranged as a needle cannula having two pointed ends, one arranged at the proximal end of the needle cannula and one arranged at the distal end. The needle hub 4 grips the middle portion of the needle cannula 3 so that both the distal and the proximal ends of the needle protrude axially relative to the hub 4, i.e. respectively forming a front needle and a rear needle. In the shown embodiment, the hub 4 is mounted fixedly relative to the housing 6. During use, the proximal end of the needle 3a is arranged to engage with a container 2 containing the medicament which is to be injected while the distal end 3b is arranged to pierce the skin of the user to inject the medicament into the body of the user. In the shown embodiment, the container 2 forms a cartridge with a body 2a having a distally arranged outlet covered by a seal structure in the form of a cartridge septum 2b adapted to be pierced by a needle cannula for establishing fluid communication with the cartridge interior. The body of the cartridge accommodates a slidably arranged piston 2c. In the state where a needle has pierced cartridge septum 2b, piston 2c is drivable towards the outlet in order to dispense medicament from the cartridge 2. As can also be seen from FIG. 1, the proximal end 3a of the needle is covered by a proximal needle cover 10a forming a flexible pierceable needle cover and the distal end of the needle 3b is covered by a distal needle cover 10b also forming a flexible pierceable needle cover. The needle covers 10a and 10b will also be referred to as a rear cover and front cover, respectively. Likewise, the part of the needle cannula that extends in a proximal direction from the hub will be referred to as the rear needle, whereas the part of the needle cannula extending in a distal direction from the hub will be referred to as the front needle. It is to be noted that in FIGS. 1 to 4 the shape of the needle covers 10a and 10b are only schematically depicted. The rear and front needle covers 10a and 10b are arranged to allow the needle to be sterilized and then ensure that the needle itself is not contaminated by further handling of the needle assembly. In FIG. 2, the needle shield 5 has been retracted with respect to the needle hub 4 such that the distal end 3b of the needle now extends distally past the distal end 5b of the needle shield. In this way the distal end of the needle 3b is now exposed and ready for fluid communication with a user. As can also be seen in FIG. 2, the act of retracting the needle shield has caused the distal needle cover 10b to be pulled back. This causes the distal end of the needle to pierce through the needle cover thereby uncovering the distal end of the needle. Due to the flexible nature of the needle cover, the needle cover is easily retracted. In the shown embodiment, the act of retracting the needle shield relative to the needle hub triggers the device and thus activates the expelling assembly 7. The expelling assembly primarily consists of a plunger which is biased distally by a force provided by a compression spring. Prior to triggering the compression spring is in a pre-tensed state where the plunger is retained axially by a retaining mechanism. Upon triggering, the plunger is axially released and urged forward by the energy released from the compression spring. The plunger then exerts a distally directed force onto the piston 2c of the cartridge 2. In the shown device, the plunger of the expelling assembly 7 provides a force which initially moves the cartridge 2 relative to the housing 6 and subsequently moves the piston 2c in the cartridge 2 so that the a dose of the medicament in the cartridge 2 is expelled through the needle assembly 50. In FIG. 3, it can be seen that the expelling assembly 7 has pushed the medicament containing cartridge 2 forward in a distal direction to engage with the proximal end of the needle. The proximal end of the needle punctures the septum 2b of the cartridge thereby establishing a fluid path from the cartridge through the needle and to the distal end of the needle whereby the medicament can be injected into the user at the selected injection site. As can also be seen in FIG. 3, the proximal needle cover 10a has also been compressed by the motion of the cartridge towards the needle. This thereby uncovers the proximal end of the needle and allows it to engage with the cartridge. In FIG. 4, the expelling assembly has pushed the piston 2c arranged in the cartridge distally, thereby causing the medicament in the cartridge to be injected through the needle into the injection site. After the medicament has been injected, the needle shield is again pushed forward with respect to the needle hub to shield the distal end of the needle. In the shown embodiment, this occurs as a consequence of the user manually retracting the housing 6 of the injection device relative to the injection site. In the shown embodiment, the needle shield 5 is biased in the distal direction by means of a needle shield spring. The description above with respect to FIGS. 1 to 4 has been provided to give background information of the use of an exemplary injection device. The injection device described is one of many different available injection devices. It should be noted that the needle cannula and the injection needle assembly of the current invention can be used with different types of injection devices wherein a septum equipped cartridge is cooperating with a separate needle assembly, not just the one described above with respect to FIGS. 1 to 4. FIG. 5 shows a cross sectional detailed view of a distal portion of an injection device similar to the injection device shown in FIGS. 1-4, but incorporating an embodiment of a needle assembly 150 in accordance with an aspect of the present invention. The proximal needle cover 110a is formed similar to the needle cover 10 shown in FIGS. 11a-11d of WO 2016/116614 A1. Tests have shown that for conventional double pointed needles, due to particular conditions during triggering of the device, there is a risk that the proximal portion of the needle cannula 130, as it penetrates through the proximal needle cover 10a and the cartridge septum 2b, generates cut-out cores of material of the needle cover 10a and/or the cartridge septum 2b. It has been shown that the risk is most noticeable with high connecting velocities during penetration but also when penetration occurs in low ambient temperatures corresponding to temperatures used during long-time storage of medicament, e.g. refrigerator temperature. Detailed tests have been performed with different samples of an example 30 G needle cannula having a proximal portion with a beveled surface provided as a planar angular cut that forms an angle of 28 degrees with the central axis of the needle cannula. Individual penetration tests have been performed with such example 30 G needle cooperating only with the proximal needle cover 10a, i.e. without cooperating with a cartridge septum, but also with such example 30 G needle cooperating only with the cartridge septum, i.e. without cooperating with the proximal needle cover 10a. All tests have been performed at an ambient temperature of 5 deg. Celsius and with a penetration speed corresponding to the speed obtained by triggering an auto-injector such as shown in FIGS. 1-4. In both situations the example 30 G needle has performed acceptably with no traces of fracturing of the needle cover and the cartridge septum, respectively. However, in a real-life setup corresponding to the configuration shown in FIGS. 1-4 wherein the needle assembly including the example 30 G needle is incorporated in an autoinjector having both a needle cover 10a and a cartridge septum 2b, traces of fracturing elements have been observed. Most likely, the effect of coring in such setup is likely to be associated with the interfacing forces between the needle cover 10a and the septum 2b during needle cannula penetration. Further detailed tests have been performed with an example needle cannula having a proximal portion with an end surface provided as a planar angular cut forming a 90 degree angle with the central axis of the needle cannula, and wherein all edges at the proximal end of the needle cannula subsequently have been made smooth by polishing. Such blunt needle cannula performs acceptably in a setup where the needle cannula only cooperates with a cartridge septum. However, in a real-life setup corresponding to the configuration shown in FIGS. 1-4 wherein the needle assembly including the example blunt needle with a 90 degree cut is incorporated in an autoinjector having both a needle cover 10a and a cartridge septum 2b, traces of fracturing elements have been observed. In accordance with the present invention a plurality of different needle cannulas of needle gauge 27 G and 30 G have been prepared wherein each of the needle cannulas have been formed by cutting the proximal end at an angle (α) with respect to the central axis of the needle cannula. A large number of sample needle cannulas have been prepared by cutting the proximal portion of the needle cannula to provide a bevel surface having an angle (α) relative to the central axis of the needle cannula of 76 degrees, 80 degrees and 85 degrees, respectively. Subsequently to cutting, the edge surfaces of the planar angled cut tip have been exerted to a finishing process in the form of glass blasting exerted at an angle of 90 degrees relative to the planar end surface of the needle cannula. All tests have been performed at an ambient temperature of 5 deg. Celsius as well as room temperature (20 deg. Celsius) and with a penetration speed corresponding to the speed obtained by triggering an auto-injector such as shown in FIGS. 1-4. No traces of fracturing of the needle cover and the cartridge septum has been observed. FIG. 6 shows a magnified side view of the proximal portion of such a needle cannula 130 with an 80 degree cut bevel end. The single cut angled end surface 134 exhibits a leading portion 134.1 defining the most proximal point of the needle cannula. Surface 134 further exhibits a trailing portion 134.2, i.e. a heel portion. FIG. 7 shows a cross sectional side view of the proximal portion of a needle cannula 130 with an 80 degree cut bevel and with a portion of the proximal needle cover 10a also visible. The needle cannula according to the invention has proven that the slightly angled bevel surface of needle cannula enables the leading portion 134.1 to provide a well-defined contact point for the needle cannula to establish a connection with the needle cover and the cartridge septum, respectively. The slightly angled grinding surface is proven to be a suitable balance between ensuring the needle indicates where the cover must break by establishing a well-defined tip and not introducing sharp, knife-like structures at locations in the vicinity of the heel portion of the proximal end of the needle cannula. The objective of forming a non-coring needle cannula the grinding is considered effective with a proximal end of the needle cannula exhibiting an angle relative to the central axis within a range from 70-85 degrees. It is to be noted that, in accordance with the invention, the slightly angled surface may be provided as only covering a fraction of the proximal end surface circumscribing the proximal opening. However, it is contemplated that a main part of the proximal surface, in particular the surface area covering the heel portion is formed with an angle of inclination with respect to the central axis of the needle cannula within said angle interval of 70 to 85 degrees. The optional subsequent finishing (glass blasting, polishing, etc) is to ensure that no sharp edges, structures or flanges, especially at the “heel” of the needle tube, will potentially cause coring of cooperating cover or seal elements. FIGS. 8a to 8c schematically show manufacturing steps for providing a further exemplary needle cannula according to the invention. The proximal portion of such a needle cannula 130 is manufactured in a simple 3-step process. In the shown embodiment, as shown in FIG. 8a, an oblique cut is firstly made to the proximal end 133a of the needle cannula by grinding the needle cannula with a 70 degree grinding angle relative to the central axis. This causes a proximally facing angled surface 134a to be formed which defines a leading portion 134.1 and a heel portion 134.2. In a second procedural step, shown in FIG. 8b, the proximal end of the needle cannula is exerted to a sand blasting operation, or similar blunting operation, so as to cause the initially formed sharp edges to become blunted. Hence, at areas 134b where the angled surface 134a intersects with the sidewall of the cannula 130, the areas 134b form blunted circumferentially running edges, both at edges facing radially inwards and at edges facing radially outwards. In a third procedural step, shown in FIG. 8c a second grinding step is performed by exclusively grinding areas arranged at the leading portion 134.1 of the needle cannula with an 80 degree grinding angle relative to the central axis thereby forming a planar surface portion 134c symmetrically disposed around the leading portion 134.1. Hereby edges disposed only at the leading portion 134.1 are sharpened. In the shown embodiment, and further explained with reference to FIG. 9, the first grinding operation may be provided by a grinding angle α1 with respect to the central axis, whereas the second grinding operation may be provided by a grinding angle α2 with respect to the central axis. In further exemplary embodiments, the grinding angle of the first grinding operation may be performed so as to provide a surface portion 134a at the heel portion 134.2 forming an angle α1 within the interval 50 to 75 degrees with respect to the central axis. Typically, in such embodiments, the grinding angle of the second grinding operation may be performed so as to provide a surface portion 134c at the leading portion 134.1 forming an angle α2 with respect to the central axis which is 5 to 10 degrees greater than the angle α1 of surface portion 134a. By using the simple 3-step operation described above, an inexpensive and effective non-coring solution is provided where the proximal end 133a of the needle cannula 130 exhibits a proximally facing end surface having a sharp leading portion as well as a blunt heel portion. It is to be noted that, since the distal portion 133b of the needle cannula that carries the skin penetrating tip is very fragile, it is important that this end will be protected during manufacture/assembly, and not used to position the needle. Hence, it is preferable if the proximal portion of the needle 133a can serve this purpose. Hence, the design of the proximal portion of the needle is provided with a robust design allowing positioning of the needle by engaging the proximal portion with a supporting surface until the needle cannula is fixedly attached relative to a needle hub element. Furthermore, some preferred embodiments have been shown in the foregoing, but it should be stressed that the invention is not limited to these, but may be embodied in other ways within the subject matter defined in the following claims and within the remaining disclosure. 16473948 novo nordisk a/s USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open Apr 5th, 2022 04:33PM Apr 5th, 2022 04:33PM Novo Nordisk Health Care Pharmaceuticals & Biotechnology
nyse:nvo Novo Nordisk Sep 2nd, 2008 12:00AM Jul 16th, 2002 12:00AM https://www.uspto.gov?id=US07419949-20080902 Single-dose administration of factor VIIa The present invention provides methods for preventing and/or treating bleeding episodes by administering a single dose of Factor VIIa or a Factor VIIa equivalent. Preferably, the single dose comprises between about 150 and about 500 μg/kg Factor VIIa or Factor VIIa equivalent. 7419949 1. A method for treating a bleeding episode in a human subject in need of such treatment, said method comprising administering intravenously to said human subject purified Factor VIIa, wherein said administering is in a single dose and said dose comprises a single-dose-effective amount of said Factor VIIa, wherein said single-dose-effective amount comprises between 250 and 500 μg/kg Factor VIIa and wherein, subsequent to said administration, no further Factor VIIa is administered to said subject for a period of at least 4 hours. 2. A method as defined in claim 1, wherein said period is at least 24 hours. 3. A method as defined in claim 1, wherein said single dose is administered over a period of less than 5 minutes. 4. A method as defined in claim 1, wherein said single-dose-effective amount comprises between 300 and 500 μg/kg Factor VIIa. 5. A method as defined in claim 1, further comprising administering, in said single dose, or substantially simultaneously with said single dose, a second coagulant agent. 6. A method as defined in claim 5, wherein said second coagulant agent is selected from the group consisting of Factor VIII, Factor IX, and Factor XIII. 7. A method as defined in claim 1, further comprising administering an anticoagulant, wherein said anticoagulant is administered in said single dose, or substantially simultaneously with said single dose. 8. A method for treating a bleeding episode, said method comprising administering intravenously to a human subject in need of such treatment (i) a first amount of purified Factor VIIa and (ii) a second amount of second coagulant agent, wherein said first amount consists of between 250 and 500 μg/kg Factor VIIa and wherein said first and second amounts together comprise an aggregate effective amount for treating said bleeding and said aggregate effective amount is administered in a single dose. 9. A method for treating a bleeding episode, said method comprising administering intravenously to a human subject in need of such treatment an effective amount for treating said bleeding of purified human Factor VIIa, wherein: (i) said effective amount is administered in a single dose over a period of less than 5 minutes; (ii) said effective amount comprises between 300 and 500 μg/kg human Factor VIIa; and (iii) subsequent to said administration, no further Factor VIIa is administered to said subject for a period of at least 1 hour. 9 CROSS REFERENCE TO RELATED APPLICATIONS This application claims priority under 35 U.S.C. 119 of U.S. application Ser. No. 60/305,720 filed on Jul. 16, 2001, the contents of which are fully incorporated herein by reference. FIELD OF THE INVENTION The present invention relates to methods for preventing and/or treating bleeding using coagulation factors. BACKGROUND OF THE INVENTION Factor VII is a plasma coagulation factor, which, once activated to Factor VIIa, initiates the normal haemostatic process by forming a complex with tissue factor (TF), a cell surface glycoprotein that is exposed to the circulation as a result of injury to the vessel wall. Subsequently, the Factor VIIa-TF complex activates Factor IX and Factor X into their activated forms (Factor IXa and Factor Xa, respectively). Factor Xa converts limited amounts of prothrombin to thrombin on the tissue factor-bearing cell. Thrombin activates platelets and Factors V and VIII into Factors Va and VIIIa, both cofactors in the further process leading to the full thrombin burst. Thrombin finally converts fibrinogen to fibrin resulting in formation of a fibrin clot. Fibrin clots formed in the presence of high thrombin concentrations comprise a tighter network and are more resistant to proteolysis than clots formed in lower concentrations of thrombin. Accordingly, a full thrombin burst is likely to be important for forming a hemostatic plug that is resistant to fibrinolysis and thus to facilitate full hemostasis and wound healing. Factor VIIa, as well as Factor VIII and Factor IX, have been used to control bleeding disorders that are caused by clotting factor deficiencies (such as, e.g. haemophilia A and B or deficiency of coagulation Factors XI or VII) or clotting factor inhibitors. Factor VIIa has also been used to control excessive bleeding caused by defective platelet function, thrombocytopenia or von Willebrand's disease. Typically, however, patients are treated with multiple injections or infusions of a coagulation factor before the bleeding is stopped. In the case of Factor VIII and Factor IX administration, a considerable number of injections are needed to maintain haemostasis until the injury causing the bleeding is completely healed. A quicker and more effective treatment, as well as a reduction in the number of injections needed before the bleeding is stopped, represent important benefit to such patients. It would also be a considerable benefit to a patient needing frequent injections or infusions with a haemostatic agent that the injection frequency be reduced. Thus, there is a need in the art for methods for preventing and/or treating bleeding episodes that reduce the duration of administration and provide a more rapid hemostasis. SUMMARY OF THE INVENTION The present invention relates to methods for preventing and/or treating a bleeding episode in a subject in need of such treatment, which are carried out by administering to the subject, in a single dose, a single-dose-effective amount of Factor VIIa or a Factor VIIa equivalent. Preferably, subsequent to the administration, no further Factor VIIa or protein having Factor VIIa coagulant activity is administered to the subject for an interval of at least about 1 hour. In some embodiments, the interval is at least about 4 hours; in other embodiments, the interval is at least about 24 hours; and in some embodiments, no further Factor VIIa or protein having Factor VIIa coagulant activity is administered during the particular bleeding episode that is being treated. In some embodiments, the single-dose-effective amount comprises between about 150 and about 500 μg/kg Factor VIIa or a corresponding amount of a Factor VIIa equivalent; in other embodiments, the single-dose-effective amount comprises between about 200 and about 500 μg/kg; between about 250 and about 500 μg/kg; between about 300 and about 500 μg/kg; between about 350 and 500 μg/kg; between about 400 and about 500 μg/kg; between about 450 and about 500 μg/kg; and greater than 500 μg/kg, respectively, of Factor VIIa or a corresponding amount of a Factor VIIa equivalent. In some embodiments, the Factor VIIa equivalent exhibits at least about 30% of the coagulant activity of Factor VIIa on a molar basis. Non-limiting examples of a Factor VIIa equivalent include S52A-FVII, S60A-FVII; L305V-FVII, L305V/M306D/D309S-FVII, L3051-FVII, L305T-FVII, F374P-FVII, V158T/M298Q-FVII, V158D/E296V/M298Q-FVII K337A-FVII, M298Q-FVII, V158D/M298Q-FVII, L305V/K337A-FVII, V 158D/E296V/M298Q/L305V-FVII, V158D/E296V/M298Q/K337A-FVII, V158D/E296V/M298Q/L305V/K337A-FVII, K157A-FVII, E296V-FVII, E296V/M298Q-FVII, V158D/E296V-FVII, V158D/M298K-FVII, and S336G-FVII; Factor VIIa variants exhibiting increased proteolytic stability as disclosed in U.S. Pat. No. 5,580,560; Factor VIIa that has been proteolytically cleaved between residues 290 and 291 or between residues 315 and 316; oxidized forms of Factor VIIa; Factor VII-sequence variants wherein the amino acid residue in positions 290 and/or 291 (of SEQ ID NO:1), preferably 290, have been replaced, and Factor VII-sequence variants wherein the amino acid residue in positions 315 and/or 316 (of SEQ ID NO:1), preferably 315, have been replaced. In some embodiments, the method further comprises administering, with or substantially simultaneously with the single dose, a second coagulant agent. Non-limiting examples of a second coagulant agent include Factor VIII, Factor IX, and Factor XIII. In some embodiments, the invention provides a method for treating a bleeding episode, which is carried out by administering to a subject in need of such treatment (i) a first amount of Factor VIIa or a Factor VIIa equivalent and (ii) a second amount of second coagulant agent, wherein the first and second amounts together comprise an aggregate effective amount for treating the bleeding episode and the aggregate effective amount is administered in a single dose. In some embodiments, the Factor VIIa used in practicing the invention is recombinant human Factor VIIa. In one embodiment, the invention provides a method for treating a bleeding episode, which is carried out by administering to a human subject in need of such treatment an effective amount for treating said bleeding of human Factor VIIa or a human Factor VIIa equivalent, wherein: (i) said effective amount is administered in a single dose over a period of less than about 5 minutes; (ii) said effective amount comprises between about 300 and about 500 ug/kg human Factor VIIa or human Factor VIIa equivalent or a corresponding amount of a Factor VIIa equivalent; and (iii) subsequent to said administration, no further Factor VIIa or Factor VIIa equivalent is administered to said subject for a period of at least about 1 hour. In practicing the present invention, administration may be achieved by any mode of administration, including, without limitation, intravenous, intramuscular, subcutaneous, mucosal, and pulmonary routes of administration. In another aspect, the invention provides a method for preventing a bleeding episode, which is carried out by administering to a human subject in need of such prevention an effective amount for preventing the bleeding episode of human Factor VIIa or a human Factor VIIa equivalent, wherein: (i) the effective amount is administered in a single dose over a period of less than about 5 minutes; (ii) the effective amount comprises between about 250 and about 500 μg/kg human Factor VIIa or human Factor VIIa equivalent or a corresponding amount of a Factor VIIa equivalent; and (iii) subsequent to the administration, no further Factor VIIa or Factor VIIa equivalent is administered to said subject for a period of at least about 1 hour. In some embodiments, the subject suffers from hemophilia A or B. In some embodiments, the bleeding is joint bleeding. In some embodiments, the subject has not been treated therapeutically with an anticoagulant for at least about 48 hours prior to administration of Factor VIIa or a Factor VIIa equivalent. In some embodiments, the subject has not been treated therapeutically with a Vitamin K antagonist for at least about 48 hours prior to said administering. DETAILED DESCRIPTION OF THE INVENTION The present invention relates to methods for preventing and/or treating bleeding in animals, particularly in humans. The invention is based on the discovery that administration of a single dose comprising a predetermined amount of Factor VIIa is effective in treating bleeding episodes, including major bleeding episodes, and can also be used to prevent an anticipated bleeding episode. Without wishing to be bound by theory, it is believed that Factor VIIa enhances thrombin generation on tissue factor-bearing cells and on activated platelets at the site of injury and that administration of a single dose of Factor VIIa according to the present invention provides a full thrombin burst, thereby facilitating the formation of a tight, strong fibrin network that is relatively resistant to premature fibrinolysis and obviating the need for further administration of Factor VIIa. As used herein, prevention refers to prophylactic administration of Factor VIIa so as to minimize or inhibit an anticipated bleeding episode, such as, e.g., prior to surgery. Treatment refers to regulation of an already-occurring bleeding, such as, for example, in trauma, with the purpose of inhibiting or minimizing the bleeding. It will be understood that efficacy in prevention and/or treatment according to the present invention encompasses the absence of significant side-effects, including, without limitation, disseminated intravascular coagulation (DIC), that would counterindicate to those of ordinary skill in the art the use of any particular therapeutic regimen. Bleeding refers to extravasation of blood from any component of the circulatory system. The term “bleeding episode” includes, without limitation, bleeding (including, without limitation, excessive, uncontrolled bleeding, i.e., haemorrhaging) in connection with surgery or trauma, such as, for example, in connection with acute haemarthroses (bleedings in joints), chronic haemophilic arthropathy, haematomas, (e.g., muscular, retroperitoneal, sublingual and retropharyngeal), bleedings in other tissue, haematuria (bleeding from the renal tract), cerebral haemorrhage, surgery (e.g., hepatectomy), dental extraction, and gastrointestinal bleedings (e.g., UGI bleeds). Also included are hemorrhages in organs such as the brain, inner ear region and eyes with limited possibility for surgical haemostasis; as well as hemorrhages in connection with biopsies of various organs (liver, lung, tumour tissue, gastrointestinal tract) as well as laparoscopic surgery. Common to these situations is the difficulty in providing haemostasis using surgical techniques (such as, e.g., sutures or clips), which is also the case when bleeding is diffuse. In practicing the invention, Factor VIIa or a Factor VIIa equivalent is administered to a patient as a single dose comprising a single-dose-effective amount. Administration of a single dose refers to administration of an entire dose of Factor VIIa as a bolus over a period of less than about 5 minutes. In some embodiments, the administration occurs over a period of less than about 2.5 minutes, and, in some, over less than about 1 min. A single-dose-effective amount of Factor VIIa or a Factor VIIa equivalent refers to the amount of Factor VIIa or equivalent which, when administered in a single dose according to the invention, produces a measurable improvement in at least one clinical parameter of haemostasis known to those of ordinary skill in the art (see below). Typically, a single-dose effective amount comprises at least 150 μg/kg Factor VIIa. In different embodiments, a single-dose-effective amount of Factor VIIa comprises between about 150-500 μg/kg; 250-500 μg/kg; 300-500 μg/kg 350-500 μg/kg; 400-500 μg/kg; 450-500 μg/kg; or more than 500 μg/kg, respectively. When Factor VIIa equivalents are administered according to the present invention, a single-dose effective amount corresponding to the above-cited amounts may be determined by comparing the coagulant activity of the Factor VIIa equivalent with that of Factor VIIa (see below) and adjusting the amount to be administered proportionately. It will be understood that a single-dose-effective amount of Factor VIIa may vary according to the subject's haemostatic status, which, in turn, may be reflected in one or more clinical parameters, including, e.g., relative levels of circulating coagulation factors; amount of blood lost; rate of bleeding; hematocrit, and the like. It will be further understood that the single-dose-effective amount may be determined by those of ordinary skill in the art by routine experimentation, by constructing a matrix of values and testing different points in the matrix. In some embodiments, following administration of a single-dose of Factor VIIa or a Factor VIIa equivalent according to the invention, the patient receives no further Factor VIIa or Factor VIIa equivalent for an interval of at least about 1 hour. In some embodiments the post-administration interval is at least about 4 hours; in other embodiments, the post-administration interval is at least about 24 hours. In still other embodiments, no further Factor VIIa or Factor VIIa equivalent is administered to treat the particular bleeding episode. According to the invention, Factor VIIa or a Factor VIIa equivalent may be administered by any effective route, including, without limitation, intravenous, intramuscular, subcutaneous, mucosal, and pulmonary routes of administration. Preferably, administration is by an intravenous route. Factor VIIa and Factor VIIa equivalents: In practicing the present invention, any Factor VIIa or equivalent may be used that is effective in preventing or treating bleeding when administered in a single dose. In some embodiments, the Factor VIIa is human Factor VIIa, as disclosed, e.g., in U.S. Pat. No. 4,784,950 (wild-type Factor VII). The term “Factor VII” is intended to encompass Factor VII polypeptides in their uncleaved (zymogen) form, as well as those that have been proteolytically processed to yield their respective bioactive forms, which may be designated Factor VIIa. Typically, Factor VII is cleaved between residues 152 and 153 to yield Factor VIIa. Factor VIIa equivalents include, without limitation, Factor VII polypeptides that have either been chemically modified relative to human Factor VIIa and/or contain one or more amino acid sequence alterations relative to human Factor VIIa. Such equivalents may exhibit different properties relative to human Factor VIIa, including stability, phospholipid binding, altered specific activity, and the like. In one series of embodiments, a Factor VIIa equivalent includes polypeptides that exhibit at least about 10%, preferably at least about 30%, more preferably at least about 50%, and most preferably at least about 70%, of the specific biological activity of human Factor VIIa. For purposes of the invention, Factor VIIa biological activity may be quantified by measuring the ability of a preparation to promote blood clotting using Factor VII-deficient plasma and thromboplastin, as described, e.g., in U.S. Pat. No. 5,997,864. In this assay, biological activity is expressed as the reduction in clotting time relative to a control sample and is converted to “Factor VII units” by comparison with a pooled human serum standard containing 1 unit/ml Factor VII activity. Alternatively, Factor VIIa biological activity may be quantified by (i) measuring the ability of Factor VIIa or a Factor VIIa equivalent to produce of Factor Xa in a system comprising TF embedded in a lipid membrane and Factor X. (Persson et al., J. Biol. Chem. 272:19919–19924, 1997); (ii) measuring Factor X hydrolysis in an aqueous system (see, Example 5 below); (iii) measuring the physical binding of Factor VIIa or a Factor VIIa equivalent to TF using an instrument based on surface plasmon resonance (Persson, FEBS Letts. 413:359–363, 1997) and (iv) measuring hydrolysis of a synthetic substrate by Factor VIIa and/or a Factor VIIa equivalent. Non-limiting examples of Factor VII-related polypeptides having substantially the same or improved biological activity as wild-type Factor VII include S52A-FVII, S60A-FVII (lino et al., Arch. Biochem. Biophys. 352: 182–192, 1998); L305V-FVII, L305V/M306D/D309S-FVII, L3051-FVII, L305T-FVII, F374P-FVII, V158T/M298-Q-FVII, V158D/E296V/M298Q-FVII, K337A-FVII, M298Q-FVII, V158D/M298Q-FVII, L305V/K337A-FVII, V158D/E296V/M298Q/L305V-FVII, V158D/E296V/M298Q/K337A-FVII, V158D/E296V/M298Q/L305V/K337A-FVII, K157A-FVII, E296V-FVII, E296V/M298Q-FVII, V158D/E296V-FVII, V158D/M298K-FVII, and S336G-FVII; Factor VIIa variants exhibiting increased proteolytic stability as disclosed in U.S. Pat. No. 5,580,560; Factor VIIa that has been proteolytically cleaved between residues 290 and 291 or between residues 315 and 316 (Mollerup et al., Biotechnol. Bioeng. 48:501–505, 1995); oxidized forms of Factor VIIa (Komfelt et al., Arch. Biochem. Biophys. 363:43–54, 1999), Factor VII-sequence variants wherein the amino acid residue in positions 290 and/or 291 (of SEQ ID NO:1), preferably 290, have been replaced, and Factor VII-sequence variants wherein the amino acid residue in positions 315 and/or 316 (of SEQ ID NO:1), preferably 315, have been replaced. Preparations and formulations: The present invention encompasses therapeutic administration of Factor VIIa or Factor VIIa equivalents, which is achieved using formulations that comprise Factor VIIa preparations. As used herein, a “Factor VII preparation” refers to a plurality of Factor VIIa polypeptides or Factor VIIa equivalent polypeptides, including variants and chemically modified forms, that have been separated from the cell in which they were synthesized, whether a cell of origin or a recombinant cell that has been programmed to synthesize Factor VIIa or a Factor VIIa equivalent. Separation of polypeptides from their cell of origin may be achieved by any method known in the art, including, without limitation, removal of cell culture medium containing the desired product from an adherent cell culture; centrifugation or filtration to remove non-adherent cells; and the like. Optionally, Factor VII polypeptides may be further purified. Purification may be achieved using any method known in the art, including, without limitation, affinity chromatography, such as, e.g., on an anti-Factor VII antibody column (see, e.g., Wakabayashi et al., J. Biol. Chem. 261:11097, 1986; and Thim et al., Biochem. 27:7785, 1988); hydrophobic interaction chromatography; ion-exchange chromatography; size exclusion chromatography; electrophoretic procedures (e.g., preparative isoelectric focusing (IEF), differential solubility (e.g., ammonium sulfate precipitation), or extraction and the like. See, generally, Scopes, Protein Purification, Springer-Verlag, New York, 1982; and Protein Purification, J.-C. Janson and Lars Ryden, editors, VCH Publishers, New York, 1989. Following purification, the preparation preferably contains less than about 10% by weight, more preferably less than about 5% and most preferably less than about 1%, of non-Factor VII proteins derived from the host cell. Factor VII and Factor VII-related polypeptides may be activated by proteolytic cleavage, using Factor XIIa or other proteases having trypsin-like specificity, such as, e.g., Factor IXa, kallikrein, Factor Xa, and thrombin. See, e.g., Osterud et al., Biochem. 11:2853 (1972); Thomas, U.S. Pat. No. 4,456,591; and Hedner et al., J. Clin. Invest. 71:1836 (1983). Alternatively, Factor VII may be activated by passing it through an ion-exchange chromatography column, such as Mono Q® (Pharmacia) or the like. The resulting activated Factor VII may then be formulated and administered as described below. Pharmaceutical compositions or formulations for use in the present invention comprise a Factor VIIa preparation in combination with, preferably dissolved in, a pharmaceutically acceptable carrier, preferably an aqueous carrier or diluent. A variety of aqueous carriers may be used, such as water, buffered water, 0.4% saline, 0.3% glycine and the like. The preparations of the invention can also be formulated into liposome preparations for delivery or targeting to the sites of injury. Liposome preparations are generally described in, e.g., U.S. Pat. Nos. 4,837,028, 4,501,728, and 4,975,282. The compositions may be sterilised by conventional, well-known sterilisation techniques. The resulting aqueous solutions may be packaged for use or filtered under aseptic conditions and lyophilised, the lyophilised preparation being combined with a sterile aqueous solution prior to administration. The compositions may contain pharmaceutically acceptable auxiliary substances or adjuvants, including, without limitation, pH adjusting and buffering agents and/or tonicity adjusting agents, such as, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, etc. Combinations: The present invention encompasses combined single-dose administration of an additional agent in concert with Factor VIIa or a Factor VIIa equivalent. In some embodiments, the additional agent comprises a coagulant, including, without limitation, a coagulation factor such as, e.g., Factor VIII, Factor IX, or Factor XIII; or an inhibitor of the fibrinolytic system, such as, e.g., aprotinin, ε-aminocaproic acid or tranexamic acid. In other embodiments, the additional agent comprises an anticoagulant, including, without limitation, heparin, warfarin, coumarin, and modified Factor VII polypeptides, such as, e.g., R152E-Factor VIIa (Wildgoose et al., Biochem 29:3413–3420, 1990), S344A-Factor VIIa (Kazama et al., J. Biol. Chem. 270:66–72, 1995), FFR-Factor VIIa (Hoist et al., Eur. J. Vasc. Endovasc. Surg. 15:515–520, 1998), Factor VIIa lacking the Gla domain, (Nicolaisen et al., FEBS Letts. 317:245–249, 1993), and chemically modified Factor VII polypeptides (U.S. Pat. No. 5,997,864). It will be understood that, in embodiments comprising single-dose administration of combinations of Factor VIIa with other agents, the dosage of Factor VIIa or Factor VIIa equivalent may on its own comprise a single-dose-effective amount. Alternatively, the combination of Factor VIIa or equivalent and the second agent may together comprise a single-dose-effect amount for preventing or treating bleeding episodes. Indications: The present invention encompasses single-dose administration of Factor VIIa or a Factor VIIa equivalent to any patient who either anticipates a bleeding episode or who is actively experiencing a bleeding episode. Such patients include, without limitation, those suffering from bleeding disorders that are caused by clotting factor deficiencies (e.g. haemophilia A and B, or deficiency of coagulation Factors XI or VII); clotting factor inhibitors; defective platelet function; thrombocytopenia; or von Willebrand's disease. The methods of the invention may also be applied to patients who are about to undergo surgery, preferably major surgery, whether or not they suffer from a bleeding disorder; as well as trauma patients. Furthermore, any type of profuse bleeding from the gastrointestinal tract, or any bleeding occurring postoperatively (including that occurring in patients not suffering from a bleeding disorder) may benefit from treatment according to the present invention. In some embodiments, the invention does not encompass administration of Factor VIIa or equivalent to patients undergoing minor surgery. In other embodiments, the invention does not encompass administration of Factor VIIa or equivalent to patients not suffering from a clotting disorder who had been administered an anticoagulant (such as, e.g., acetocoumerol) within 48 hours prior to Factor VIIa administration. In other embodiments, the invention does not encompass administration of Factor VIIa or equivalent to patients not suffering from a clotting disorder who had been administered a Vitamin K antagonist within 48 hours prior to Factor VIIa administration. The present invention also provides the benefit of allowing a patient to selfadminister an effective dose of Factor VIIa or a Factor VIIa equivalent in order to facilitate effective management of anticipated or current bleeding episodes. Many variations of the present invention will suggest themselves to those skilled in the art in light of the above detailed description. Such obvious variations are within the full intended scope of the invention. All patents, patent applications, and literature references referred to herein are hereby incorporated by reference in their entirety. The following examples are intended as non-limiting illustrations of the present invention. EXAMPLE 1 High-dose Factor VIIa Administration Patients suffering from hemophilia (including, e.g., patients with clotting factor inhibitors, acquired inhibitor patients, patients suffering from Factor VII deficiency, and patients suffering from von Willebrands disease) are enrolled in a registry that tracks the outcome of bleeding episodes whose treatment includes bolus administration of Factor VIIa. The bleeding episodes are characterized as spontaneous, related to traumatic injury, or other (including, e.g., surgical or dental procedures). The dosage groups are characterized as ≦100 μg/kg; 100-150 μg/kg; 150-200 μg/kg and ≧200 μg/kg. Responses to treatment (as assessed at 72 h) are characterized as cessation of bleeding, slowing of bleeding; or no response. EXAMPLE 2 Single High-dose Factor VIIa Administration Description of Clinical Trial: A randomized, multicenter, cross-over, double-blind study is performed to evaluate the efficacy and safety of Factor VIIa (by two different blinded dose schedules) in producing hemostasis in joint bleeds in a home-treatment setting. Subjects with congenital hemophilia A or B and inhibitors to Factor VIII or Factor IX receive treatment and are assessed for at least 9 hours after the dosing. The success or failure of the treatment is ascertained using a pilot algorithm to assess changes in pain and joint mobility. rFVIIa will be given as an intravenous bolus injection, either at 270 μg/kg body weight dose at hour 0, or at 90 μg/kg body weight doses given at hours 0, 3 and 6 and placebo solutions will be administered to blind subject as to the dose regimen of rFVIIa being administered. If additional doses of Factor VIIa are administered within the first 9 hours to achieve hemostasis, then the treatment efficacy is graded as a failure. Trial Population: Twenty-four patients with congenital hemophilia A or B and inhibitors to factor VIII or IX have been enrolled in this trial. Patients must have experienced two or more mild or moderate joint bleeds during the past 12 months. Assessments Treatment efficacy is based on the evaluation of pain, joint mobility and measure of circumference of the elbow or knee at the midpoint of the joint in extension. These variables are graded and entered in the diary by the patient/caregiver. Pain and mobility are assessed as more, no difference or less than before the treatment and circumference is measured in millimeters. An independent blinded committee reviews the diary data on pain and joint mobility and judges the response to treatment as success or failure based on a pilot algorithm developed for this study. 10196902 novo noridsk healthcare a/g USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open 514/2 Mar 31st, 2022 02:17PM Mar 31st, 2022 02:17PM Novo Nordisk Health Care Pharmaceuticals & Biotechnology
nyse:nvo Novo Nordisk Aug 15th, 2006 12:00AM Sep 4th, 2003 12:00AM https://www.uspto.gov?id=US07091245-20060815 Compounds, their preparation and use Novel compounds of the general formula (I), the use of these compounds as pharmaceutical compositions, pharmaceutical compositions comprising the compounds and methods of treatment employing these compounds and compositions. The present compounds may be useful in the treatment and/or prevention of conditions mediated by Peroxisome Proliferator-Activated Receptors (PPAR), in particular the PPARδ suptype. 7091245 1. A compound of formula (I): wherein X1 is aryl or heteroaryl each of which is optionally substituted with one or more substituents selected from halogen, hydroxy, cyano, amino or carboxy; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-6-alkylthio, arylthio, C3-6-cycloalkylthio, C1-6-alkylcarbonyl, arylcarbonyl, C1-6-alkylsulfonyl, arylsulfonyl, C1-6-alkylamido, arylamido, C1-6-alkylaminocarbonyl, C1-6-dialkylaminocarbonyl, C1-6-alkylamino, C1-6-dialkylamino or C3-6-cycloalkylamino each of which is optionally substituted with one or more halogens; and X2 is arylene or heteroarylene each of which is optionally substituted with one or more substituents selected from halogen, hydroxy, cyano, amino or carboxy; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-6-alkylthio, C3-6-cycloalkylthio, C1-6-alkylamino, C1-6-dialkylamino or C3-6-cycloalkylamino each of which is optionally substituted with one or more halogens; and X3 is aryl or heteroaryl each of which is optionally substituted with one or more substituents selected from halogen, hydroxy, cyano, amino or carboxy; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-6-alkylthio, arylthio, C3-6-cycloalkylthio, C1-6-alkylcarbonyl, arylcarbonyl, C1-6-alkylsulfonyl, arylsulfonyl, C1-6-alkylamido, arylamido, C1-6-alkylaminocarbonyl, C1-6-dialkylaminocarbonyl, C1-6-alkylamino, C1-6-dialkylamino or C3-6-cycloalkylamino each of which is optionally substituted with one or more halogens; and X4 is arylene or heteroarylene each of which is optionally substituted with one or more substituents selected from halogen, hydroxy, cyano, amino or carboxy; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-6-alkylthio, C3-6-cycloalkylthio, C1-6-alkylamino, C1-6-dialkylamino or C3-6-cycloalkylamino each of which is optionally substituted with one or more halogens; and Ar is arylene which is optionally substituted with one or more substituents selected from halogen, hydroxy or cyano; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-6-alkylthio, arylthio or C3-6-cycloalkylthio each of which is optionally substituted with one or more halogens; and Y1 is O or S; and Y2 is O or S; and Z is —(CH2)n— wherein n is 1, 2 or 3; and R1 is hydrogen, halogen or a substituent selected from C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aralkyl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-6-alkylthio, arylthio or C3-6-cycloalkylthio each of which is optionally substituted with one or more halogens; and R2 is hydrogen, C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, C4-6-alkenynyl or aryl; or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable solvate thereof, or any tautomeric forms, stereoisomers, mixture of stereoisomers, racemic mixture, or polymorphs thereof. 2. A compound according to claim 1, wherein X1 is aryl optionally substituted with one or more substituents selected from halogen, hydroxy; or C1-6-alkyl, aryl, heteroaryl, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkylcarbonyl, C1-6-alkylsulfonyl, arylamido, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 3. A compound according to claim 2, wherein X1 is aryl optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 4. A compound according to claim 2, wherein X1 is phenyl optionally substituted with one or more substituents selected from halogen, hydroxy; or C1-6-alkyl, aryl, heteroaryl, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkylcarbonyl, C1-6-alkylsulfonyl, arylamido, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 5. A compound according to claim 4, wherein X1 is phenyl optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 6. A compound according to claim 1, wherein X1 is heteroaryl optionally substituted with one or more substituents selected from halogen, hydroxy; or C1-6-alkyl, aryl, heteroaryl, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkylcarbonyl, C1-6-alkylsulfonyl, arylamido, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 7. A compound according to claim 6, wherein X1 is heteroaryl optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 8. A compound according to claim 7, wherein X1 is furyl, thienyl, pyrrolyl, thiazolyl, imidazolyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl or pyrimidinyl, each of which is optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 9. A compound according to claim 8, wherein X1 is pyridyl optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 10. A compound according to claim 1, wherein X2 is arylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. 11. A compound according to claim 10, wherein X2 is phenylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. 12. A compound according to claim 11, wherein X2 is phenylene. 13. A compound according to claim 1, wherein X2 is heteroarylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. 14. A compound according to claim 13 wherein X2 is furylene, thienylene, pyrrolylene, thiazolylene, imidazolylene, oxazolylene, isoxazolylene, isothiazolylene, pyridylene or pyrimidinylene, each of which is optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. 15. A compound according to claim 1, wherein X3 is aryl optionally substituted with one or more substituents selected from halogen, hydroxy; or C1-6-alkyl, aryl, heteroaryl, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkylcarbonyl, C1-6-alkylsulfonyl, arylamido, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 16. A compound according to claim 15, wherein X3 is aryl optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 17. A compound according to claim 15, wherein X3 is phenyl optionally substituted with one or more substituents selected from halogen, hydroxy; or C1-6-alkyl, aryl, heteroaryl, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkylcarbonyl, C1-6-alkylsulfonyl, arylamido, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 18. A compound according to claim 17, wherein X3 is phenyl optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 19. A compound according to claim 1, wherein X3 is heteroaryl optionally substituted with one or more substituents selected from halogen, hydroxy; or C1-6-alkyl, aryl, heteroaryl, C1-6-alkoxy, C1-6-alkylthio, C1-6-alkylcarbonyl, C1-6-alkylsulfonyl, arylamido, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 20. A compound according to claim 19, wherein X3 is heteroaryl optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 21. A compound according to claim 20, wherein X3 is furyl, thienyl, pyrrolyl, thiazolyl, imidazolyl, oxazolyl, isoxazolyl, isothiazolyl, pyridyl or pyrimidinyl, each of which is optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 22. A compound according to claim 21 wherein X3 is pyridyl optionally substituted with one or more substituents selected from C1-6-alkyl, phenyl, C1-6-alkylsulfonyl, C1-6-alkylaminocarbonyl or C1-6-dialkylaminocarbonyl each of which is optionally substituted with one or more halogens. 23. A compound according to claim 1, wherein X4 is arylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. 24. A compound according to claim 23, wherein X4 is phenylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. 25. A compound according to claim 24, wherein X4 is phenylene. 26. A compound according to claim 1, wherein X4 is heteroarylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. 27. A compound according to claim 26 wherein X4 is furylene, thienylene, pyrrolylene, thiazolylene, imidazolylene, oxazolylene, isoxazolylene, isothiazolylene, pyridylene or pyrimidinylene, each of which is optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. 28. A compound according to claim 1, wherein Ar is phenylene which is optionally substituted with one or more substituents selected from halogen, hydroxy or cyano; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-6-alkylthio, arylthio or C3-6-cycloalkylthio each of which is optionally substituted with one or more halogens. 29. A compound according to claim 28, wherein Ar is phenylene which is optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl, C1-6-alkoxy, aryloxy, aralkoxy or phenyl, each of which is optionally substituted with one or more halogens. 30. A compound according to claim 28, wherein Ar is phenylene which is optionally substituted with methyl or trifluoromethyl. 31. A compound according to claim 30, wherein Ar is phenylene. 32. A compound according to claim 1, wherein Y1 is S. 33. A compound according to claim 1, wherein Y1 is O. 34. A compound according to claim 1, wherein Y2 is O. 35. A compound according to claim 1, wherein Y2 is S. 36. A compound according to claim 1, wherein n is 1. 37. A compound according to claim 1, wherein R1 is hydrogen or a substituent selected from C1-6-alkyl, aralkyl, C1-6-alkoxy, aryloxy, aralkoxy each of which is optionally substituted with one or more halogens. 38. A compound according to claim 37, wherein R1 is hydrogen or a substituent selected from C1-6-alkyl, C1-6-alkoxy each of which is optionally substituted with one or more halogens. 39. A compound according to claim 38, wherein R1 is hydrogen. 40. A compound according to claim 1, wherein R2 is hydrogen. 41. A compound according to claim 1, which is selected from the following: [4-(3,3-Bis-biphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, and {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-2-trifluoromethyl-phenoxy}-acetic acid, or a salt thereof with a pharmaceutically acceptable acid or base, optical isomer or mixture of optical isomers, racemic mixture, or tautomeric forms thereof. 42. A compound according to claim 1, which is selected from the following: [4-(3,3-Bis-biphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, [4-(3,3-Bis-biphenyl-4-yl-allyloxy)-2-methyl-phenoxy]-acetic acid, (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-methyl-fu ran-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yd)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methyl-biphenyl-4-yd)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yd)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-tert-butyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yd)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yd)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, [4-(3,3-Bis-biphenyl-4-yl-allylsulfanyl)-phenoxy]-acetic acid, [4-(3,3-Bis-biphenyl-4-yl-allyloxy)-phenoxy]-acetic acid, (4-{3,3-Bis-[4-(5-methyl-fu ran-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(-furan-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allylsulfanyl]-phenoxy}-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-phenoxy)-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-phenoxy]-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-phenoxy]-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-phenoxy)-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allyloxy]-phenoxy}acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-tert-butyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid, {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid, [4-(3,3-Bis-biphenyl-4-yl-2-ethyl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, [4-(3,3-Bis-biphenyl-4-yl-2-ethyl-allyloxy)-2-methyl-phenoxy]-acetic acid, (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-2-ethyl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-2-ethyl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, (4-{3,3-Bis-[4-(4-methyl-fu ran-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-methyl-fu ran-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid, {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-tert-butyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, [4-(3,3-Bis-biphenyl-4-yl-2-ethoxy-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, [4-(3,3-Bis-biphenyl-4-yl-2-ethoxy-allyloxy)-2-methyl-phenoxy]-acetic acid, (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid(4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-2-ethoxy-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-2-ethoxy-allylsulfanyl)-2-methyl-phenoxy]-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl] 2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid, {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-tert-butyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid, [4-(3,3-Bis-biphenyl-4-yl-allylsulfanyl)-2-chloro-phenoxy]-acetic acid, [4-(3,3-Bis-biphenyl-4-yl-allyloxy)-2-chloro-phenoxy]-acetic acid, (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-allyloxy}-2-chloro-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(-furan-2-yl)-phenyl]-allyloxy}-2-chloro-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-allylsulfanyl}-2-chloro-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-allylsulfanyl}-2-chloro-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-chloro-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allyloxy}-2-chloro-phenoxy)-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-chloro-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-2-chloro-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-chloro-phenoxy)-acetic acid, (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-chloro-phenoxy)-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-chloro-phenoxy]-acetic acid, [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-chloro-phenoxy]-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid, and {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid, or a salt thereof with a pharmaceutically acceptable acid or base, optical isomer or mixture of optical isomers, racemic mixture, or tautomeric forms thereof. 43. A compound according to claim 1, which is a PPARδ agonist. 44. A compound according to claim 43, which is a selective PPARδ agonist. 45. A pharmaceutical composition comprising, as an active ingredient, at least one compound according to claim 1 together with one or more pharmaceutically acceptable carriers or excipients. 46. A pharmaceutical composition according to claim 45 in unit dosage form, comprising from about 0.05 mg to about 1000 mg per day of the compound. 47. A pharmaceutical composition according to claim 46 for oral, nasal, transdermal, pulmonal, or parenteral administration. 48. A method for the treatment of conditions mediated by the Peroxisome Proliferator-Activated Receptors (PPAR), wherein the condition is selected from type I diabetes, type II diabetes, impaired glucose tolerance (IGT), insulin resistence, dyslipidemin, Syndrome X, hypertension, obesity, hyperglyccmia, atherloselerosis, hyperlipidemia, coronary artery disease, myocardial ischemia, glomerulonephritis, glomeruloselerosis, nephritic syndrome, hypertensive nephrosclerosis, diabetic complications, loss of cognative function in dementia, psoriasis, Polycystic Ovarian Syndrome (PCOS), osteoporosis, progression from IGT to type II diabetes, and progression from non insulin-requiring type II diabetes to insulin-requiring type II diabetes, the method comprising administering to a subject in need thereof an effective amount of a compound according to claim 1 or a pharmaceutical composition comprising the same. 49. A method for the treatment of type I diabetes, type II diabetes, impaired glucose tolerance, insulin resistance or obesity, the method comprising administering to a subject in need thereof an effective amount of a compound according to claim 1 or of a pharmaceutical composition comprising the same. 50. The method according to claim 48 wherein the effective amount of the compound is in the range of from about 0.05 mg to about 1000 mg per day. 51. The method according to claim 49 wherein the effective amount of the compound is in the range of from about 0.05 mg to about 1000 mg per day. 52. A pharmaceutical composition according to claim 45 in unit dosage form, comprising from about 0.05 mg to about 1000 mg per day of the compound. 53. A pharmaceutical composition according to claim 45 in unit dosage form, comprising from about 0.5 mg to about 200 mg per day of the compound. 54. The method according to claim 48 wherein the effective amount of the compound is in the range of from about 0.1 to about 500 mg per day. 55. The method according to claim 48 wherein the effective amount of the compound is in the range of from about 0.5 mg to about 200 mg per day. 56. The method according to claim 49 wherein the effective amount of the compound is in the range of from about 0.1 to about 500 mg per day. 57. The method according to claim 49 wherein the effective amount of the compound is in the range of from about 0.5 mg to about 200 mg per day. 57 CROSS-REFERENCE TO RELATED APPLICATIONS This application claims priority under 35 U.S.C. 119 of Danish application no. PA 2002 01301 filed Sep. 5, 2002, Danish application no. PA 2003 00784 filed May 23, 2003 and U.S. application Ser. No. 60/409,814 filed Sep. 11, 2002, the contents of which are fully incorporated herein by reference. FIELD OF THE INVENTION The present invention relates to novel compounds, to the use of these compounds as pharmaceutical compositions, to pharmaceutical compositions comprising the compounds and to a method of treatment employing these compounds and compositions. More specifically, the compounds of the invention can be utilised in the treatment and/or prevention of conditions mediated by the Peroxisome Proliferator-Activated Receptors (PPAR), in particular the PPARδ subtype. BACKGROUND OF THE INVENTION Coronary artery disease (CAD) is the major cause of death in Type 2 diabetic and metabolic syndrome patients (i.e. patients that fall within the ‘deadly quartet’ category of impaired glucose tolerance, insulin resistance, hypertriglyceridaemia and/or obesity). The hypolipidaemic fibrates and antidiabetic thiazolidinediones separately display moderately effective triglyceride-lowering activities although they are neither potent nor efficacious enough to be a single therapy of choice for the dyslipidaemia often observed in Type 2 diabetic or metabolic syndrome patients. The thiazolidinediones also potently lower circulating glucose levels of Type 2 diabetic animal models and humans. However, the fibrate class of compounds are without beneficial effects on glycaemia. Studies on the molecular actions of these compounds indicate that thiazolidinediones and fibrates exert their action by activating distinct transcription factors of the peroxisome proliferator activated receptor (PPAR) family, resulting in increased and decreased expression of specific enzymes and apolipoproteins respectively, both key-players in regulation of plasma triglyceride content. Fibrates, on the one hand, are PPARα activators, acting primarily in the liver. Thiazolidinediones, on the other hand, are high affinity ligands for PPARγ acting primarily on adipose tissue. Adipose tissue plays a central role in lipid homeostasis and the maintenance of energy balance in vertebrates. Adipocytes store energy in the form of triglycerides during periods of nutritional affluence and release it in the form of free fatty acids at times of nutritional deprivation. The development of white adipose tissue is the result of a continuous differentiation process throughout life. Much evidence points to the central role of PPARγ activation in initiating and regulating this cell differentiation. Several highly specialised proteins are induced during adipocyte differentiation, most of them being involved in lipid storage and metabolism. The exact link from activation of PPARγ to changes in glucose metabolism, most notably a decrease in insulin resistance in muscle, has not yet been clarified. A possible link is via free fatty acids such that activation of PPARγ induces Lipoprotein Lipase (LPL), Fatty Acid Transport Protein (FATP) and Acyl-CoA Synthetase (ACS) in adipose tissue but not in muscle tissue. This, in turn, reduces the concentration of free fatty acids in plasma dramatically, and due to substrate competition at the cellular level, skeletal muscle and other tissues with high metabolic rates eventually switch from fatty acid oxidation to glucose oxidation with decreased insulin resistance as a consequence. PPARα is involved in stimulating β-oxidation of fatty acids. In rodents, a PPARα-mediated change in the expression of genes involved in fatty acid metabolism lies at the basis of the phenomenon of peroxisome proliferation, a pleiotropic cellular response, mainly limited to liver and kidney and which can lead to hepatocarcinogenesis in rodents. The phenomenon of peroxisome proliferation is not seen in man. In addition to its role in peroxisome proliferation in rodents, PPARα is also involved in the control of HDL cholesterol levels in rodents and humans. This effect is, at least partially, based on a PPARα-mediated transcriptional regulation of the major HDL apolipoproteins, apo A-I and apo A-II. The hypotriglyceridemic action of fibrates and fatty acids also involves PPARα and can be summarised as follows: (I) an increased lipolysis and clearance of remnant particles, due to changes in lipoprotein lipase and apo C-III levels, (II) a stimulation of cellular fatty acid uptake and their subsequent conversion to acyl-CoA derivatives by the induction of fatty acid binding protein and acyl-CoA synthase, (III) an induction of fatty acid β-oxidation pathways, (IV) a reduction in fatty acid and triglyceride synthesis, and finally (V) a decrease in VLDL production. Hence, both enhanced catabolism of triglyceride-rich particles as well as reduced secretion of VLDL particles constitutes mechanisms that contribute to the hypolipidemic effect of fibrates. PPARδ activation was initially reported not to be involved in modulation of glucose or triglyceride levels. (Berger et al., j. Biol. Chem., 1999, Vol 274, pp. 6718–6725). Later it has been shown that PPARδ activation leads to increased levels of HDL cholesterol in dbldb mice (Leibowitz et al. FEBS letters 2000, 473, 333–336). Further, a PPARδ agonist when dosed to insulin-resistant middle-aged obese rhesus monkeys caused a dramitic dose-dependent rise in serum HDL cholesterol while lowering the levels of small dense LDL, fasting triglycerides and fasting insulin (Oliver et al. PNAS 2001, 98, 5306–5311). The same paper also showed that PPARδ activation increased the reverse cholesterol transporter ATP-binding cassette A1 and induced apolipoprotein A1-specific cholesterol efflux. The involvement of PPARδ in fatty acid oxidation in muscles was further substantiated in PPARα knock-out mice. Muoio et al. (J. Biol. Chem. 2002, 277, 26089–26097) showed that the high levels of PPARδ in skeletal muscle can compensate for deficiency in PPARα. Taken together these observations suggest that PPARδ activation is useful in the treatment and prevention of cardiovascular diseases and conditions including atherosclerosis, hypertriglyceridemia, and mixed dyslipidaemia (WO 01/00603). A number of PPARδ compounds have been reported to be useful in the treatment of hyperglycemia, hyperlipidemia and hypercholesterolemia (WO 02/59098, WO 01/603, WO 01/25181, WO 02/14291, WO 01/79197, WO 99/4815, WO 97/28149, WO 98/27974, WO 97/28115, WO 97/27857, WO 97/28137, WO 97/27847). Glucose lowering as a single approach does not overcome the macrovascular complications associated with Type 2 diabetes and metabolic syndrome. Novel treatments of Type 2 diabetes and metabolic syndrome must therefore aim at lowering both the overt hypertriglyceridaemia associated with these syndromes as well as alleviation of hyperglycaemia. This indicate that research for compounds displaying various degree of PPARα, PPARγ and PPARδ activation should lead to the discovery of efficacious triglyceride and/or cholesterol and/or glucose lowering drugs that have great potential in the treatment of diseases such as type 2 diabetes, dyslipidemia, syndrome X (including the metabolic syndrome, i.e. impaired glucose tolerance, insulin resistance, hypertrigyceridaemia and/or obesity), cardiovascular diseases (including atherosclerosis) and hypercholesteremia. In WO 97/48674, various antimicrobial diaryls has been described as anti-infective agents. The invention comprises compounds of the formula: wherein L may be selected from the group consisting of N, CH and C; G, E may independently be selected from phenyl, substituted phenyl, phenylC1-4-alkyl, substituted phenylC1-4-alkyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-thienyl and 3-thienyl; J may be CH or O; X may be selected from the group consisting of is O, S, NR, and C(O)NR; Ar may be aryl or substituted aryl; W may be O or S; A may be selected from the group consisting of i.a. NRR, amidino, COOH; CHRCOOH, CH═CHR, CH═C(COOH)2; q, p may independently be are 0 or 1; and m, n may indenpendently be 0–6. Definitions In the structural formulas given herein and throughout the present specification the following terms have the indicated meaning: The term “C1-6-alkyl” as used herein, alone or in combination, represent a linear or branched, saturated hydrocarbon chain having the indicated number of carbon atoms. Representative examples include, but are not limited to methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, hexyl, isohexyl and the like. The term “C1-6-alkylcarbonyl as used herein, represents a “C1-6-alkyl” group as defined above having the indicated number of carbon atoms linked through a carbonyl group. Representative examples include, but are not limited to, methylcarbonyl, ethylcarbonyl, n-propylcarbonyl, isopropylcarbonyl, butylcarbonyl, isobutylcarbonyl, sec-butylcarbonyl, tert-butylcarbonyl, n-pentylcarbonyl, isopentylcarbonyl, neopentylcarbonyl, tert-pentylcarbonyl, n-hexylcarbonyl, isohexylcarbonyl and the like. The term “C1-6-alkylsulfonyl” as used herein refers to a monovalent substituent comprising a “C1-6-alkyl” group as defined above linked through a sulfonyl group. Representative examples include, but are not limited to, methylsulfonyl, ethylsulfonyl, n-propylsulfonyl, iso-propylsulfonyl, n-butylsulfonyl, isobutylsulfonyl, sec-butylsulfonyl, tert-butylsulfonyl, n-pentylsulfonyl, isopentylsulfonyl, neopentylsulfonyl, tert-pentylsulfonyl, n-hexylsulfonyl, iso-hexylsulfonyl and the like. The term “C1-6-alkylamido” as used herein, refers to an acyl group linked through an amino group; Representative examples include, but are not limited to acetylamino, propionyl-amino, butyrylamino, isobutyrylamino, pivaloylamino, valerylamino and the like. The term “C3-6-cycloalkyl” as used herein, alone or in combination, represent a saturated monocyclic hydrocarbon group having the indicated number of carbon atoms. Representative examples include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like. The term “C2-6-alkenyl” as used herein, represent an olefinically unsaturated branched or straight hydrocarbon group having from 2 to the specified number of carbon atoms and at least one double bond. Representative examples include, but are not limited to, vinyl, 1-propenyl, 2-propenyl, allyl, iso-propenyl, 1,3-butadienyl, 1-butenyl, hexenyl, pentenyl and the like. The term “C2-6-alkynyl” as used herein, represent an unsaturated branched or straight hydrocarbon group having from 2 to the specified number of carbon atoms and at least one triple bond. Representative examples include, but are not limited to, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl and the like. The term “C4-6-alkenynyl” as used herein, represent an unsaturated branched or straight hydrocarbon group having from 4 to the specified number of carbon atoms and both at least one double bond and at least one triple bond. Representative examples include, but are not limited to, 1-penten-4-ynyl, 3-penten-1-ynyl, 1,3-hexadiene-5-ynyl and the like. The term “C1-6-alkoxy” as used herein, alone or in combination, refers to a straight or branched configuration linked through an ether oxygen having its free valence bond from the ether oxygen. Examples of linear alkoxy groups are methoxy, ethoxy, propoxy, butoxy, pentoxy, hexoxy and the like. Examples of branched alkoxy are isopropoxy, sec-butoxy, tert-butoxy, isopentyloxy, isohexyloxy and the like. The term “C3-6-cycloalkoxy” as used herein, alone or in combination, represent a saturated monocyclic hydrocarbon group having the indicated number of carbon atoms linked through an ether oxygen having its free valence bond from the ether oxygen. Examples of cycloalkoxy groups are cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy and the like. The term “C1-6-alkylthio” as used herein, alone or in combination, refers to a straight or branched monovalent substituent comprising a “C1-6-alkyl” group as defined above linked through a divalent sulfur atom having its free valence bond from the sulfur atom and having 1 to 6 carbon atoms. Representative examples include, but are not limited to, methylthio, ethylthio, propylthio, butylthio, pentylthio and the like. The term “C3-6-cycloalkylthio” as used herein, alone or in combination, represent a saturated monocyclic hydrocarbon group having the indicated number of carbon atoms linked through a divalent sulfur atom having its free valence bond from the sulfur atom. Examples of cycloalkoxy groups are cyclopropylthio, cyclobutylthio, cyclopentylthio, cyclohexylthio and the like. The term “C1-6-alkylamino” as used herein, alone or in combination, refers to a straight or branched monovalent substituent comprising a “C1-6-alkyl” group as defined above linked through amino having a free valence bond from the nitrogen atom. Representative examples include, but are not limited to, methylamino, ethylamino, propylamino, butylamino, pentylamino and the like. The term “C1-6-alkylaminocarbonyl” as used herein refers to a monovalent substituent comprising a C1-monoalkylamino group linked through a carbonyl group such as e.g. methylaminocarbonyl, ethylaminocarbonyl, n-propylaminocarbonyl, isopropylaminocarbonyl, n-butylaminocarbonyl, sec-butylaminocarbonyl, isobutylaminocarbonyl, tert-butylaminocarbonyl, n-pentylaminocarbonyl, 2-methylbutylaminocarbonyl, 3-methylbutylaminocarbonyl, n-hexylaminocarbonyl, 4-methylpentylaminocarbonyl, neopentylaminocarbonyl, n-hexylaminocarbonyl and 2-2-dimethylpropylaminocarbonyl and the like. The term “C3-6-cycloalkylamino” as used herein, alone or in combination, represent a saturated monocyclic hydrocarbon group having the indicated number of carbon atoms linked through amino having a free valence bond from the nitrogen atom. Representative examples include, but are not limited to, cyclopropylamino, cyclobutylamino, cyclopentylamino, cyclohexylamino and the like. The term “C1-6-alkoxyC1-6-alkyl” as used herein, alone or in combination, refers to a “C1-6-alkyl” group as defined above whereto is attached a “C1-6-alkoxy” group as defined above. Representative examples include, but are not limited to, methoxymethyl, ethoxymethyl, methoxyethyl, ethoxyethyl and the like. The term “aryl” as used herein refers to an aromatic monocyclic or an aromatic fused bi- or tricyclic hydrocarbon group. Representative examples include, but are not limited to, phenyl, naphthyl, anthracenyl, phenanthrenyl, azulenyl and the like. The term “arylene” as used herein refers to divalent aromatic monocyclic or a divalent aromatic fused bi- or tricyclic hydrocarbon group. Representative examples include, but are not limited to, phenylene, naphthylene and the like. The term “arylcarbonyl” as used herein represents an “aryl” group as defined above linked through a carbonyl group. Representative examples include, but are not limited to, phenylcarbonyl, naphthylcarbonyl, anthracenylcarbonyl, phenanthrenylcarbonyl, azulenylcarbonyl and the like The term “arylsulfonyl” as used herein refers to an “aryl” group as defined above linked through a sulfonyl group. Representative examples include, but are not limited to, phenylsulfonyl, naphthylsulfonyl, anthracenylsulfonyl, phenanthrenylsulfonyl, azulenylsulfonyl, and the like. The term “arylamido” as used herein refers to an arylcarbonyl group linked through an amino group. Representative examples include, but are not limited to phenylcarbonylamino, naphthylcarbonylamino, anthracenylcarbonylamino, phenanthrenylcarbonylamino, azulenylcarbonylamino and the like. The term “halogen” means fluorine, chlorine, bromine or iodine. The term “perhalomethyl” means trifluoromethyl, trichloromethyl, tribromomethyl or triiodomethyl. The term “perhalomethoxy” means trifluoromethoxy, trichloromethoxy, tribromomethoxy or triiodomethoxy. The term “C1-6-dialkylamino” as used herein refers to an amino group wherein the two hydrogen atoms independently are substituted with a straight or branched, saturated hydrocarbon chain having the indicated number of carbon atoms. Representative examples include, but are not limited to, dimethylamino, N-ethyl-N-methylamino, diethylamino, dipropylamino, N-(n-butyl)-N-methylamino, di(n-pentyl)amino and the like. The term “C1-6-dialkylaminocarbonyl” as used herein refers to a monovalent substituent comprising a C1-1-dialkylamino group linked through a carbonyl group such as dimethyl-aminocarbonyl, N-ethyl-N-methylaminocarbonyl, diethylaminocarbonyl, dipropylaminocarbonyl, N-(n-butyl)-N-methylaminocarbonyl, di(n-pentyl)aminocarbonyl, and the like. The term “acyl” as used herein refers to a monovalent substituent comprising a “C1-6-alkyl” group as defined above linked through a carbonyl group. Representative examples include, but are not limited to, acetyl, propionyl, butyryl, isobutyryl, pivaloyl, valeryl and the like. The term “heteroaryl” as used herein, alone or in combination, refers to a monovalent substituent comprising a 5–7 membered monocyclic aromatic system or a 8–10 membered bicyclic aromatic system containing one or more heteroatoms selected from nitrogen, oxygen and sulfur, e.g. furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, isothiazolyl, isoxazolyl, oxazolyl, oxadiazolyl, thiadiazolyl, quinolyl, isoquinolyl, quinazolinyl, quinoxalinnyl, indolyl, benzimidazolyl, benzofuranyl, pteridinyl and purinyl and the like. The term “heteroarylene” as used herein, alone or in combination, refers to divalent 5–7 membered monocyclic aromatic system or a 8–10 membered bicyclic aromatic system containing one or more heteroatoms selected from nitrogen, oxygen and sulfur, e.g. furylene, thienylene, pyrrolylene, imidazolylene, pyrazolylene, triazolylene, pyrazinylene, pyrimidinylene, pyridazinylene, isothiazolylene, isoxazolylene, oxazolylene, oxadiazolylene, thiadiazolylene, quinolylene, isoquinolylene, quinazolinylene, quinoxalinnylene, indolylene, benzimidazolylene, benzofuranylene, pteridinylene and purinylene and the like. The term “heteroaryloxy” as used herein, alone or in combination, refers to a heteroaryl as defined herein linked to an oxygen atom having its free valence bond from the oxygen atom e.g. pyrrolyloxy, imidazolyloxy, pyrazolyloxy, triazolyloxy, pyrazinyloxy, pyrimidinyloxy, pyridazinyloxy, isothiazolyloxy, isoxazolyloxy, oxazolyloxy, oxadiazolyloxy, thiadiazolyloxy, quinolinyloxy, isoquinolinyloxy, quinazolinyloxy, quinoxalinyloxy, indoltloxy, benzimidazolyloxy, benzofuranyloxy, pteridinyloxy and purinyloxy and the like. The term “aralkyl” as used herein refers to a straight or branched saturated carbon chain containing from 1 to 6 carbons substituted with an aromatic carbohydride. Representative examples include, but are not limited to, benzyl, phenethyl, 3-phenylpropyl, 1-naphthyl-methyl, 2-(1-naphthyl)ethyl and the like. The term “aryloxy” as used herein refers to phenoxy, 1-naphthyloxy, 2-naphthyloxy and the like. The term “aralkoxy” as used herein refers to a C1-6-alkoxy group substituted with an aromatic carbohydride, such as benzyloxy, phenethoxy, 3-phenylpropoxy, 1-naphthyl-methoxy, 2-(1-naphtyl)ethoxy and the like. The term “heteroaralkyl” as used herein refers to a straight or branched saturated carbon chain containing from 1 to 6 carbons substituted with a heteroaryl group; such as (2-furyl)methyl, (3-furyl)methyl, (2-thienyl)methyl, (3-thienyl)methyl, (2-pyridyl)methyl, 1-methyl-1-(2-pyrimidyl)ethyl and the like. The term “heteroaralkoxy” as used herein refers to a heteroarylalkyl as defined herein linked to an oxygen atom having its free valence bond from the oxygen atom. Representative examples include, but are not limited to, (2-furyl)methyl, (3-furyl)methyl, (2-thienyl)methyl, (3-thienyl)methyl, (2-pyridyl)methyl, 1-methyl-1-(2-pyrimidyl)ethyl linked to oxygen, and the like. The term “arylthio” as used herein, alone or in combination, refers to an aryl group linked through a divalent sulfur atom having its free valence bond from the sulfur atom, the aryl group optionally being mono- or polysubstituted with C1-6-alkyl, halogen, hydroxy or C1-6-alkoxy. Representative examples include, but are not limited to, phenylthio, (4-methylphenyl)-thio, (2-chlorophenyl)thio and the like. Certain of the above defined terms may occur more than once in the structural formulae, and upon such occurrence each term shall be defined independently of the other. The term “optionally substituted” as used herein means that the groups in question are either unsubstituted or substituted with one or more of the substituents specified. When the groups in question are substituted with more than one substituent the substituents may be the same or different. DESCRIPTION OF THE INVENTION The present invention relates to compounds of the general formula (1): wherein X1 is aryl or heteroaryl each of which is optionally substituted with one or more substituents selected from halogen, hydroxy, cyano, amino or carboxy; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-6-alkylthio, arylthio, C3-6-cycloalkylthio, C1-6-alkylcarbonyl, arylcarbonyl, C1-6-alkylsulfonyl, arylsulfonyl, C1-6-alkylamido, arylamido, C1-6-alkylaminocarbonyl, C1-6-alkylamino, C1-6-dialkylamino or C3-6-cycloalkylamino each of which is optionally substituted with one or more halogens; and X2 is arylene or heteroarylene each of which is optionally substituted with one or more substituents selected from halogen, hydroxy, cyano, amino or carboxy; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-6-alkylthio, C3-6-cycloalkylthio, C1-6-alkylamino, C1-6-dialkylamino or C3-6-cycloalkylamino each of which is optionally substituted with one or more halogens; and X3 is aryl or heteroaryl each of which is optionally substituted with one or more substituents selected from halogen, hydroxy, cyano, amino or carboxy; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-6-alkylthio, arylthio, C3-6-cycloalkylthio, C1-6-alkylcarbonyl, arylcarbonyl, C1-6-alkylsulfonyl, arylsulfonyl, C1-6-alkylamido, arylamido, C1-6-alkylaminocarbonyl, C1-6-alkylamino, C1-6-dialkylamino or C3-6-cycloalkylamino each of which is optionally substituted with one or more halogens; and X4 is arylene or heteroarylene each of which is optionally substituted with one or more substituents selected from halogen, hydroxy, cyano, amino or carboxy; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, C1-6-alkoxy, C3-6-cycloalkoxy, C1-6-alkylthio, C3-6-cycloalkylthio, C1-6-alkylamino, C1-6-dialkylamino or C3-6-cycloalkylamino each of which is optionally substituted with one or more halogens; and Ar is arylene which is optionally substituted with one or more substituents selected from halogen, hydroxy or cyano; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-6-alkylthio, arylthio or C3-6-cycloalkylthio each of which is optionally substituted with one or more halogens; and Y1 is O or S; and Y2 is O or S; and Z is —(CH2)n— wherein n is 1, 2 or 3; and R1 is hydrogen, halogen or a substituent selected from C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aralkyl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-1-alkylthio, arylthio or C3-6-cycloalkylthio each of which is optionally substituted with one or more halogens; and R2 is hydrogen, C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, C4-6-alkenynyl or aryl; or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable solvate thereof, or any tautomeric forms, stereoisomers, mixture of stereoisomers including a racemic mixture, or polymorphs. In one embodiment, the present invention is concerned with compounds of formula (I) wherein X1 is aryl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl, aryl, C1-6-alkoxy or C1-6-alkylsulfonyl each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein XI is aryl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X1 is phenyl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl, aryl, C1-6-alkoxy or C1-6-alkylsulfonyl each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X1 is phenyl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X1 is heteroaryl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl, aryl, C1-6-alkoxy or C1-6-alkylsulfonyl each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X1 is heteroaryl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X1 is furyl or thienyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X2 is arylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X2 is phenylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X2 is phenylene. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X3 is aryl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl, aryl, C1-6-alkoxy or C1-6-alkylsulfonyl each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X3 is aryl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X3 is phenyl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl, aryl, C1-6-alkoxy or C1-6-alkylsulfonyl each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X3 is phenyl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X3 is heteroaryl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl, aryl, C1-6-alkoxy or C1-6-alkylsulfonyl each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X3 is heteroaryl optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X3 is furyl or thienyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X4 is arylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X4 is phenylene optionally substituted with one or more substituents selected from halogen or C1-6-alkyl optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein X4 is phenylene. In another embodiment, the present invention is concerned with compounds of formula (I) wherein Ar is phenylene which is optionally substituted with one or more substituents selected from halogen, hydroxy or cyano; or C1-6-alkyl, C3-6-cycloalkyl, C2-6-alkenyl, C2-6-alkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, C1-6-alkoxy, C3-6-cycloalkoxy, aryloxy, aralkoxy, heteroaralkoxy, C1-6-alkylthio, arylthio or C3-6-cycloalkylthio each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein Ar is phenylene which is optionally substituted with one or more substituents selected from halogen; or C1-6-alkyl, C1-6-alkoxy, aryloxy or aralkoxy each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein Ar is phenylene which is optionally substituted with methyl. In another embodiment, the present invention is concerned with compounds of formula (I) wherein Ar is phenylene. In another embodiment, the present invention is concerned with compounds of formula (I) wherein Y1 is S. In another embodiment, the present invention is concerned with compounds of formula (I) wherein Y2 is O. In another embodiment, the present invention is concerned with compounds of formula (I) wherein n is 1. In another embodiment, the present invention is concerned with compounds of formula (I) wherein R1 is hydrogen or a substituent selected from C1-6-alkyl, aralkyl, C1-6-alkoxy, aryloxy, aralkoxy each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein R1 is hydrogen or a substituent selected from C1-6-alkyl, C1-6-alkoxy each of which is optionally substituted with one or more halogens. In another embodiment, the present invention is concerned with compounds of formula (I) wherein R1 is hydrogen. In another embodiment, the present invention is concerned with compounds of formula (I) wherein R2 is hydrogen. In another embodiment, the present invention is concerned with compounds of formula I wherein alkyl is methyl or ethyl. In another embodiment, the present invention is concerned with compounds of formula I wherein alkenyl is vinyl or 1-propenyl. In another embodiment, the present invention is concerned with compounds of formula I wherein alkynyl is 1-propynyl. In another embodiment, the present invention is concerned with compounds of formula I wherein alkenynyl is 1-pentene-4-yne. In another embodiment, the present invention is concerned with compounds of formula I wherein alkoxy is methoxy, ethoxy, isopropoxy or cyclopropoxy. In another embodiment, the present invention is concerned with compounds of formula I wherein aryl is phenyl. In another embodiment, the present invention is concerned with compounds of formula I wherein arylene is phenylene. In another embodiment, the present invention is concerned with compounds of formula I wherein halogen is fluorine or chlorine. In another embodiment, the present invention is concerned with compounds of formula I wherein perhalomethyl is trifluoromethyl. In another embodiment, the present invention is concerned with compounds of formula I wherein perhalomethoxy is trifluoromethoxy, In another embodiment, the present invention is concerned with compounds of formula I wherein heteroaryl is furyl or thienyl. In another embodiment, the present invention is concerned with compounds of formula I wherein aralkyl is benzyl. In another embodiment, the present invention is concerned with compounds of formula I wherein aryloxy is phenoxy. In another embodiment, the present invention is concerned with compounds of formula I wherein aralkoxy is benzyloxy. In another embodiment, the present invention is concerned with compounds of formula I wherein the substituents R1 and X4 are arranged in a trans-configuration. In another embodiment, the present invention is concerned with compounds of formula I wherein the substituents R1 and X4 are arranged in a cis-configuration. In another embodiment, the present invention is concerned with compounds of formula I which are PPARδ agonists. In another embodiment, the present invention is concerned with compounds of formula I which are selective PPARδ agonists. Examples of compounds of the invention are: [4-(3,3-Bis-biphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid [4-(3,3-Bis-biphenyl-4-yl-allyloxy)-2-methyl-phenoxy]-acetic acid (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yd)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allyoxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-2-trifluoromethyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methyl-biphenyl-4-yd)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yd)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-tert-butyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yd)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-allysulfanyl]-2-methyl-phenoxy}-acetic acid [4-(3,3-Bis-biphenyl-4-yl-allylsulfanyl)-phenoxy]-acetic acid [4-(3,3-Bis-biphenyl-4-yl-allyloxy)-phenoxy]-acetic acid (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allylsulfanyl]-phenoxy}-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-phenoxy)-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-phenoxy]-acetic acid [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-phenoxy]-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-phenoxy)-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-tert-butyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-phenoxy}-acetic acid {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-allylsulfanyl]-phenoxy}-acetic acid [4-(3,3-Bis-biphenyl-4-yl-2-ethyl-allysulfanyl)-2-methyl-phenoxy]-acetic acid [4-(3,3-Bis-biphenyl-4-yl-2-ethyl-allyloxy)-2-methyl-phenoxy]-acetic acid (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-2-ethyl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-2-ethyl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (4-{3,3-Bis-[4-(4-methyl-fu ran-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-methyl-phenoxy)-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-tert-butyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethyl-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethyl-allylsulfanyl]-2-methyl-phenoxy}-acetic acid [4-(3,3-Bis-biphenyl-4-yl-2-ethoxy-allylsulfanyl)-2-methyl-phenoxy]-acetic acid [4-(3,3-Bis-biphenyl-4-yl-2-ethoxy-allyloxy)-2-methyl-phenoxy]-acetic acid (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-methyl-fu ran-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid(4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-2-ethoxy-allylsulfanyl)-2-methyl-phenoxy]-acetic acid [4-(3,3-Bis-[1,1′;4′,1”]terphenyl-4-yl-2-ethoxy-allylsulfanyl)-2-methyl-phenoxy]-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (4-{3,3-Bis-[4-(4-methyl-fu ran-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-methyl-furan-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-chloro-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl] 2-ethoxy-allyloxy}-2-methyl-phenoxy)-acetic acid (4-{3,3-Bis-[4-(4-acetyl-thiophen-2-yl)-phenyl]-2-ethoxy-allylsulfanyl}-2-methyl-phenoxy)-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-tert-butyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-acetyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-phenoxy-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(3′-isopropyl-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,4′-difluoro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′,5′-dichloro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethoxy-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(2′-chloro-4′-fluoro-biphenyl-4-yl)-2-ethoxy-allylsulfanyl]-2-methyl-phenoxy}-acetic acid [4-(3,3-Bis-biphenyl-4-yl-allylsulfanyl)-2-chloro-phenoxy]-acetic acid [4-(3,3-Bis-biphenyl-4-yl-allyloxy)-2-chloro-phenoxy]-acetic acid (4-{3,3-Bis-[4-(5-methyl-fu ran-2-yl)-phenyl]-allyloxy}-2-chloro-phenoxy)-acetic acid (4-{3,3-Bis-[4-(-furan-2-yl)-phenyl]-allyloxy}-2-chloro-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-methyl-furan-2-yl)-phenyl]-allylsulfanyl}-2-chloro-phenoxy)-acetic acid (4-{3,3-Bis-[4-(furan-2-yl)-phenyl]-allylsulfanyl}-2-chloro-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-chloro-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-chloro-thiophen-2-yl)-phenyl]-allyloxy}-2-chloro-phenoxy)-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4-thiophen-2-yl-phenyl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allylsulfanyl}-2-chloro-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-allyloxy}-2-chloro-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allyloxy}-2-chloro-phenoxy)-acetic acid (4-{3,3-Bis-[4-(5-acetyl-thiophen-2-yl)-phenyl]-2-ethyl-allylsulfanyl}-2-chloro-phenoxy)-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(3′-methylsulfanyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(3′-amino-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-amino-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4-pyridin-2-yl-phenyl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methoxy-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethoxy-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-methyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-trifluoromethyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-chloro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-tert-butyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-chloro-phenoxy]-acetic acid [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-chloro-phenoxy]-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-acetyl-biphenyl-4-yl)-allyloxy]-2-methyl-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-phenoxy-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(4′-isopropyl-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(3′,4′-difluoro-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(3′,5′-dichloro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allyloxy]-2-chloro-phenoxy}-acetic acid {4-[3,3-Bis-(3′-chloro-4′-fluoro-biphenyl-4-yl)-allylsulfanyl]-2-chloro-phenoxy}-acetic acid or a salt thereof with a pharmaceutically acceptable acid or base, or any optical isomer or mixture of optical isomers, including a racemic mixture, or any tautomeric forms. The present invention also encompasses pharmaceutically acceptable salts of the present compounds. Such salts include pharmaceutically acceptable acid addition salts, pharmaceutically acceptable base addition salts, pharmaceutically acceptable metal salts, ammonium and alkylated ammonium salts. Acid addition salts include salts of inorganic acids as well as organic acids. Representative examples of suitable inorganic acids include hydrochloric, hydrobromic, hydroiodic, phosphoric, sulfuric, nitric acids and the like. Representative examples of suitable organic acids include formic, acetic, trichloroacetic, trifluoroacetic, propionic, benzoic, cinnamic, citric, fumaric, glycolic, lactic, maleic, malic, malonic, mandelic, oxalic, picric, pyruvic, salicylic, succinic, methanesulfonic, ethanesulfonic, tartaric, ascorbic, pamoic, bismethylene salicylic, ethanedisulfonic, gluconic, citraconic, aspartic, stearic, palmitic, EDTA, glycolic, p-aminobenzoic, glutamic, benzenesulfonic, p-toluenesulfonic acids, sulphates, nitrates, phosphates, perchlorates, borates, acetates, benzoates, hydroxynaphthoates, glycerophosphates, ketoglutarates and the like. Further examples of pharmaceutically acceptable inorganic or organic acid addition salts include the pharmaceutically acceptable salts listed in J. Pharm. Sci. 1977, 66, 2, which is incorporated herein by reference. Examples of metal salts include lithium, sodium, potassium, magnesium, zinc, calcium salts and the like. Examples of amines and organic amines include ammonium, methylamine, dimethylamine, trimethylamine, ethylamine, diethylamine, propylamine, butylamine, tetramethylamine, ethanolamine, diethanolamine, triethanolamine, meglumine, ethylenediamine, choline, N,N′-dibenzylethylenediamine, N-benzylphenylethylamine, N-methyl-D-glucamine, guanidine and the like. Examples of cationic amino acids include lysine, arginine, histidine and the like. The pharmaceutically acceptable salts are prepared by reacting the compound of formula I with 1 to 4 equivalents of a base such as sodium hydroxide, sodium methoxide, sodium hydride, potassium t-butoxide, calcium hydroxide, magnesium hydroxide and the like, in solvents like ether, THF, methanol, t-butanol, dioxane, isopropanol, ethanol etc. Mixture of solvents may be used. Organic bases like lysine, arginine, diethanolamine, choline, guandine and their derivatives etc. may also be used. Alternatively, acid addition salts wherever applicable are prepared by treatment with acids such as hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, p-toluenesulphonic acid, methanesulfonic acid, acetic acid, citric acid, maleic acid salicylic acid, hydroxynaphthoic acid, ascorbic acid, palmitic acid, succinic acid, benzoic acid, benzenesulfonic acid, tartaric acid and the like in solvents like ethyl acetate, ether, alcohols, acetone, THF, dioxane etc. Mixture of solvents may also be used. The stereoisomers of the compounds forming part of this invention may be prepared by using reactants in their single enantiomeric form in the process wherever possible or by conducting the reaction in the presence of reagents or catalysts in their single enantiomer form or by resolving the mixture of stereoisomers by conventional methods. Some of the preferred methods include use of microbial resolution, enzymatic resolution, resolving the diastereomeric salts formed with chiral acids such as mandelic acid, camphorsulfonic acid, tartaric acid, lactic acid, and the like wherever applicable or chiral bases such as brucine, (R)- or (S)-phenylethylamine, cinchona alkaloids and their derivatives and the like. Commonly used methods are compiled by Jaques et al in “Enantiomers, Racemates and Resolution” (Wiley Interscience, 1981). More specifically the compound of formula I may be converted to a 1:1 mixture of diastereomeric amides by treating with chiral amines, aminoacids, aminoalcohols derived from aminoacids; conventional reaction conditions may be employed to convert acid into an amide; the dia-stereomers may be separated either by fractional crystallization or chromatography and the stereoisomers of compound of formula I may be prepared by hydrolysing the pure diastereomeric amide. Various polymorphs of compound of general formula I forming part of this invention may be prepared by crystallization of compound of formula I under different conditions. For example, using different solvents commonly used or their mixtures for recrystallization; crystallizations at different temperatures; various modes of cooling, ranging from very fast to very slow cooling during crystallizations. Polymorphs may also be obtained by heating or melting the compound followed by gradual or fast cooling. The presence of polymorphs may be determined by solid probe nmr spectroscopy, ir spectroscopy, differential scanning calorimetry, powder X-ray diffraction or such other techniques. The invention also encompasses prodrugs of the present compounds, which on administration undergo chemical conversion by metabolic processes before becoming active pharmacological substances. In general, such prodrugs will be functional derivatives of the present compounds, which are readily convertible in vivo into the required compound of the formula (I). Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in “Design of Prodrugs”, ed. H. Bundgaard, Elsevier, 1985. The invention also encompasses active metabolites of the present compounds. The invention also relates to pharmaceutical compositions comprising, as an active ingredient, at least one compound of the formula I or any optical or geometric isomer or tautomeric form thereof including mixtures of these or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers or diluents. Furthermore, the invention relates to the use of compounds of the general formula I or their tautomeric forms, their stereoisomers, their polymorphs, their pharmaceutically acceptable salts or pharmaceutically acceptable solvates thereof for the preparation of a pharmaceutical composition for the treatment and/or prevention of conditions mediated by nuclear receptors, in particular the Peroxisome Proliferator-Activated Receptors (PPAR) such as the conditions mentioned above. In another aspect, the present invention relates to a method of treating and/or preventing Type I or Type II diabetes. In a still further aspect, the present invention relates to the use of one or more compounds of the general formula I or pharmaceutically acceptable salts thereof for the preparation of a pharmaceutical composition for the treatment and/or prevention of Type I or Type II diabetes. In a still further aspect, the present compounds are useful for the treatment and/or prevention of IGT. In a still further aspect, the present compounds are useful for the treatment and/or prevention of Type 2 diabetes. In a still further aspect, the present compounds are useful for the delaying or prevention of the progression from IGT to Type 2 diabetes. In a still further aspect, the present compounds are useful for the delaying or prevention of the progression from non-insulin requiring Type 2 diabetes to insulin requiring Type 2 diabetes. In another aspect, the present compounds reduce blood glucose and triglyceride levels and are accordingly useful for the treatment and/or prevention of ailments and disorders such as diabetes and/or obesity. In still another aspect, the present compounds are useful for the treatment and/or prophylaxis of insulin resistance (Type 2 diabetes), impaired glucose tolerance, dyslipidemia, disorders related to Syndrome X such as hypertension, obesity, insulin resistance, hyperglycaemia, atherosclerosis, hyperlipidemia, coronary artery disease, myocardial ischemia and other cardiovascular disorders. In still another aspect, the present compounds are effective in decreasing apoptosis in mammalian cells such as beta cells of Islets of Langerhans. In still another aspect, the present compounds are useful for the treatment of certain renal diseases including glomerulonephritis, glomerulosclerosis, nephrotic syndrome, hypertensive nephrosclerosis. In still another aspect, the present compounds may also be useful for improving cognitive functions in dementia, treating diabetic complications, psoriasis, polycystic ovarian syndrome (PCOS) and prevention and treatment of bone loss, e.g. osteoporosis. The present compounds may also be administered in combination with one or more further pharmacologically active substances eg., selected from antiobesity agents, antidiabetics, antihypertensive agents, agents for the treatment and/or prevention of complications resulting from or associated with diabetes and agents for the treatment and/or prevention of complications and disorders resulting from or associated with obesity. Thus, in a further aspect of the invention the present compounds may be administered in combination with one or more antiobesity agents or appetite regulating agents. Such agents may be selected from the group consisting of CART (cocaine amphetamine regulated transcript) agonists, NPY (neuropeptide Y) antagonists, MC4 (melanocortin 4) agonists, orexin antagonists, TNF (tumor necrosis factor) agonists, CRF (corticotropin releasing factor) agonists, CRF BP (corticotropin releasing factor binding protein) antagonists, urocortin agonists, β3 agonists, MSH (melanocyte-stimulating hormone) agonists, MCH (melanocyte-concentrating hormone) antagonists, CCK (cholecystokinin) agonists, serotonin re-uptake inhibitors, serotonin and noradrenaline re-uptake inhibitors, mixed serotonin and noradrenergic compounds, 5HT (serotonin) agonists, bombesin agonists, galanin antagonists, growth hormone, growth hormone releasing compounds, TRH (thyreotropin releasing hormone) agonists, UCP 2 or 3 (uncoupling protein 2 or 3) modulators, leptin agonists, DA agonists (bromocriptin, doprexin), lipase/amylase inhibitors, RXR (retinoid X receptor) modulators or TR β agonists. In one embodiment of the invention the antiobesity agent is leptin. In another embodiment the antiobesity agent is dexamphetamine or amphetamine. In another embodiment the antiobesity agent is fenfluramine or dexfenfluramine. In still another embodiment the antiobesity agent is sibutramine. In a further embodiment the antiobesity agent is orlistat. In another embodiment the antiobesity agent is mazindol or phentermine. Suitable antidiabetics comprise insulin, GLP-1 (glucagon like peptide-1) derivatives such as those disclosed in WO 98/08871 to Novo Nordisk A/S, which is incorporated herein by reference as well as orally active hypoglycaemic agents. The orally active hypoglycaemic agents preferably comprise sulphonylureas, biguanides, meglitinides, glucosidase inhibitors, glucagon antagonists such as those disclosed in WO 99/01423 to Novo Nordisk A/S and Agouron Pharmaceuticals, Inc., GLP-1 agonists, potassium channel openers such as those disclosed in WO 97/26265 and WO 99/03861 to Novo Nordisk A/S which are incorporated herein by reference, DPP-IV (dipeptidyl peptidase-IV) inhibitors, inhibitors of hepatic enzymes involved in stimulation of gluconeogenesis and/or glycogenolysis, glucose uptake modulators, compounds modifying the lipid metabolism such as antihyperlipidemic agents and antilipidemic agents as HMG CoA inhibitors (statins), compounds lowering food intake, RXR agonists and agents acting on the ATP-dependent potassium channel of the β-cells. In one embodiment of the invention the present compounds are administered in combination with insulin. In a further embodiment the present compounds are administered in combination with a sulphonylurea eg. tolbutamide, glibenclamide, glipizide or glicazide. In another embodiment the present compounds are administered in combination with a biguanide eg. metformin. In yet another embodiment the present compounds are administered in combination with a meglitinide eg. repaglinide or senaglinide. In a further embodiment the present compounds are administered in combination with an α-glucosidase inhibitor eg. miglitol or acarbose. In another embodiment the present compounds are administered in combination with an agent acting on the ATP-dependent potassium channel of the β-cells eg. tolbutamide, glibenclamide, glipizide, glicazide or repaglinide. Furthermore, the present compounds may be administered in combination with nateglinide. In still another embodiment the present compounds are administered in combination with an antihyperlipidemic agent or antilipidemic agent eg. cholestyramine, colestipol, clofibrate, gemfibrozil, lovastatin, pravastatin, simvastatin, probucol or dextrothyroxine. In a further embodiment the present compounds are administered in combination with more than one of the above-mentioned compounds eg. in combination with a sulphonylurea and metformin, a sulphonylurea and acarbose, repaglinide and metformin, insulin and a sulphonylurea, insulin and metformin, insulin, insulin and lovastatin, etc. Furthermore, the present compounds may be administered in combination with one or more antihypertensive agents. Examples of antihypertensive agents are β-blockers such as alprenolol, atenolol, timolol, pindolol, propranolol and metoprolol, ACE (angiotensin converting enzyme) inhibitors such as benazepril, captopril, enalapril, fosinopril, lisinopril, quinapril and ramipril, calcium channel blockers such as nifedipine, felodipine, nicardipine, isradipine, nimodipine, diltiazem and verapamil, and α-blockers such as doxazosin, urapidil, prazosin and terazosin. Further reference can be made to Remington: The Science and Practice of Pharmacy, 19th Edition, Gennaro, Ed., Mack Publishing Co., Easton, Pa., 1995. It should be understood that any suitable combination of the compounds according to the invention with one or more of the above-mentioned compounds and optionally one or more further pharmacologically active substances are considered to be within the scope of the present invention. The present invention also relates to a process for the preparation of the above said novel compounds, their derivatives, their analogs, their tautomeric forms, their stereoisomers, their polymorphs, their pharmaceutically acceptable salts or pharmaceutically acceptable solvates. Pharmaceutical Compositions The compounds of the invention may be administered alone or in combination with pharmaceutically acceptable carriers or excipients, in either single or multiple doses. The pharmaceutical compositions according to the invention may be formulated with pharmaceutically acceptable carriers or diluents as well as any other known adjuvants and excipients in accordance with conventional techniques such as those disclosed in Remington: The Science and Practice of Pharmacy, 19th Edition, Gennaro, Ed., Mack Publishing Co., Easton, Pa., 1995. The compositions may appear in conventional forms, for example capsules, tablets, aerosols, solutions, suspensions or topical applications. Typical compositions include a compound of formula I or a pharmaceutically acceptable acid addition salt thereof, associated with a pharmaceutically acceptable excipient which may be a carrier or a diluent or be diluted by a carrier, or enclosed within a carrier which can be in the form of a capsule, sachet, paper or other container. In making the compositions, conventional techniques for the preparation of pharmaceutical compositions may be used. For example, the active compound will usually be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier which may be in the form of a ampoule, capsule, sachet, paper, or other container. When the carrier serves as a diluent, it may be solid, semi-solid, or liquid material which acts as a vehicle, excipient, or medium for the active compound. The active compound can be adsorbed on a granular solid container for example in a sachet. Some examples of suitable carriers are water, salt solutions, alcohols, polyethylene glycols, polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatine, lactose, terra alba, sucrose, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid or lower alkyl ethers of cellulose, silicic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose and polyvinylpyrrolidone. Similarly, the carrier or diluent may include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax. The formulations may also include wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavouring agents. The formulations of the invention may be formulated so as to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art. The pharmaceutical compositions can be sterilized and mixed, if desired, with auxiliary agents, emulsifiers, salt for influencing osmotic pressure, buffers and/or colouring substances and the like, which do not deleteriously react with the active compounds. The route of administration may be any route, which effectively transports the active compound to the appropriate or desired site of action, such as oral, nasal, pulmonary, transdermal or parenteral e.g. rectal, depot, subcutaneous, intravenous, intraurethral, intramuscular, intranasal, ophthalmic solution or an ointment, the oral route being preferred. If a solid carrier is used for oral administration, the preparation may be tabletted, placed in a hard gelatin capsule in powder or pellet form or it can be in the form of a troche or lozenge. If a liquid carrier is used, the preparation may be in the form of a syrup, emulsion, soft gelatin capsule or sterile injectable liquid such as an aqueous or non-aqueous liquid suspension or solution. For nasal administration, the preparation may contain a compound of formula I dissolved or suspended in a liquid carrier, in particular an aqueous carrier, for aerosol application. The carrier may contain additives such as solubilizing agents, e.g. propylene glycol, surfactants, absorption enhancers such as lecithin (phosphatidylcholine) or cyclodextrin, or preservatives such as parabenes. For parenteral application, particularly suitable are injectable solutions or suspensions, preferably aqueous solutions with the active compound dissolved in polyhydroxylated castor oil. Tablets, dragees, or capsules having talc and/or a carbohydrate carrier or binder or the like are particularly suitable for oral application. Preferable carriers for tablets, dragees, or capsules include lactose, corn starch, and/or potato starch. A syrup or elixir can be used in cases where a sweetened vehicle can be employed. A typical tablet which may be prepared by conventional tabletting techniques may contain: Core: Active compound (as free compound or salt thereof) 5 mg Colloidal silicon dioxide (Aerosil) 1.5 mg Cellulose, microcryst. (Avicel) 70 mg Modified cellulose gum (Ac-Di-Sol) 7.5 mg Magnesium stearate Ad. Coating: HPMC approx. 9 mg *Mywacett 9–40 T approx. 0.9 mg *Acylated monoglyceride used as plasticizer for film coating. If desired, the pharmaceutical composition of the invention may comprise the compound of formula (I) in combination with further pharmacologically active substances such as those described in the foregoing. The compounds of the invention may be administered to a mammal, especially a human in need of such treatment, prevention, elimination, alleviation or amelioration of diseases related to the regulation of blood sugar. Such mammals include also animals, both domestic animals, e.g. household pets, and non-domestic animals such as wildlife. The compounds of the invention are effective over a wide dosage range. A typical oral dosage is in the range of from about 0.001 to about 100 mg/kg body weight per day, preferably from about 0.01 to about 50 mg/kg body weight per day, and more preferred from about 0.05 to about 10 mg/kg body weight per day administered in one or more dosages such as 1 to 3 dosages. The exact dosage will depend upon the frequency and mode of administration, the sex, age, weight and general condition of the subject treated, the nature and severity of the condition treated and any concomitant diseases to be treated and other factors evident to those skilled in the art. The formulations may conveniently be presented in unit dosage form by methods known to those skilled in the art. A typical unit dosage form for oral administration one or more times per day such as 1 to 3 times per day may contain of from 0.05 to about 1000 mg, preferably from about 0.1 to about 500 mg, and more preferred from about 0.5 mg to about 200 mg. Any novel feature or combination of features described herein is considered essential to this invention. The invention also relate to methods of preparing the above mentioned compounds, comprising: General Procedure (A) Step A: Reacting a compound of formula II wherein X1, X2, X3 and X4 are defined as above, through a Wittig-like process with for example (EtO)2PO(CHR1)COOR6 (wherein R6 is an alkyl group), in the presence of a base such as sodium hydride, EtONa and the like to give a compound of formula III wherein X1, X2, X3, X4, R1 and R6 are defined as above Step B: Reducing the compound of formula III, wherein X1, X2, X3, X4, R1 and R6 are defined as above with a suitable reagent such as diisobutylaluminium hydride, to give a compound of formula IV wherein X1, X2, X3, X4 and R1 are defined as above, and Step C: Reacting the compound of formula IV, wherein X1, X2, X3, X4, and R1 are defined as above, (except that when X1, X2, X3 or X4, are substituted with hydroxy, this functionality has to be protected) with a compound of formula V wherein Y1, Ar, Y2, Z and R2 are defined as above, except that R2 is not hydrogen under Mitsunobu conditions, using a reagent such as triphenylphosphine/diethylazodicarboxylate and the like to obtain a compound of formula I, wherein X1, X2, X3, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above, except that R2 is not hydrogen. General Procedure (B) Step A: Converting the —OH functionality in the compound of formula IV, wherein X1, X2, X3, X4, and R1 are defined as above, to an appropriate leaving group (L) such as p-toluenesulfonate, methanesulfonate, halogen (for example by methods according to: Houben-Weyl, Methoden der organischen Chemie, Alkohole III, 6/1 b, Thieme-Verlag 1984, 4th Ed., pp. 927–939; Comprehensive Organic Transformations. A guide to functional group preparations, VCH Publishers 1989, 1st Ed., pp. 353–363 and J. Org. Chem., Vol. 36 (20), 3044–3045, 1971), triflate and the like, to give a compound of formula VI wherein, X1, X2, X3, X4, R1 and L are defined as above. Step B: Reacting the compound of formula VI wherein L is a leaving group such as p-toluenesulfonate, methanesulfonate, halogen, triflate and the like and wherein X1, X2, X3, X4 and R1 are defined as above with a compound of formula V wherein Y1, Ar, Y2, Z and R2 are defined as above, except that R2 is not hydrogen to give a compound of formula I wherein X1, X2, X3, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above, except that R2 is not hydrogen. General Procedure (C) Step A: Reacting an compound of formula VII wherein X2, X4 and R1 are defined as above, with an boronic acid derivative of X1 or X3 under appropriate coupling conditions as Pd2(dba)3/Pd(P(t-Bu)3)2/KF/THF, to give a compound of formula IV, wherein X1, X2, X3, X4 and R1 are defined as above, and Step B: Reacting a compound of formula IV as described inder procedure A step C, to obtain a compound of formula I, wherein X1, X2, X3, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above, except that R2 is not hydrogen. General Procedure (D) Step A: By chemical or enzymatic saponification of a compound of formula I wherein X1, X2, X3, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above, except that R2 is not hydrogen to give a compound of formula I wherein X1, X2, X3, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above, except that R2 is hydrogen. General Procedure (E) Step A: Reacting a compound of formula VII, wherein X2, X4 and R1 are defined as above, with a compound of formula V, wherein Y1, Ar, Y2, Z and R2 are defined as above, except that R2 is not hydrogen under Mitsunobu conditions, using a reagent such as triphenylphosphine/diethylazodicarboxylate and the like to obtain a compound of formula VIII wherein X2, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above, except that R2 is not hydrogen. Step B: By chemical or enzymatic saponification of a compound of formula VIII wherein X2, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above, except that R2 is not hydrogen, to give a compound of formula VIII wherein X2, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above, except that R2 is hydrogen. Step C: Reacting a compound of formula VIII, wherein X2, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above, with an boronic acid derivative of X1 or X3 under appropriate coupling conditions as Pd2(dba)3/Pd(P(t-Bu)3)2/KF/THF, to give a compound of formula I, wherein X1, X2, X3, X4, Y1, Y2, Ar, Z, R1 and R2 are defined as above. EXAMPLE 1 General Procedure (E) [4-(3,3-Bis-biphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid Step A: To a solution of NaH (3.53 g, 88.2 mmol) in dry toluene (300 ml) was added dropwise at 0° C. a solution of trietylphosphonoacetate (13.2 g, 58.8 mmol) in toluene (100 ml). The reaction mixture was stirred for 30 min. after which a solution of 4,4-dibromobenzophenone (10.0 g, 29.4 mmol) in THF (100 ml) was added. The reaction mixture was stirred for 48 h. Ethanol (10 ml) and water (300 ml) were added and the mixture was extracted with ethyl acetate-methanol (2%, 2×150 ml). The combined organic phases were washed with brine, dried with MgSO4, filtered and evaporated. The residue was purified by column chromatography (eluent: ether) to give ethyl 3,3-bis-(4-bromophenyl)-acrylate as a gum. Crystallization from hexanes gave white crystals in 8.77 g (73%) yield. 1H NMR (CDCl3, 300 MHz); δ 1.20 (3H, t), 4.05 (2H, q), 6.35 (1H, s), 7.0–7.1 (4H, dm), 7.40–7.52 (4H, dm). Ethyl 3,3-bis-(4-bromophenyl)-acrylate (8.75 g, 21.3 mmol) was dissolved in dry THF (35 ml). DIBAL-H (1.5 M in toluene, 43 ml, 64.0 mmol) was added at −15° C. and the reaction mixture was stirred for 30 min. A solution of ammonium chloride in water was added and the mixture was extracted with ethyl acetate (3×50 ml). The combined organic phases were washed with brine, dried with MgSO4, filtered and evaporated to give 3,3-bis-(4-bromophenyl)-pro-2-en-1-ol in 6.0 g (76%) yield. 1H NMR (CDCl3, 300 MHz); δ 1.15 (1H, br s), 4.16–4.20 (2H, dd), 6.25 (1H, t), 7.0–7.1 (4H, dm), 7.40–7.52 (4H, dm). 3,3-Bis-(4-bromophenyl)-pro-2-en-1-ol (2.98 g, 8.1 mmol) and tributylphosphine (2.4 g, 12.1 mmol) were dissolved in dry THF (150 ml) and cooled to 0° C. under an atmosphere of nitrogen. 1,1′-(Azodicarbonyl)dipiperidine (ADDP) (3.1 g, 12.1 mmol) was added and the reaction mixture was stirred for 5 min. (4-Mercapto-2-methyl-phenoxy)-acetic acid methyl ester (2.06 g, 9.7 mmol; Bioorg. Med. Chem. Lett. 2003, 13, 1517) was slowly added (5 min) and the stirring continued for 2 h at 0° C. Water (100 ml) was added and the mixture was extracted with dichloromethane (2×150 ml). The combined organic phases were dried with MgSO4, filtered and evaporated. The residue was purified by column chromatography (eluent: dichloromethane) to give 4.0 g (88%) of {4-[3,3-bis-(4-bromo-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid methyl ester. 1H NMR (CDCl3, 300 MHz); δ 2.20 (3H, s), 3.44 (2H, d), 3.78 (3H, s), 4.64 (2H, s), 6.11 (1H, t), 6.55 (1H, d), 6.73 (2H, d), 6.98 (2H, d), 7.10 (2H, bs), 7.38 (2H, d), 7.43 (2H, d). Step B: A solution of {4-[3,3-bis-(4-bromo-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid methyl ester (530 mg, 0.94 mmol) in ethanol (20 ml) and 1M NaOH (2.0 ml, 2.0 mmol) was stirred at room temp. for 2 h. The reaction mixture added water (20 ml) and 1N HCl (3.0 ml). The water phase was extracted with dichloromethane (2×50 ml) and the combined organic phases were dried with MgSO4, filtered and evaporated to give 482 mg (93%) of {4-[3,3-bis-(4-bromo-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid. 1H NMR (CDCl3, 300 MHz) δ2.20 (3H, s), 3.45 (2H, d), 4.68 (2H, s), 6.10 (1H, t), 6.58 (1H, d), 6.75 (2H, d), 6.98 (2H, d), 7.10–7.13 (2H, m), 7.38 (2H, d), 7.43 (2H, d). Step C: In an evaporated schlenk flask kept under nitrogen atmosphere were added {4-[3,3-bis-(4-bromo-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (297 mg, 0.54 mmol), phenylboronic acid (152 mg, 1.2 mmol), KF (104 mg, 1.79 mmol), Pd2(dba)3 (30 mg, 33 mmol) and Pd(P(t-Bu)3)2 (33 mg, 65 mmol). THF (6 ml) was added to the solid mixture keeping the mixture under nitrogen. The reaction mixture was stirred at room temperature for 1 h, followed by 4 h at 50° C. A saturated solution of ammonium chloride (5 ml) was added, and the mixture was extracted with dichloromethane (2×20 ml). The organic phases were dried and evaporated. The residue was purified by column chromatography (eluent: dichloromethane:THF (8:3)) to give the title compound in 155 mg (53%) yield. 1H NMR (CDCl3, 300 MHz) δ 2.22 (3H, s), 3.61 (2H, d), 4.60 (2H, s), 6.21 (1H, t), 6.59 (1H, d), 7.00–7.77 (20H, m). EXAMPLE 2 General Procedure (E) {4-[3,3-Bis-(4′-methanesulfonyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid Step C: In an evaporated schlenk flask kept under nitrogen atmosphere were added {4-[3,3-bis-(4-bromo-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (208 mg, 0.38 mmol, example 1, step A–B), 4-(methansulfonyl)phenylboronic acid (280 mg, 1.4 mmol), KF (77 mg, 1.33 mmol), Pd2(dba)3 (21 mg, 23 mmol) and Pd(P(t-BU)3)2 (23 mg, 46 mmol). THF (5 ml) was added to the solid mixture keeping the mixture under nitrogen. The reaction mixture was stirred at room temperature for 1 h, followed by 4 h at 50° C. A saturated solution of ammonium chloride (5 ml) was added, and the mixture was extracted with dichloromethane (2×20 ml). The organic phases were dried and evaporated. The residue was purified by column chromatography (eluent: ethyl acetate:methanol (9:1)) to give the title compound in 95 mg (36%) yield. 1H NMR (CDCl3, 300 MHz) δ 2.18 (3H, s), 3.10 (3H, s), 3.12 (3H, s), 3.57 (2H, d), 4.62 (2H, s), 6.26 (1H, t), 6.60 (1H, d), 7.12 (4H, m), 7.31 (2H, d), 7.53 (2H, d), 7.85 (2H, d), 7.74 (2H, d), 7.81 (2H, d), 8.00 (4H, m). EXAMPLE 3 General Procedure (E) {4-[3,3-Bis-(3′-trifluoromethyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid Step C: In an evaporated schlenk flask kept under nitrogen atmosphere were added {4-[3,3-bis-(4-bromo-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (231 mg, 0.42 mmol, example 1, step A–B), 3-(trifluoromethyl)phenylboronic acid (203 mg, 1.1 mmol), KF (81 mg, 1.39 mmol), Pd2(dba)3 (23 mg, 25 mmol) and Pd(P(t-Bu)3)2 (26 mg, 51 mmol). THF (5 ml) was added to the solid mixture keeping the mixture under nitrogen. The reaction mixture was stirred at room temperature for 1 h, followed by 10 h at 50° C. A saturated solution of ammonium chloride (5 ml) was added, and the mixture was extracted with dichloromethane (2×20 ml). The organic phases were dried and evaporated. The residue was purified by HPLC to give the title compound in 140 mg (49%) yield. 1H NMR (CDCl3, 300 MHz) δ 2.22 (3H, s), 3.59 (2H, d), 4.66 (2H, s), 6.24 (1H, t), 6.61 (1H, d), 7.08–7.90 (18H, m). EXAMPLE 4 General Procedure (E) [4-(3,3-Bis-[1,1′;4′,1″]terphenyl-4-yl-allylsulfanyl)-2-methyl-phenoxy]-acetic acid Step C: In an evaporated schlenk flask kept under nitrogen atmosphere were added {4-[3,3-bis-(4-bromo-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (225 mg, 0.41 mmol, example 1, step A–B, biphenylboronic acid (163 mg, 0.8 mmol), KF (79 mg, 1.35 mmol), Pd2(dba)3 (23 mg, 25 mmol) and Pd(P(t-Bu)3)2 (26 mg, 51 mmol). THF (6 ml) was added to the solid mixture keeping the mixture under nitrogen. The reaction mixture was stirred at 50° C. for 2 h, followed by 10 h at 70° C. A saturated solution of ammonium chloride (5 ml) was added, and the mixture was extracted with dichloromethane (2×20 ml). The organic phases were dried and evaporated. The residue was purified by HPLC to give the title compound in 55 mg (19%) yield. 1H NMR (DMSO, 300 MHz) δ 2.12 (3H, s), 3.60 (2H, d), 4.68 (2H, s), 6.24 (1H, t), 6.77 (1H, d), 7.02–7.90 (28H, m). EXAMPLE 5 General Procedure (E) {4-[3,3-Bis-(3′-dimethylcarbamoyl-biphenyl-4-yl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid Step C: In an evaporated schlenk flask kept under nitrogen atmosphere were added {4-[3,3-bis-(4-bromo-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (225 mg, 0.41 mmol, example 1, step A–B), N,N-dimethylbenzamide-3-boronic acid (182 mg, 0.94 mmol), KF (79 mg, 1.35 mmol), Pd2(dba)3 (23 mg, 25 mmol) and Pd(P(t-Bu)3)2 (26 mg, 51 mmol). THF (6 ml) was added to the solid mixture keeping the mixture under nitrogen. The reaction mixture was stirred at room temperature for 1 h, followed by 1 h at 50° C. A saturated solution of ammonium chloride (5 ml) was added, and the mixture was extracted with dichloromethane (3×20 ml). The organic phases were dried and evaporated. The residue was purified by column chromatography (eluent: dichloromethane:methanol (9:1)) to give the title compound in 90 mg (32%) yield. 1H NMR (CDCl3, 300 MHz) δ 2.19 (3H, s), 2.99 (3H, s), 3.07 (3H, s), 3.14 (3H, s), 3.17 (3H,s), 3.48 (2H, d), 4.57 (2H, s), 6.25 (1H, t), 6.64 (1H, d), 6.74 (2H, d), 6.93 (1H, d), 7.17–8.00 (15H, m). EXAMPLE 6 General Procedure (E) {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid Step C: In a vial for micro wave oven was added under nitrogen {4-[3,3-bis-(4-bromophenyl)-allylsulfanyl]-2-methyl-phenoxy}-acetic acid (175 mg, 0.34 mmol, example 1, step A–B), pyridine-3-boronic acid (91 mg, 0.74 mmol), sodium carbonate (215 mg, 2.0 mmol), Pd(PPh3)2Cl (15 mg, 0.02 mmol), dimethyl ether (2.9 ml), water (1.25 ml) and ethanol (99%, 0.8 ml). The reaction was run for 5 min at 140° C. in the micro wave oven. The reaction was filtered and the filtrate evaporated. The residue was purified by column chromatography (eluent: dichloromethane:methanol (4:1)) to give the title compound in 102 mg (56%) yield. 1H NMR (MeOD, 400 MHz) δ 2.17 (3H, s), 3.55 (2H, d), 4.49 (2H, s), 6.36 (1H, t), 6.68 (1H, d), 7.00–7–65 (12H, m), 8.03–8.14 (2H, m), 8.51 (2H, m), 8.77 (1H, s), 8.85 (1H, s). EXAMPLE 7 {4-[3,3-Bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-2-trifluoromethyl-phenoxy}-acetic acid Step A: To a 0° C. solution of triethylphosphonoacetate (11.7 g, 52.4 mmol) in THF (230 ml) was added over 5 min a solution of NaH 60% in oil (2.6 g; 109 mmol). The reaction mixture was stirred for 30 min after which 4,4-diiodobenzophenone (18.8 g, 42.4 mmol; Bull. Chem. Soc. Jpn. 1999, 72, 115–120) was added over 10 min. The reaction mixture was stirred over night at room temperature. Water (5 ml) was added followed by decalite. The mixture was evaporated and the solid residue was extracted with dichloromethane (3×200 ml). The combined organic phases were evaporated to give crude product in 17.9 g (80%) yield. Purification by column chromatography (eluent: dichloromethane) gave ethyl 3,3-bis-(4-iodophenyl)-acrylate as an oil in 9.6 g (43%) yield. 1H NMR (CDCl3, 300 MHz); δ 1.15 (3H, t), 4.07 (2H, q), 6.35 (1H, s), 6.90–7.02 (4H, m), 7.63–7.75 (4H, m). Step B: To a solution of ethyl 3,3-bis-(4-iodophenyl)-acrylate (706 mg, 1.4 mmol) in THF (1.5 ml) was added over 45 min a solution of DIBAL-H (1.5 M in toluene, 6.3 ml, 9.5 mmol) at −20° C. The reaction mixture was stirred for a further 1 h. A solution of ammonium chloride was and to the mixture was added ethyl acetate (40 ml) and decalite. The mixture was filtered and the filter washed with ethyl acetate (100 ml). The combined filtrates were evaporated and the residue was purified by column chromatography (eluent: dichloromethane:THF (8:3)) to give 3,3-bis-(4-iodophenyl)-pro-2-en-1-ol in 541 mg (84%) yield. 1H NMR (CDCl3, 300 MHz); δ 1.60 (1H, br s), 4.20 (2H, d), 6.23 (1H, t), 6.84–7.00 (4H, m), 7.57–7.75 (4H, m). Step C: In a vial for micro wave oven was added under nitrogen 3,3-bis-(4-iodophenyl)-pro-2-en-1-ol (326 mg, 0.7 mmol), pyridine-3-boronic acid (199 mg, 1.6 mmol), sodium carbonate (449 mg, 4.2 mmol), Pd(PPh3)2Cl (32 mg, 0.04 mmol), dimethyl ether (4.2 ml), water (1.8 ml) and ethanol (99%, 1.2 ml). The reaction was run for 7 min at 140° C. in the micro wave oven. The reaction was filtered and the filtrate evaporated. The residue was purified by column chromatography (eluent: dichloromethane:THF (1:1)) to give 3,3-bis-(4-pyridin-3-yl-phenyl)-prop-2-en-1-ol in 205 mg (72%) yield. 1H NMR (MeOD, 400 MHz) δ 4.18 (2H, d), 6.36 (1H, t), 7.32 (2H, d), 7.38 (2H, d), 7.44–7.54 (2H, m), 7.59 (2H, d), 7.69 (2H, d), 8.02–8.14 (2H, m), 8.51 (2H, m), 8.75–8.85 (2H, m). Triphenylphosphine (138 mg, 0.68 mmol) and ADDP (173 mg, 0.68 mmol) were added to a solution of 3,3-bis-(4-pyridin-3-yl-phenyl)-prop-2-en-1-ol (100 mg, 0.27 mmol) in THF (5 ml) at 0° C. The reaction mixture was stirred for 5 min under nitrogen atmosphere. (4-Mercapto-2-trifluoromethyl-phenoxy)-acetic acid ethyl ester (prepared analogous to procedure in BioOrg. Med. Chem. Lett. 2003, 13, 1517) (92 mg, 0.32 mmol) was slowly added to the reaction mixture and the reaction was stirred for further 2 h at 0° C. The reaction mixture was evaporated and the residue was triturated with ether (2×15 ml). The filtered ether phases were evaporated and purified by column chromatography (eluent: ethyl acetate) to give {4-[3,3-bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-2-trifluoromethyl-phenoxy}-acetic acid ethyl ester in 100 mg (60%) yield. 1H NMR (CDCl3, 400 MHz) δ 1.24 (3H, t), 3.61 (2H, d), 4.22 (2H, q), 4.70 (2H, s), 6.23 (1H, t), 6.77 (1H, d), 7.05 (2H, d), 7.25–7.62 (10H, m), 7.74–7.95 (2H, m), 8.60 (2H, m), 8.82–8.94 (2H, m). A solution of {4-[3,3-bis-(4-pyridin-3-yl-phenyl)-allylsulfanyl]-2-trifluoromethyl-phenoxy}-acetic acid ethyl ester (70 mg, 0.1 mmol) in ethanol (10 ml) and 1N NaOH (0.5 ml) was stirred at 75° C. for 1 h. The reaction mixture was evaporated and the residue was treated with 1N HCl (0.7 ml) and extracted with methylene chloride (2×25 ml). The organic phases were dried and evaporated to give the title compound in 23 mg (34%) yield. 1H NMR (CDCl3, 400 MHz) δ 3.45 (2H, d), 4.89 (2H, s), 6.27 (1H, t), 6.80 (3H, m), 7.27 (2H, d), 7.38–7.75 (8H, m), 8.06 (1H, m), 8.33 (1H, m), 8.58 (2H, m), 8.85 (1H, s), 9.27 (1H, s). Pharmacological Methods In Vitro PPARalpha, PPARgamma and PPARdelta Activation Activity The PPAR transient transactivation assays are based on transient transfection into human HEK293 cells of two plasmids encoding a chimeric test protein and a reporter protein respectively. The chimeric test protein is a fusion of the DNA binding domain (DBD) from the yeast GAL4 transcription factor to the ligand binding domain (LBD) of the human PPAR proteins. The PPAR-LBD moiety harbored in addition to the ligand binding pocket also the native activation domain (activating function 2=AF2) allowing the fusion protein to function as a PPAR ligand dependent transcription factor. The GAL4 DBD will direct the chimeric protein to bind only to Gal4 enhancers (of which none existed in HEK293 cells). The reporter plasmid contained a Gal4 enhancer driving the expression of the firefly luciferase protein. After transfection, HEK293 cells expressed the GAL4-DBD-PPAR-LBD fusion protein. The fusion protein will in turn bind to the Gal4 enhancer controlling the luciferase expression, and do nothing in the absence of ligand. Upon addition to the cells of a PPAR ligand luciferase protein will be produced in amounts corresponding to the activation of the PPAR protein. The amount of luciferase protein is measured by light emission after addition of the appropriate substrate. Cell Culture and Transfection HEK293 cells were grown in DMEM +10% FCS. Cells were seeded in 96-well plates the day before transfection to give a confluency of 50–80% at transfection. A total of 0.8 μg DNA containing 0.64 μg pM1α/γLBD, 0.1 μg pCMVβGal, 0.08 μg pGL2(Gal4)5 and 0.02 μg pADVANTAGE was transfected per well using FuGene transfection reagent according to the manufacturers instructions (Roche). Cells were allowed to express protein for 48 h followed by addition of compound. Plasmids: Human PPAR α, γ and δ was obtained by PCR amplification using cDNA synthesized by reverse transcription of mRNA from human liver, adipose tissue and plancenta respectively. Amplified cDNAs were cloned into pCR2.1 and sequenced. The ligand binding domain (LBD) of each PPAR isoform was generated by PCR (PPARα: aa 167—C-terminus; PPARγ: aa 165—C-terminus; PPARδ: aa 128—C-terminus) and fused to the DNA binding domain (DBD) of the yeast transcription factor GAL4 by subcloning fragments in frame into the vector pM1 (Sadowski et al. (1992), Gene 118, 137) generating the plasmids pM1αLBD, pM1γLBD and pM1δ. Ensuing fusions were verified by sequencing. The reporter was constructed by inserting an oligonucleotide encoding five repeats of the GAL4 recognition sequence (5×CGGAGTACTGTCCTCCG(AG)) (Webster et al. (1988), Nucleic Acids Res. 16, 8192) into the vector pGL2 promotor (Promega) generating the plasmid pGL2(GAL4)5. pCMVβGal was purchased from Clontech and pADVANTAGE was purchased from Promega. In Vitro Transactivation Assay Compounds: All compounds were dissolved in DMSO and diluted 1:1000 upon addition to the cells. Compounds were tested in quadruple in concentrations ranging from 0.001 to 300 μM. Cells were treated with compound for 24 h followed by luciferase assay. Each compound was tested in at least two separate experiments. Luciferase assay: Medium including test compound was aspirated and 100 μl PBS incl. 1 mM Mg++ and Ca++ was added to each well. The luciferase assay was performed using the LucLite kit according to the manufacturers instructions (Packard Instruments). Light emission was quantified by counting on a Packard LumiCounter. To measure β-galactosidase activity 25 μl supernatant from each transfection lysate was transferred to a new microplate. β-galactosidase assays were performed in the microwell plates using a kit from Promega and read in a Labsystems Ascent Multiscan reader. The β-galactosidase data were used to normalize (transfection efficiency, cell growth etc.) the luciferase data. Statistical Methods The activity of a compound is calculated as fold induction compared to an untreated sample. For each compound the efficacy (maximal activity) is given as a relative activity compared to Wy14,643 for PPARα, Rosiglitazone for PPARγ and Carbacyclin for PPARδ. The EC50 is the concentration giving 50% of maximal observed activity. EC50 values were calculated via non-linear regression using GraphPad PRISM 3.02 (GraphPad Software, San Diego, Calif.). The results were expressed as means ±SD. 10654699 novo novdisk a/s USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open 514/571 Mar 31st, 2022 02:17PM Mar 31st, 2022 02:17PM Novo Nordisk Health Care Pharmaceuticals & Biotechnology
nyse:nvo Novo Nordisk Feb 22nd, 2022 12:00AM Oct 9th, 2019 12:00AM https://www.uspto.gov?id=US11253596-20220222 Phenylalkylcarboxylic acid delivery agents The present invention provides phenylalkylcarboxylic acid compounds and compositions containing such compounds which facilitate the delivery of biologically active agents. 11253596 1. A composition comprising: (A) at least one biologically active agent; and (B) at least one delivery agent compound represented by formula I or a pharmaceutically acceptable salt thereof, wherein n is 1-12, and R1-R5 are independently hydrogen, C1-C6, alkyl, C2-C4 alkenyl, halogen, C1-C4 alkoxy, hydroxyl, C6-C14 aryloxy, or C1-C6 alkylhalo group, with the proviso that at least one of R1-R5 is methyl, C1-C4 alkoxy, hydroxyl or halogen. 2. The composition of claim 1, wherein the delivery agent is selected from 4-(4-methoxyphenyl)butanoic acid, 5-(2-methoxyphenyl)pentanoic acid, 5-(3-fluorophenyl)pentanoic acid, 5-(3-methoxyphenyl)pentanoic acid, 6-(3-fluorophenyl)hexanoic acid, 3-(4-n-propoxyphenyl)propanoic acid, 3-(4-isopropoxyphenyl)propanoic acid, 3-(4-n-butoxyphenyl)propanoic acid, 3-(3-phenoxyphenyl)propanoic acid, 3-(3-ethoxyphenyl)propanoic acid, 3-(3-isopropoxyphenyl)propanoic acid, 3-(3-n-butoxyphenyl)propanoic acid, 3-(3-n-propoxyphenyl)propanoic acid, 3-(3-isobutoxyphenyl)propanoic acid, 3-(4-isobutoxyphenyl)propanoic acid, or pharmaceutically acceptable salts thereof. 3. The composition of claim 1, wherein the delivery agent is conjugated to a polymer by a linkage group selected from the group consisting of —NHC(O)NH—, —C(O)NH—, —NHC(O)—; —OOC—, —COO—, —NHC(O)O—, —OC(O)NH—, —CH2NH—NHCH2—, —CH2NHC(O)O—, —OC(O)NHCH2—, —CH2NHCOCH2O—, —OCH2C(O)NHCH2—, —NHC(O)CH2O—, —OCH2C(O)NH—, —NH—, —O—, and carbon-carbon bond. 4. The composition of claim 1, wherein the biologically active agent is a protein, polypeptide, peptide, hormone, polysaccharide, mucopolysaccharide, carbohydrate, lipid, or combination thereof. 5. The composition of claim 1, wherein the biologically active agent is selected from the group consisting of: argatroban, BIBN-4096BS, growth hormones, human growth hormones recombinant human growth hormones (rhGH), bovine growth hormones, porcine growth hormones, growth hormone releasing hormones, growth hormone releasing factor, glucagon, interferons, α-interferon, β-interferon, γ-interferon, interleukin-1, interleukin-2, insulin, porcine insulin, bovine insulin, human insulin, human recombinant insulin, insulin-like growth factor (IGF), IGF-1, heparin, unfractionated heparin, heparinoids, dermatans, chondroitins, low molecular weight heparin, very low molecular weight heparin, ultra low molecular weight heparin, calcitonin, salmon calcitonin, eel calcitonin, human calcitonin; erythropoietin (EPO), atrial naturetic factor, antigens, monoclonal antibodies, somatostatin, protease inhibitors, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, leutinizing-hormone-releasing-hormone, follicle stimulating hormone, glucocerebrosidase, thrombopoeitin, filgrastim, postaglandins, cyclosporin, vasopressin, cromolyn sodium, sodium chromoglycate, disodium chromoglycate, vancomycin, desferrioxamine (DFO), parathyroid hormone (PTH), fragments of PTH, glucagon-like peptide 1 (GLP-1), antimicrobials, anti-fungal agents, vitamins; analogs, fragments, mimetics and polyethylene glycol (PEG)-modified derivatives of these compounds; gallium or gallium salts; glucagons; zanamivir, sumatriptan, almotriptan, naratriptan, rizatriptan, frovatriptan, eletriptan, caspofungin acetate, CPHPC, siRNA and any combination thereof. 6. The composition of claim 1, further comprising at least one enzyme inhibitor. 7. The composition of claim 1, wherein the weight ratio of delivery agent to active agent ranges from about 800:1 to about 10:1. 8. A method of preparing pharmaceutical composition, comprising mixing at least one delivery agent compound of claim 1, and a biologically active agent. 9. A dosage unit form comprising: (A) a delivery agent according to claim 1; and (B) a biologically active agent; and (C) (a) an excipient, (b) a diluent, (c) a disintegrant, (d) a lubricant, (e) a plasticizer, (f) a colorant, (g) an enzyme inhibitor (h) a dosing vehicle, or (i) any combination thereof. 10. The dosage unit form of claim 9, wherein the biologically active agent comprises at least one protein, polypeptide, peptide, hormone, polysaccharide, mucopolysaccharide, carbohydrate, or lipid. 11. The dosage unit form of claim 10, wherein the biologically active agent is selected from the group consisting of: argatroban, BIBN-4096BS, growth hormones, human growth hormones (hGH), recombinant human growth hormones (rhGH), bovine growth hormones, porcine growth hormones, growth hormone releasing hormones, growth hormone releasing factor, interferons, glucagon, α-interferon, β-interferon, γ-interferon, interleukin-1, interleukin-2, insulin, porcine insulin, bovine insulin, human insulin, human recombinant insulin, insulin-like growth factor, insulin-like growth factor-1, heparin, unfractionated heparin, heparinoids, dermatans, chondroitins, low molecular weight heparin, very low molecular weight heparin, ultra low molecular weight heparin, calcitonin, salmon calcitonin, eel calcitonin, human calcitonin; erythropoietin, atrial naturetic factor, antigens, monoclonal antibodies, somatostatin, protease inhibitors, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, leutinizing-hormone-releasing-hormone, follicle stimulating hormone, glucocerebrosidase, thrombopoeitin, filgrastim, postaglandins, cyclosporin, vasopressin, cromolyn sodium, sodium chromoglycate, disodium chromoglycate, vancomycin, desferrioxamine, parathyroid hormone, fragments of PTH, glucagon-like peptide 1 (GLP-1), antimicrobials, anti-fungal agents, vitamins; analogs, fragments, mimetics and polyethylene glycol-modified derivatives of these compounds; gallium or gallium salts; glucagons, zanamivir, sumatriptan, almotriptan, naratriptan, rizatriptan, frovatriptan, eletriptan, capsofungin acetate, CPHPC, SiRNA and any combination thereof. 12. A method for administering a biologically-active agent to an animal in need of the agent, the method comprising administering orally to the animal the composition of claim 1. 13. A method for administering a biologically-active agent to an animal in need of the agent, the method comprising administering orally to the animal the dosage unit form of claim 9. 14. A method for administering a biologically-active agent to an animal in need of the agent, the method comprising administering to the animal the composition of claim 1 intranasally, sublingually, intraduodenally, subcutaneously, buccally, intracolonicly, rectally, vaginally, mucosally, pulmonary, transdermally, intradermally, parenterally, intravenously, intramuscularly, via the ocular system, or by traversing the blood-brain barrier. 15. A method for administering a biologically-active agent to an animal in need of the agent, the method comprising administering to the animal the dosage unit of claim 9 intranasally, sublingually, intraduodenally, subcutaneously, buccally, intracolonicly, rectally, vaginally, mucosally, pulmonary, transdermally, intradermally, parenterally, intravenously, intramuscularly, via the ocular system, or by traversing the blood-brain barrier. 16. A method for administering a biologically active agent comprising administering a delivery agent compound of claim 1, followed by administration of a biologically active agent. 17. The method of claim 16, wherein the biologically active agent is a protein, polypeptide, peptide, hormone, polysaccharide, mucopolysaccharide, carbohydrate, or lipid. 18. A method for increasing the bioavailability of a biologically active agent comprising administering to an animal a composition of claim 1. 19. A method for preparing a composition comprising mixing: (A) at least one active agent; (B) at least one delivery agent compound of claim 1; (C) optionally, an enzyme inhibitor; and (D) optionally, a dosing vehicle. 20. A compound selected from the group consisting of 4-(4-methoxyphenyl)butanoic acid, 5-(2-methoxyphenyl)pentanoic acid, 5-(3-fluorophenyl)pentanoic acid, 5-(3-methoxyphenyl)pentanoic acid, 6-(3-fluorophenyl)hexanoic acid, 3-(4-t-butylphenyl) propanoic acid, 3-(4-n-butylphenyl)propanoic acid, 3-(4-n-propylphenyl)propanoic acid, 3-(4-n-propoxyphenyl)propanoic acid, 3-(4-isopropoxyphenyl)propanoic acid, 3-(4-n-butoxyphenyl)propanoic acid, 3-(3-phenoxyphenyl)propanoic acid, 3-(3-ethoxyphenyl)propanoic acid, 3-(3-isopropoxyphenyl)propanoic acid, 3-(3-n-butoxyphenyl)propanoic acid, 3-(3-n-propoxyphenyl)propanoic acid, 3-(3-isobutoxyphenyl)propanoic acid, 3-(4-isobutoxyphenyl)propanoic acid, 4-(4-ethylphenyl)butanoic acid, 4-(4-isopropylphenyl)butanoic acid, and 5-(4-ethylphenyl)pentanoic acid, or pharmaceutically acceptable salts thereof. 20 This application is a continuation of U.S. patent application Ser. No. 15/198,392, filed Jun. 30, 2016, which is a continuation of U.S. patent application Ser. No. 13/474,491, filed May 17, 2012, now abandoned, which is a continuation of U.S. patent application Ser. No. 12/522,464, filed Oct. 14, 2009, now abandoned, which is the U.S. national phase of International Application No. PCT/US08/53429, filed Feb. 8, 2008, which claims the benefit of U.S. Provisional Application No. 60/888,927, filed Feb. 8, 2007. FIELD OF THE INVENTION The present invention relates phenylalkylcarboxylic acid compounds and compositions which facilitate the delivery of active agents. BACKGROUND OF THE INVENTION Conventional means for delivering active agents are often severely limited by biological, chemical and physical barriers. Typically, these barriers are imposed by the environment through which delivery occurs, the environment of the target for delivery, and/or the target itself. Biologically and chemically active agents are particularly vulnerable to such barriers. In the delivery to animals of biologically active and chemically active pharmacological and therapeutic agents, barriers are imposed by the body. Examples of physical barriers are the skin, lipid bi-layers and various organ membranes that are relatively impermeable to certain active agents but must be traversed before reaching a target, such as the circulatory system. Chemical barriers include, but are not limited to, pH variations in the gastrointestinal (GI) tract and degrading enzymes. These barriers are of particular significance in the design of oral delivery systems. Oral delivery of many biologically or chemically active agents would be the route of choice for administration to animals if not for biological, chemical, and physical barriers. Among the numerous agents which are not typically amenable to oral administration are biologically or chemically active peptides, such as calcitonin and insulin; polysaccharides, and in particular mucopolysaccharides including, but not limited to, heparin; heparinoids; antibiotics; and other organic substances. These agents may be rapidly rendered ineffective or destroyed in the gastro-intestinal tract by acid hydrolysis, enzymes, and the like. In addition, the size and structure of macromolecular drugs may prohibit absorption. Earlier methods for orally administering vulnerable pharmacological agents have relied on the co-administration of adjuvants (e.g., resorcinols and non-ionic surfactants such as polyoxyethylene oleyl ether and n-hexadecylpolyethylene ether) to increase artificially the permeability of the intestinal walls, as well as the co-administration of enzymatic inhibitors (e.g., pancreatic trypsin inhibitors, diisopropylfluorophosphate (DFF) and trasylol) to inhibit enzymatic degradation. Liposomes have also been described as drug delivery systems for insulin and heparin. However, broad spectrum use of such drug delivery systems is precluded because: (1) the systems require toxic amounts of adjuvants or inhibitors; (2) suitable low molecular weight cargos, i.e. active agents, are not available; (3) the systems exhibit poor stability and inadequate shelf life; (4) the systems are difficult to manufacture; (5) the systems fail to protect the active agent (cargo); (6) the systems adversely alter the active agent; or (7) the systems fail to allow or promote absorption of the active agent. Proteinoid microspheres have been used to deliver pharmaceuticals. See, for example, U.S. Pat. Nos. 5,401,516; 5,443,841; and Re. 35,862. In addition, certain modified amino acids have been used to deliver pharmaceuticals. See, for example, U.S. Pat. Nos. 5,629,020; 5,643,957; 5,766,633; 5,776,888; and 5,866,536. More recently, a polymer has been conjugated to a modified amino acid or a derivative thereof via a linkage group to provide for polymeric delivery agents. The modified polymer may be any polymer, but preferred polymers include, but are not limited to, polyethylene glycol (PEG), and derivatives thereof. See, for example, International Patent Publication No. WO 00/40203. However, there is still a need for simple, inexpensive delivery systems which are easily prepared and which can deliver a broad range of active agents by various routes. SUMMARY OF THE INVENTION The present invention provides phenylalkylcarboxylic acid compounds and compositions which facilitate the delivery of active agents (e.g. biologically active agents). Delivery agent compounds of the present invention include those having the formula: and pharmaceutically acceptable salts thereof, wherein n is 1-12, and R1-R5 are independently hydrogen, C1-C6 alkyl, C2-C4 alkenyl, halogen, C1-C4 alkyloxy, hydroxyl, C6-C14 aryloxy, or C1-C6 alkylhalo (e.g. C1 alkylhalo) group. According to one embodiment, n ranges from 1 to 9. For example, n may be 1-9, 2-9, 3-9, 4-9, 5-9, 6-9, 7-9, 8-9, 1-8, 2-8, 3-8, 4-8, 5-8, 6-8, 7-8, 1-7, 2-7, 3-7, 4-7, 5-7, 6-7, 1-6, 2-6, 3-6, 4-6, 5-6, 1-5, 2-5, 3-5, 4-5, 1-4, 2-4, 3-4, 1-3, 2-3 or 1-2. According to another embodiment, at least one of R1 to R5 is methyl, methoxy, hydroxy or halogen group (e.g., Cl or F). Mixtures of these delivery agent compounds may also be used. The invention also provides a pharmaceutical composition comprising at least one delivery agent compound of the present invention, and at least one active agent (e.g. a biologically active agent). When administered with an active agent, delivery agents of the present application improve the bioavailability of the active agent compared to administration of the active agent without the delivery agent compound. Also provided is a dosage unit form comprising a pharmaceutical composition of the present invention. The dosage unit form may be in the form of a liquid or a solid, such as a tablet, capsule or particle, including a powder or sachet. Another embodiment is a method for administering an active agent to an animal, particularly an animal in need of the active agent, by administering a pharmaceutical composition comprising at least one of delivery agent compound of the present invention and the active agent to the animal. Preferred routes of administration include the oral and intracolonic routes, particularly the oral route. Yet another embodiment of the present invention is a method of treating a disease or for achieving a desired physiological effect in an animal (e.g. a human) by administering to the animal the pharmaceutical composition of the present invention. Yet another embodiment of the present invention is a method of preparing a pharmaceutical composition of the present invention by mixing at least one delivery agent compound of the present invention, and at least one active agent. Yet another embodiment of the present invention is a method of increasing the bioavailability (e.g., the oral bioavailability) of a pharmaceutical composition containing an active agent (e.g., abiologically active agent) comprising adding a delivery agent compound of the present invention to the pharmaceutical composition. DETAILED DESCRIPTION OF THE INVENTION Definitions The term “alkyl” refers to a straight-chained, branched, or substituted monovalent aliphatic hydrocarbon group containing no double or triple carbon-carbon bonds. Examples of alkyl group include, but are not limited to, methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl, and 1-dimethylethyl (t-butyl). The term “alkenyl” refers to a straight-chained, branched, or substituted monovalent aliphatic hydrocarbon group containing at least one carbon-carbon double bond. Examples of alkenyl groups include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl (allyl), iso-propenyl, 2-methyl-1-propenyl, 1-butenyl, and 2-butenyl. The term “alkylene” refers to a straight-chained, branched or substituted divalent aliphatic hydrocarbon group containing no double or triple bonds. The term “alkyloxy” refers to an alkyl group attached via an oxygen linkage to the rest of the molecule. Examples of alkyloxy groups include, but are not limited to, —OCH3, and —OC2H5 groups. The term “aryl” refers to an monovalent C6-C14 aromatic group, i.e. a monovalent group having one or more unsaturated carbon rings. Examples of aryl groups, include, but are not limited to, phenyl, naphthyl, tetrahydronapthyl, indanyl, and biphenyl. The term “alkyl(arylene)” refers to a divalent group containing an aromatic group with an alkyl group before and/or after the aromatic group. The term “aryloxy” refers to an C6-C14 aryl group attached via an oxygen linkage to the rest of the molecule, such as —OC6H5. The term “insulin” includes recombinant forms of insulin (e.g. recombinant human insulin), analogs of insulin lispro or Humalog®) as well as regular forms of insulin of human or other animal origin. The term “heparin” includes unfractionated heparin, low molecular weight heparin, very low molecular weight heparin, of recombinant, human, or other animal origin. The term “LHRH” or “luteinizing hormone-releasing hormone” refers to a hormone produced by the hypothalamus that signals the anterior pituitary gland to begin secreting luteinizing hormone and follicle-stimulating hormone. The term “rhGH” refers to recombinant human growth hormone. The term “caspofungin” or “caspofungin acetate” refers to a water-soluble, semisynthetic lipopeptide derived from the fungus, Glarea lozoyensis, that has activity against Aspergillus and Candida species. Caspofugin acetate (Cancidas®) has been approved by the FDA and is indicated for the treatment of invasive aspergillosis in patients who are refractory to or intolerant of other antifungal agents. Unless otherwise specified, the term “substituted” as used herein refers to substitution with any one or any combination of the following substituents: hydroxy, C1-C6 alkyl, including methyl, ethyl, propyl, isopropyl, normal or iso-butyl; C2-C4 alkenyl, C1-C4 alkyloxy, aryl, halo, alkylhalo, or aryloxy groups. The term “about” means generally means within 10%, preferably within 5%, and more preferably within 1% of a given range. The term “short stature” refers to a subject with a size (e.g. a height) that is significantly below what is considered normal. Growth hormone, e.g., human growth hormone, is indicated for short stature. Delivery Agent Compounds Delivery agent compounds of the present invention include those compounds represented by Formula I below, and pharmaceutically acceptable salts thereof: wherein n is 1-12; and R1-R5 are independently hydrogen, C1-C6 alkyl, C2-C4 alkenyl, halo, C1-C4 alkyloxy, hydroxyl, C6-C14 aryloxy, or C1-C6 alkylhalo group (e.g. C1 alkylhalo). In various embodiments, n may be 1-9, 2-9, 3-9, 4-9, 5-9, 6-9, 7-9, 8-9, 1-8, 2-8, 3-8, 4-8, 5-8, 6-8, 7-8, 1-7, 2-7, 3-7, 4-7, 5-7, 6-7, 1-6, 2-6, 3-6, 4-6, 5-6, 1- 5, 2-5, 3-5, 4-5, 1-4, 2-4, 3-4, 1-3, 2-3 or 1-2. In another embodiment of the present invention, delivery agent compounds of the present invention include those compounds represented by Formula I above in which at least one of R1-R5 is a methyl, methoxy, alkyloxy, hydroxy or halogen group. In a preferred embodiment, delivery agent compounds include those in which n is defined as in the preceding paragraph and at least one of R1-R5 is a methyl, methoxy, alkyloxy, hydroxy, or halogen group. In one embodiment of the present invention, delivery agent compounds are selected from Formula I above, in which at least one of R1-R5 is a methyl group. In another embodiment, delivery agent compounds are selected from Formula I above in which at least one of R1-R5 is a methoxy group. In another embodiment, delivery agent compounds are selected from Formula I above in which at least one of R1-R5 is a hydroxy group. In another embodiment, delivery agent compounds are selected from Formula I above in which at least one of R1-R5 is an aryloxy group. In another embodiment, delivery agent compounds are selected from Formula I above in which at least one of R1-R5 is an alkyloxy group. In another embodiment, delivery agent compounds are selected from Formula I above in which at least one of R1-R5 is a C1 alkylhalo group. In another embodiment, delivery agent compounds are selected from Formula I above in which at least one of R1-R5 is a halogen, preferably at least one of R1-R5 is a chlorine atom or at least one of R1-R5 is a fluorine atom. In one embodiment of the present invention, the compounds listed in Table 1 are excluded as delivery agents of Formula I. However, in various embodiments these compounds may be included in compositions that further include an active agent (e.g., a biologically active agent). The delivery agent compounds may be in the form of the free base or pharmaceutically acceptable salts thereof, such as pharmaceutically acceptable acid addition salts. Suitable salts include, but are not limited to, organic and inorganic salts, for example ammonium, acetate salt, citrate salt, halide (preferably hydrochloride), hydroxide, sulfate, nitrate, phosphate, alkyloxy, perchlorate, tetrafluoroborate, carboxylate, mesylate, fumerate, malonate, succinate, tartrate, acetate, gluconate, and maleate. Preferred salts include, but are not limited to, citrate and mesylate salts. The salts may also be solvates, including ethanol solvates, and hydrates. Salts of the delivery agent compounds of the present invention may be prepared by methods known in the art. For example, citrate salts and mesylate salts may be prepared in ethanol, toluene and citric acid. The delivery agent compound may be purified by recrystallization or by fractionation on one or more solid chromatographic supports, alone or linked in tandem. Suitable recrystallization solvent systems include, but are not limited to, ethanol, water, heptane, ethyl acetate, acetonitrile, acetone, methanol, and tetrahydrofuran (THF) and mixtures thereof. Fractionation may be performed on a suitable chromatographic support such as alumina, using methanol/n-propanol mixtures as the mobile phase; reverse phase chromatography using trifluoroacetic acid/acetonitrile mixtures as the mobile phase; and ion exchange chromatography using water or an appropriate buffer as the mobile phase. When anion exchange chromatography is performed, preferably a 0-500 mM sodium chloride gradient is employed. The delivery agent may contain a polymer conjugated to it by a linkage group selected from the group consisting of —NHC(O)NH—, —C(O)NH—, —NHC(O)—; —OOC—, —COO—, —NHC(O)O—, —OC(O)NH—, —CH2NH—NHCH2—, —CH2NHC(O)O—, —OC(O)NHCH2—, —CH2NHCOCH2O—, —OCH2C(O)NHCH2—, —NHC(O)CH2O—, —OCH2C(O)NH—, —NH—, —O—, and carbon-carbon bond, with the proviso that the polymeric delivery agent is not a polypeptide or polyamino acid. The polymer may be any polymer including, but not limited to, alternating copolymers, block copolymers and random copolymers, which are safe for use in mammals. Preferred polymers include, but are not limited to, polyethylene; polyacrylates; polymethacrylates; poly(oxyethylene); poly(propylene); polypropylene glycol; polyethyleneglycol (PEG); and derivatives thereof and combinations thereof. The molecular weight of the polymer typically ranges from about 100 to about 200,000 daltons. The molecular weight of the polymer preferably ranges from about 200 to about 10,000 daltons. In one embodiment of the present invention, the molecular weight of the polymer ranges from about 200 to about 600 daltons and more preferably ranges from about 300 to about 550 daltons. Non-limiting examples of delivery agent compounds of Formula I include those shown below and pharmaceutically acceptable salts thereof: Compounds 22-74 (Table 1) were purchased from commercially available sources for utilization as delivery agents. TABLE 1 Commercial compounds utilized as delivery agents Delivery Agent Compound # Purchased from Chemical name 22 Sigma-Aldrich Benzeneacetic acid (St. Louis, MO) 23 Johnson Matthey 8-Phenyloctanoic acid (London, UK) 24 Lancaster 10-Phenyldecoic acid (Windham, NH) 25 Lancaster 4-(4-Methylphenyl)butanoic acid 26 Lancaster 3-(3-Hydroxyphenyl)propanoic acid 27 Sigma-Aldrich 3-(p-Hydroxyphenyl)propanoic acid 28 Sigma-Aldrich 5-Phenylpentanoic acid 29 Sigma-Aldrich 6-Phenylhexanoic acid 30 Matrix Scientific 2-Phenoxyphenylethanoic acid (Columbia, SC) 31 Matrix Scientific 4-Phenoxyphenylethanoic acid 32 Lancaster 7-Phenylheptanoic acid 33 Johnson Matthey 3-(4-Methylphenyl)propanoic acid 34 Johnson Matthey 3-(3,4-Dihydroxyphenyl)propanoic acid 35 Johnson Matthey 3-(2-Hydroxyphenyl)propanoic acid 36 Sigma-Aldrich 3-[4-(Trifluoromethyl)phe- nyl]propanoic acid 37 Sigma-Aldrich 3-[2,5-Bis(Trifluoromethyl)phe- nyl]propanoic acid 38 Trans World 3-(2-Fluorophenyl)propanoic acid Chemicals (Rockville, MD) 39 Trans World 3-(3-Fluorophenyl)propanoic acid Chemicals 40 Sigma-Aldrich 3-(3,4-Difluorophenyl)propanoic acid 41 Trans World 3-(4-Fluorophenyl)propanoic acid Chemicals 42 Trans World 3-(2-Methylphenyl)propanoic acid Chemicals 43 Matrix Scientific 2-(3-Phenoxyphenyl)ethanoic acid 44 Lancaster 4-Phenylbutanoic acid 45 Trans World 3-(2,4-Dichlorophenyl) propanoic acid Chemicals 46 Trans World 3-(2,4-Dimethylphenyl) propanoic acid Chemicals 47 Trans World 3-(2-Chlorophenyl) propanoic acid Chemicals 48 Trans World 3-(3,4-Dichlorophenyl) propanoic acid Chemicals 49 Trans World 3-(3,5-Dimethoxyphenyl)propanoic acid Chemicals 50 Trans World 3-(4-Iodophenyl)propanoic acid Chemicals 51 Trans World 3-(3-Methylphenyl) propanoic acid Chemicals 52 Trans World 3-(4-Chlorophenyl) propanoic acid Chemicals 53 Trans World 3-(4-Ethylphenyl) propanoic acid Chemicals 54 Trans World 3-(3-Iodophenyl) propanoic acid Chemicals 55 Trans World 3-(4-Isopropylphenyl) propanoic acid Chemicals 56 Sigma-Aldrich 3-(3-Chloro-4-methoxyphenyl) propanoic acid 57 Trans World 3-(3-Bromophenyl) propanoic acid Chemicals 58 Trans World 3-(3,4-Dimethylphenyl) propanoic acid Chemicals 59 Trans World 3-(3-Chlorophenyl) propanoic acid Chemicals 60 Trans World 3-(2-Bromophenyl) propanoic acid Chemicals 61 Trans World 3-(4-Bromophenyl)propanoic acid Chemicals 62 Trans World 3-(2-Methoxyphenyl)propanoic acid Chemicals 64 Sigma-Aldrich 3-(4-Methoxyphenyl)propanoic acid 65 Sigma-Aldrich 3-(2,3-Dimethoxyphenyl)propanoic acid 66 Sigma-Aldrich 3-(3,4-Dimethoxyphenyl)propanoic acid 67 Sigma-Aldrich 4-(p-Iodophenyl)butanoic acid 68 Sigma-Aldrich 3-(3,4,5-Trimethoxyphenyl)propanoic acid 69 Sigma-Aldrich 4-(3,4-Dimethoxyphenyl)butanoic acid 70 Sigma-Aldrich 3-[3,5-Bis(Trifluoromethyl)phe- nyl]propanoic acid 71 Sigma-Aldrich 3-(2,4-Dimethoxyphenyl)propanoic acid 72 Sigma-Aldrich 3-(2,5-Dimethoxyphenyl)propanoic acid 73 Oakwood 5-(4-Fluorophenyl)pentanoic acid Products Inc. (West Columbia, SC) 74 Trans World 3-(4-Ethoxyphenyl)propanoic acid Chemicals Active Agents Active agents suitable for use in the present invention include biologically active agents and chemically active agents, including, but not limited to, pesticides, pharmacological agents, and therapeutic agents. Suitable active agents include those that are rendered less effective, ineffective or are destroyed in the gastro-intestinal tract by acid hydrolysis, enzymes and the like. Also included as suitable active agents are those macromolecular agents whose physiochemical characteristics, such as, size, structure or charge, prohibit or impede absorption when dosed orally. For example, biologically or chemically active agents suitable for use in the present invention include, but are not limited to, proteins; polypeptides; peptides; hormones; polysaccharides, and particularly mixtures of muco-polysaccharides; carbohydrates; lipids; small polar organic molecules (i.e. polar organic molecules having a molecular weight of 500 daltons or less); other organic compounds; and particularly compounds which by themselves do not pass (or which pass only a fraction of the administered dose) through the gastro-intestinal mucosa and/or are susceptible to chemical cleavage by acids and enzymes in the gastro-intestinal tract; or any combination thereof. Further examples include, but are not limited to, the following, including synthetic, natural or recombinant sources thereof: growth hormones, including human growth hormones (hGH), recombinant human growth hormones (rhGH), bovine growth hormones, and porcine growth hormones; growth hormone releasing hormones; growth hormone releasing factor, interferons, including α-interferon (e.g., interferon alfacon-1 (available as Infergen® from InterMune, Inc. of Brisbane, Calif.)), β-interferon and γ-interferon; interleukin-1; interleukin-2; insulin, including porcine, bovine, human, and human recombinant, optionally having counter ions including zinc, sodium, calcium and ammonium; insulin-like growth factor, including IGF-1; heparin, including unfractionated heparin, heparinoids, dermatans, chondroitins, low molecular weight heparin, very low molecular weight heparin and ultra low molecular weight heparin; calcitonin, including salmon, eel, porcine and human; erythropoietin; atrial naturetic factor; antigens; monoclonal antibodies; somatostatin; protease inhibitors; adrenocorticotropin, gonadotropin releasing hormone; oxytocin; leutinizing-hormone-releasing-hormone; follicle stimulating hormone; glucocerebrosidase; thrombopoietin; filgrastim; prostaglandins; cyclosporin; vasopressin; cromolyn sodium (sodium or disodium chromoglycate); vancomycin; desferrioxamine (DFO); bisphosphonates, including alendronate, tiludronate, etidronate, clodronate, pamidronate, olpadronate, and incadronate; parathyroid hormone (PTH), including its fragments; anti-migraine agents such as sumatriptan, almotriptan, naratriptan, rizatriptan, frovatriptan, eletriptan, BIBN-4096BS and other calcitonin gene-related proteins antagonists; glucagon-like peptide 1 (GLP-1); Argatroban; glucagon; antimicrobials, including antibiotics, anti-bacterials and anti-fungal agents; vitamins; analogs, fragments, mimetics or polyethylene glycol (PEG)-modified derivatives of these compounds; or any combination thereof. Non-limiting examples of antibiotics include gram-positive acting, bacteriocidal, lipopeptidal and cyclic peptidal antibiotics, such as daptomycin and analogs thereof. Delivery Systems The pharmaceutical composition of the present invention comprises one or more delivery agent compounds of the present invention, and one or more active agents (e.g., biologically active agents). In one embodiment, one or more of the delivery agent compounds, or salts of these compounds, may be used as a delivery agent by mixing delivery agent compounds with the active agent prior to administration to form an administration composition. The administration compositions may be in the form of a liquid. The solution medium may be water (for example, for salmon calcitonin, parathyroid hormone, and erythropoietin), 25% aqueous propylene glycol (for example, for heparin) and phosphate buffer (for example, for rhGH). Other dosing vehicles include, but are not limited to, polyethylene glycol. Dosing solutions may be prepared by mixing a solution of the delivery agent compound with a solution of the active agent, just prior to administration. Alternately, a solution of the delivery agent compound (or active agent) may be mixed with the solid form of the active agent (or delivery agent compound). The delivery agent compound and the active agent may also be mixed as dry powders. The delivery agent compound and the active agent can also be admixed during the manufacturing process. Alternatively, the delivery agent compound and active agent can be separately administered in sequential fashion. The dosing solutions may optionally contain additives such as phosphate buffer salts, citric acid, glycols, or other dispersing agents. Stabilizing additives may be incorporated into the solution, preferably at a concentration ranging between about 0.1 and 20% (w/v). The administration compositions may alternately be in the form of a solid, such as a tablet, capsule or particle, such as a powder or sachet. Solid dosage forms may be prepared by mixing the solid form of the compound with the solid form of the active agent. Alternately, a solid may be obtained from a solution of compound and active agent by methods known in the art, such as freeze-drying (lyophilization), precipitation, crystallization and solid dispersion. The administration compositions of the present invention may also include one or more enzyme inhibitors. Such enzyme inhibitors include, but are not limited to, compounds such as actinonin or epiactinonin and derivatives thereof. Other enzyme inhibitors include, but are not limited to, aprotinin (Trasylol) and Bowman-Birk inhibitors. The amount of active agent used in an administration composition of the present invention is an amount effective to accomplish the purpose of the particular active agent for the target indication. The amount of active agent in the compositions typically is a pharmacologically, biologically, therapeutically, or chemically effective amount. However, the amount can be less than that amount when the composition is used in a dosage unit form because the dosage unit form may contain a plurality of delivery agent compound/active agent compositions or may contain a divided pharmacologically, biologically, therapeutically, or chemically effective amount. The total effective amount can then be administered in cumulative units containing, in total, an effective amount of the active agent. Generally, the amount of delivery agent compound in the composition is an amount effective to facilitate delivery of the active agent. The total amount of active agent and delivery agent to be used can be determined by methods known to the skilled artisan. However, because the compositions of the invention may deliver active agents more efficiently than compositions containing the active agent alone, lower amounts of biologically or chemically active agents than those used in prior dosage unit forms or delivery systems can be administered to the subject, while still achieving the same blood levels and/or therapeutic effects. Generally, the weight ratio of delivery agent to active agent ranges from about 1000:1 or 800:1 to about 10:1 or 1:10, and preferably ranges from about 400:1 or 200:1 to about 100:1 or 25:1. Other ranges are contemplated to be within acceptable ranges for delivery of some active compounds, such as from about 100:1 or 50:1 to about 5:1 or 2.5:1, or from about 60:1 or 30:1 to about 1:1 or 0.5:1. Such ranges and ratios can be determined by one skilled in the art. The presently disclosed delivery agent compounds facilitate the delivery of biologically and chemically active agents, particularly in oral, intranasal, sublingual, intraduodenal, subcutaneous, buccal, intracolonic, rectal, vaginal, mucosal, pulmonary, transdermal, intradermal, parenteral, intravenous, intramuscular and ocular systems, as well as traversing the blood-brain barrier. Dosage unit forms can also include any one or combination of excipients, diluents, disintegrants, lubricants, plasticizers, colorants, flavorants, taste-masking agents, sugars, sweeteners, salts, and dosing vehicles, including, but not limited to, water, 1,2-propane diol, ethanol, olive oil, or any combination thereof. The compounds and compositions of the subject invention are useful for administering biologically or chemically active agents to any animals, including but not limited to birds such as chickens; mammals, such as rodents, cows, pigs, dogs, cats, primates, and particularly humans; and insects. The system is particularly advantageous for delivering chemically or biologically active agents that would otherwise be destroyed or rendered less effective by conditions encountered before the active agent reaches its target zone (i.e. the area in which the active agent of the delivery composition is to be released) and within the body of the animal to which they are administered. Particularly, the compounds and compositions of the present invention are useful for orally administering active agents, especially those that are not ordinarily orally deliverable, or those for which improved delivery is desired. The compositions comprising the phenylalkyl carboxylic acid compounds and active agents have utility in the delivery of active agents to selected biological systems and in an increased or improved bioavailability of the active agent compared to administration of the active agent without the delivery agent. Delivery can be improved by delivering more active agent over a period of time, or in delivering the active agent in a particular time period (such as to effect quicker or delayed delivery), or in delivering the active agent at a specific time, or over a period of time (such as sustained delivery). Another embodiment of the present invention is a method for the treatment or prevention of a disease or for achieving a desired physiological effect, such as any one of the diseases or conditions listed in the table below, in an animal by administering the composition of the present invention. Preferably, an effective amount of the composition for the treatment or prevention of the desired disease or for achieving the desired physiological effect is administered. Specific indications for active agents can be found in the The Physicians' Desk Reference (58th Ed., 2004, Medical Economics Company, Inc., Montvale, N.J.), and Fauci, A S, et. al., Harrison's Principles of Internal Medicine (14th Ed., 1998, McGraw-Hill Health Professions Division, New York. Both of these references are herein incorporated by reference in their entirety. The active agents in the table below include their analogs, fragments, mimetics, and polyethylene glycol-modified derivatives (e.g., the PEGylated derivative of granulocyte colony stimulating factor sold as Neulasta®). TABLE 2 Active agent utilization Active Agent Disease and Physiological Effect Growth hormones (including human recombinant Growth disorders growth hormone and growth-hormone releasing factors and its analogs) Interferons, including α, β and γ Viral infection, including chronic cancer, hepatitis, and multiple sclerosis Interleukins (e.g. Interleukin-1; interleukin-2) Viral infection; cancer; cell mediated immunity; and transplant rejection; Insulin; Insulin-like growth factor IGF-1 Diabetes Immune Globulins, such as IVIg smallpox, rabies, and diphtheria, Alzheimer's Disease; Primary immunodeficiencies; Acute Guillain-Barré syndrome; Chronic idiopathic demyelinating polyneuropathy (CIDP); Myasthenia gravis, polymyositis, and dermatomyositis; neonatal immune thrombocytopenia, heparin-induced thrombocytopenia, and antiphospholipid antibody syndrome: Posttransfusion purpura. Heparin Treatment and Prevention of Thrombosis, including Deep Vein Thrombosis; prevention of blood coagulation Calcitonin Osteoporosis; diseases of the bone; bone pain; analgesic (including pain associated with osteoporosis or cancer) Erythropoietin, Pegylated erythropoietin. Anemia; HIV/HIV-therapy Associated Anemia; Chemotherapeutically-Induced Anemia Atrial naturetic factor Vasodilation Antigens Infection CPHPC Reduction of amyloid deposits and systemic amyloidoisis often (but not always) in connection with Alzheimer's disease,Type II diabetes, and other amyloid-based diseases Monoclonal antibodies To prevent graft rejection; cancer; used in assays to detect diseases Somatostatin/octreotide Bleeding ulcer; erosive gastritis; variceal bleeding; diarrhea; acromegaly; TSH-secreting pituitary adenomas; secretory pancreatic tumors; carcinoid syndrome; reduce proptosis/thyroid-associated ophthalmopathy; reduce macular edema/retinopathy Protease inhibitors HIV Infection/AIDS Adrenocorticotropin High cholesterol (to lower cholesterol) Gonadotropin releasing hormone Ovulatory disfunction (to stimulate ovulation) Oxytocin Labor disfunction (to stimulate contractions) Leutinizing-hormone-releasing-hormone; Regulate reproductive function Leutinizing Hormone; follicle stimulating hormone Glucocerebrosidase Gaucher disease (to metabolize lipoprotein) Thrombopoietin Thrombocytopenia Filgrastim (Granulocyte Colony Stimulating shorten the duration of chemotherapy-induced Factor); GM-CSF, (sargramostim) and their neutropenia and thus treat or prevent infection in Pegylated forms chemotherapy patients; Inhibit the growth of or to kill Mycobacterium Intracellular Avium Infection (MAC) siRNA Huntington, Alzheimers, Viral Infections (HIV, Hepatitis A, B or C, RSV), Cancers; Macular Degeneration Prostaglandins Hypertension Cyclosporin Transplant rejection; psoriasis, inflammatory alopecias; Sjogren's syndrome; Keratoconjunctivitis Sicca Vasopressin Nocturnal Enuresis; antidiuretic Cromolyn sodium; Asthma; allergies Vancomycin Treat or prevent antimicrobial-induced infections including, but not Emitted to methacillin- resistant Staphalococcus aureus and Staph. epidermiditis gallium salts (such as gallium nitrate) Osteoporosis; Paget's disease; Inhibits osteoclasts; Promotes osteoblastic activity, hypercalcemia, including cancer related hypercalcemia, urethral (urinary tract) malignancies; anti-tumors, cancers, including urethral and bladder cancers; lymphoma; malignancies (including bladder cancer); leukemia; management of bone metastases (and associated pain); muliple myeloma, attenuate immune response, including allogenic transplant rejections; disrupt iron metabolism; promote cell migration; wound repair; to attenuate or treat infectious processes of mycobacterium species, including but not limited to mycobacterium tubercolosis, and mycobacterium avium complex Desferrioxamine (DFO) Iron overload Parathyroid hormone (PTH), including its Osteoporosis; fragments. Diseases of the bone Antimicrobials Infection including but not limited to gram-positive bacterial infection Vitamins Treat and prevent Vitamin deficiencies Bisphosphonates Osteoporosis; Paget's disease; bone tumors and metastases (and associated pain); Breast cancer; including as adjuvant therapy for early stage breast cancer; management of bone metastases (and associated pain), including bone metastases associate with breast cancer, prostate cancer, and lung cancer; Inhibits osteoclasts; Promotes osteoblastic activity; treat and/or prevent bone mineral density (bmd) loss; multiple myeloma; prevention of bone complications related to malignant osteolysis; fibrous dysplasia; pediatric osteogenesis imperfecta; hypercalcemia, urethral (urinary tract) malignancies; reflex sympathetic dystropy synodrome, acute back pain after vertebral crush fracture, chronic inflammatory joint disease, renal bone disease, extrosseous calcifications, analgesic, vitamin D intoxication, periarticular ossifications BIBN4096BS - (1-Piperidinecarboxamide. N-[2-[[5- Anti-migraine; calcitonin gene- related peptide amino-1-[ [4-(4-pyridinyl)-l- antagonist piperazinyl)carbonyl]pentyl]amino]-1-[(3,5- dibromo-4-hydroxyphenyl)methyl]-2-oxoethyl]- 4(1,4-dihydro-2-oxo-3(2H0-quinazolinyl)-.[R- (R*,S*)]-) Glucagon improving glycemic control (e.g. treating hypoglycemia and controlling hypoglycemic reactions), obesity; a diagnostic aid in the radiogical examination of the stomach, duodenum, small bowel and colon; Treat acute poisoning With Cardiovascular Agents including, but not limited to, calcium channel blockers, beta blockers GLP-1, Exendin - 3, Exendin - 4, Obestatin Diabetes; improving glycemic control (e.g. treating hypoglycemia and controlling hypoglycemic reactions), obesity dipeptidyl peptidase IV (DPP-4) inhibitors Diabetes; improving glycemic control (e.g. treating hypoglycemia), obesity acyclovir Used to treat herpes infections of the skin, lip and genitals; herpes zoster (shingles); and chickenpox HIV Entry Inhibitors (e.g. Fuzeon) Inhibit entry of HIV into host cells Sumatriptin, almotriptan, naratriptan, rizatriptan, anti-migraine serotonin agonists frovatriptan and eletriptan (piperidinyloxy)phenyl, (piperidinyloxy)pyridinyl, (piperidinylsulfanyl)phenyl and (piperidinylsulfanyl)pyridinyl compounds Neuraminidase inhibitors: peramivir, zanamivir, Antivirals oseltamivir, BCX-1898, BCX-1827, BCX-1989, BCX 1923, BCX 1827 and A315675; M2 inhibitors: amantadine, rimantadine; Nucleoside/Nucleotide Reverse Transcriptase Inhibitors, Non-nucleoside Reverse Transcriptase Inhibitors, Protease Inhibitors, Fusion inhibitors: thiovir, thiophosphonoformate, foscarnet, enfuviritide, zidovudine, didanosine, zalcitabine, stavudine, lamivudine, emtricitabine, abacavir, azidothymidine, tenofovir disoproxil, delavridine, efavirenz, nevirapine, ritonavir, nelfinavir mesylate, saquinvir mesylate, indinavir sulfate, amprenavir, lopinavir, lopinavir, fosamprenavir calcium, atazanavir sulfate Peptide YY (PYY) and PYY-like Peptides (e.g. Obesity, Diabetes, Eating Disorders, Insulin- PYY[3-36]) Resistance Syndromes For example, one embodiment of the present invention is a method for treating a patient suffering from or susceptible to diabetes by administering insulin and at least one of the delivery agent compounds of the present invention. Following administration, the active agent present in the composition or dosage unit form is taken up into the circulation. The bioavailability of the agent can be readily assessed by measuring a known pharmacological activity in blood, e.g. an increase in blood clotting time caused by heparin, or a decrease in circulating calcium levels caused by calcitonin. Alternatively, the circulating levels of the active agent itself can be measured directly. One embodiment of the present invention provides a pharmaceutical composition comprising an effective amount of insulin and an effective amount of at least one of the delivery agents described herein. For example, one embodiment of the present invention provides a pharmaceutical composition comprising about 50 to 800 mg/kg (e.g. 200 mg/kg) of insulin and about 0.1 to 2.0 mg/kg (e.g. 0.5 mg/kg) of any one of the delivery agent compounds of the present invention. Yet another embodiment is method of treating diseases characterized by hyperglycemia, such as diabetes, comprising administering a pharmaceutical composition of the present invention to a subject. One embodiment of the present invention provides a pharmaceutical composition comprising an effective amount of heparin and an effective amount of at least one of the delivery agents described herein. For example, one embodiment of the present invention provides a pharmaceutical composition comprising about 5 to 125 mg/kg (e.g. 25 mg/kg or 80 mg/kg) of heparin and about 5 to 500 mg/kg (e.g. 50 mg/kg or 200 mg/kg) of any one of the delivery agent compounds of the present invention. Yet another embodiment is a method of treating or preventing disease characterized by intravascular thrombi by administering an effective amount of heparin and an effective amount of a delivery agent of the present invention to a subject. Yet another embodiment is a method of preventing Deep Vain Thrombosis (DVT) in susceptible individuals by administering an effective amount of heparin and an effective amount of a delivery agent compound of the present invention to a subject. One embodiment of the present invention provides a pharmaceutical composition comprising an effective amount of rhGH and an effective amount of at least one of the delivery agents described herein. For example, one embodiment of the present invention provides a pharmaceutical composition comprising about 0.25 to 10 mg/kg (e.g. 3 mg/kg) of rhGH and about 50 to 500 mg/kg (e.g. 200 mg/kg) of any one of the delivery agent compounds of the present invention. Yet another embodiment is a method of treating or preventing short stature by administering an effective amount of rhGH and an effective amount of at least one delivery agent compound (formula I) of the present invention to a subject. Yet another embodiment is method of treating or preventing a disease which requires supplementation of growth hormone by administering an effective amount of at least one delivery agent compound of the present invention to a subject. One embodiment of the present invention provides a pharmaceutical composition comprising an effective amount of LHRH and an effective amount of at least one of the delivery agents described herein. For example, one embodiment of the present invention provides a pharmaceutical composition comprising about 0.1 to 10 mg/kg (e.g. 1 mg/kg) of LHRH and about 50-500 mg/kg (e.g. 200 mg/kg) of any one of the delivery agent compounds of the present invention. Yet another embodiment is method of treating or preventing infertility in men or women which requires supplementation of LHRH by administering an effective amount of LHRH and an effective amount of at least one delivery agent of the present invention to a subject. Yet another embodiment is method of treating or preventing a disease which requires supplementation of LHRH by administering an effective amount of LHRH and an effective amount of at least one delivery agent of the present invention to a subject. One embodiment of the present invention provides a pharmaceutical composition comprising an effective amount of caspofungin acetate (e.g. Cancidas®) and an effective amount of at least one of the delivery agents described herein. For example, one embodiment of the present invention provides a pharmaceutical composition comprising about 5 to 125 mg/kg (e.g. 25 mg/kg) of caspofungin acetate and about 50 to 500 mg/kg (e.g. 200 mg/kg) of any one of the delivery agent compounds of the present invention. Yet another embodiment is method of treating or preventing candidiasis or other systemic or localized fungal infections by administering an effective amount of caspofungin acetate and an effective amount of a delivery agent of the present invention to the subject. EXAMPLES The following examples illustrate the present invention without limitation. Example 1—Preparation of 4-(4-Methoxyphenyl)butanoic acid (Compound 1) A 500 mL round bottom flask equipped with a magnetic stirrer bar and an inert atmosphere (nitrogen gas) was charged with 5.25 mL (48.3 mmol) of anisole, 4.83 g (48.3 mmol) of succinic anhydride, 125 mL 1,1,2,2-tetrachloroethane and 125 mL of nitrobenzene. The reaction vessel was cooled with an external ice bath and stirred for 30 minutes. Aluminum trichloride (14.2 g, 106.4 mmol) was added to the pale yellow solution, which then turned to a dark reddish brown color. The ice bath was removed, and the reaction was allowed to stir at room temperature for 36 hours. Reaction was again cooled with an external ice bath. Prepared acidic solution by pouring 1N hydrogen chloride solution into a 100 mL beaker filled with ice. This solution was added to the reaction mixture carefully, drop-wise at first until reaction became clear with white precipitate. After that point a 10 mL portion was carefully added to test for reactivity, and then the remained of the ice/acid mixture was added. A second 100 mL of ice/acid mixture was added, the external ice bath removed and the pale emulsion was stirred for 2 hours. A white precipitate was collected form the emulsion by suction filtration. This solid was dissolved in 300 mL of 0.3 M sodium hydroxide, washed with 100 mL of ethyl acetate, and acidified to ˜pH 1 with 1 M hydrochloric acid. The white precipitate that was collected upon vacuum filtration was washed with 3×100 mL de-ionized water, dried and reserved for use in next procedure. To a 50 mL rounded bottom flask was added 4.77 g (86.1 mmol) of cut zinc. To this was added a solution of 0.22 g (0.81 mmol) of mercury(II)chloride and 0.2 mL concentrated hydrochloric acid (37%) in 4 mL of water. The mixture was allowed to stir at room temperature for 10 minutes. The liquid was decanted off and immediately replaced with a fresh solution of 10 mL concentrated hydrochloric acid (37%) in 2 mL of water. 3.00 g (14.4 mmol) of 4-(4-methoxyphenyl)-4-oxobutyric acid was added to the zinc mixture followed by an additional 10 mL of concentrated hydrochloric acid (37%) and 2 mL water. The reaction was heated to reflux for three hours, with an additional 0.4 mL of concentrated hydrochloric acid (37%) being added every thirty minutes. The reaction was cool to room temperature and allowed to mix overnight. 10 mL of diethyl ether was added to the reaction mixture and stirred for thirty minutes. The liquid was decanted away from the solid into a 125 mL separatory funnel and the solid residue was rinsed with 20 mL of ether which was also decanted into the separatory funnel. The aqueous layer was separated and extracted an additional two times with 30 mL diethyl ether. The combined organic layers were dried over sodium sulfate, filtered and solvent removed under reduced pressure. The residue solid was dissolved in ˜250 mL of 0.3M sodium hydroxide solution and washed with 25 mL of ethyl acetate. The aqueous solution was acidified with ˜200 mL 1N hydrochloric acid solution and allowed to rest overnight. The product (1.42 g, 51%) was isolated as a white solid, mp 57-58° C. Combustion analysis: Found: C, 67.87, H, 7.33%; C11H14O3 requires C: 68.02, H: 7.27% 1H NMR (d6-DMSO): δ 12.0, s, 1H (COOH); δ 7.2 d, 2H (aryl H's); δ 6.8, d, 2H, (arylH's); δ 3.7, s, 3H (OMe H's); δ 2.5, t, 2H (CH2 α to aryl group); δ 2.2, t, 2H (CH2α to COOH), δ 1.75, p, 2H (middle CH2). Example 2—Preparation of 5-(2-Methoxyphenyl)pentanoic acid (Compound 2) A 250 mL 3-neck round bottom flask equipped with a thermometer and a magnetic stirring bar was charged w/ 16.0 mL (18.1 g, 72.3 mmol) of triethyl 4-phosphocrotonate and 20 mL of tetrahydrofuran (THF). The clear solution was cooled to −78° C. in a dry ice/acetone bath and treated with 72.0 mL (72.0 mmol) of 1.0M lithium hexamethylsilizaide/THF solution, added slowly over 10 min. The red solution was stirred at −78° C. for 1 hour. One third of the anion solution was transferred via cannula to a solution of 3.28 g (24.1 mmol) of 2-anisaldehyde and 15 mL of THF. The reaction mixture warmed to 45° C. upon addition and was stirred at 25° C. for 20 hour. After dilution with 2:1 methyl t-butyl ether(MTBE)/hexanes, the reaction mixture was washed with water (4×40 mL) and brine (1×40 mL), dried over sodium sulfate, decolorized with silica gel and concentrated. The ethyl 5-(2-methoxyphenyl)pentadienoate was used as is. A 500 mL Parr shaker reaction vessel was charged with the ethyl 5-(2-methoxyphenyl)pentadienoate isolated above and ethanol. This mixture was treated with 0.25 g of 10% palladium on charcoal and placed under an atmosphere of 45 psig of hydrogen gas in a Parr shaker apparatus. After hydrogen was no longer taken up, the reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to give crude ethyl 5-(2-methoxyphenyl)pentanoate. A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with the ethyl 5-(2-methoxyphenyl)pentanoate isolated above and ethanol. This solution was treated with 2N aqueous sodium hydroxide and heated to reflux. After 5 hr the clear solution was cooled to 25° C., washed with MTBE and acidified with aqueous 4% hydrochloric acid to give a red-orange solid which was isolated by filtration to give 3.44 g of 5-(2-methoxyphenyl)pentanoic acid. 1H NMR (d6-DMSO): δ 11.9, bs, 1H (COOH); δ 7.03, t, 1H, (arylH para to CH2); δ 6.99, d, 1H (arylH ortho to CH2); δ 6.80, d, 1H, (arylH ortho to OMe); δ 6.72, t, 1H (arylH para to OMe); δ 3.64, s, 3H (OCH3); δ 2.41, t, 2H, (CH2 α to aryl); δ 2.09, t, 2H (CH2 α to COOH); δ 1.38, m, 4H (CH2's β to aryl and COOH). 13C NMR (d6-DMSO): 174.42, 157.00, 129.77, 129.49, 127.04, 120.15, 110.56, 55.17, 33.53, 29.18, 28.87, 24.30. Example 3—Preparation of 5-(3-Fluorophenyl)pentanoic acid (Compound 3) A 250 mL 3-neck round bottom flask equipped with a thermometer and a magnetic stirring bar was charged w/ 16.0 mL (18.1 g, 72.3 mmol) of triethyl 4-phosphocrotonate and 20 mL of tetrahydrofuran (THF). The clear solution was cooled to −78° C. in a dry ice/acetone bath and treated with 72.0 mL (72.0 mmol) of 1.0M lithium hexamethylsilizaide/THF solution, added slowly over 10 min. The red solution was stirred at −78° C. for 1 hour. One third of the anion solution was transferred via cannula to a solution of 3.28 g (24.1 mmol) of 3-Fluorobenzaldehyde and 15 mL of THF. The reaction mixture warmed to 45° C. upon addition and was stirred at 25° C. for 20 hour. After dilution with 2:1 methyl t-butyl ether(MTBE)/hexanes, the reaction mixture was washed with water (4×40 mL) and brine (1×40 mL), dried over sodium sulfate, decolorized with silica gel and concentrated. The ethyl 5-(3-Fluorophenyl)pentadienoate was used as is. A 500 mL Parr shaker reaction vessel was charged with the ethyl 5-(3-Fluorophenyl)pentadienoate isolated above and ethanol. This mixture was treated with 0.25 g of 10% palladium on charcoal and placed under an atmosphere of 45 psig of hydrogen gas in a Parr shaker apparatus. After hydrogen was no longer taken up, the reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to give crude ethyl 5-(3-Fluorophenyl)pentanoate. A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with the ethyl 5-(3-Fluorophenyl)pentanoate isolated above and ethanol. This solution was treated with 2N aqueous sodium hydroxide and heated to reflux. After 5 hr the clear solution was cooled to 25° C., washed with MTBE and acidified with aqueous 4% hydrochloric acid to give a red-orange solid which was isolated by filtration to give 3.44 g of 5-(3-Fluorophenyl)pentanoic acid. 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.29, q, 1H, (arylH meta to F); δ 7.0, m, 3H (other arylH's); δ 2.57, t, 2H, (CH2 α to aryl); δ 2.22, t, 2H (CH2 α to COOH); δ 1.5, m, 4H (CH2's β to aryl and COOH). 13C NMR (d6-DMSO): 174.35, 162.2 (d), 145.0, 130.0, 124.36, 114.8, 112.4, 34.37, 33.40, 30.02, 23.98. Example 4—Preparation of 5-(3-Methoxyphenyl)pentanoic acid (Compound 4) A 250 mL 3-neck round bottom flask equipped with a thermometer and a magnetic stirring bar was charged w/ 16.0 mL (18.1 g, 72.3 mmol) of triethyl 4-phosphocrotonate and 20 mL of tetrahydrofuran (THF). The clear solution was cooled to −78° C. in a dry ice/acetone bath and treated with 72.0 mL (72.0 mmol) of 1.0M lithium hexamethylsilizaide/THF solution, added slowly over 10 min. The red solution was stirred at −78° C. for 1 hour. One third of the anion solution was transferred via cannula to a solution of 3.28 g (24.1 mmol) of 3-anisaldehyde and 15 mL of THF. The reaction mixture warmed to 45° C. upon addition and was stirred at 25° C. for 20 hour. After dilution with 2:1 methyl t-butyl ether(MTBE)/hexanes, the reaction mixture was washed with water (4×40 mL) and brine (1×40 mL), dried over sodium sulfate, decolorized with silica gel and concentrated. The ethyl 5-(3-methoxyphenyl)pentadienoate was used as is. A 500 mL Parr shaker reaction vessel was charged with the ethyl 5-(3-methoxyphenyl)pentadienoate isolated above and ethanol. This mixture was treated with 0.25 g of 10% palladium on charcoal and placed under an atmosphere of 45 psig of hydrogen gas in a Parr shaker apparatus. After hydrogen was no longer taken up, the reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to give crude ethyl 5-(3-methoxyphenyl)pentanoate. A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with the ethyl 5-(3-methoxyphenyl)pentanoate isolated above and ethanol. This solution was treated with 2N aqueous sodium hydroxide and heated to reflux. After 5 hr the clear solution was cooled to 25° C., washed with MTBE and acidified with aqueous 4% hydrochloric acid to give a red-orange solid which was isolated by filtration to give 3.44 g of 5-(3-methoxyphenyl)pentanoic acid. 1H NMR (d6-DMSO): δ 11.9, bs, 1H (COOH); δ 7.07, t, 1H, (arylH meta to OMe); δ 6.64, m, 3H (arylH's); δ 3.63, s, 3H (OCH3); δ 2.44, t, 2H, (CH2 α to aryl); δ 2.11, t, 2H (CH2 α to COOH); δ 1.4, m, 4H (CH2's β to aryl and COOH). 13C NMR (d6-DMSO): 174.39, 159.23, 143.58, 129.18, 120.50, 113.89, 111.05, 54.83, 34.81, 33.46, 24.08. Example 5—Preparation of 6-(3-Fluorophenyl) hexanoic acid (Compound 5) A 250 mL 3-neck round bottom flask equipped with a thermometer and a magnetic stirring bar was charged w/ 6.02 g (13.6 mmol) of 4-carboxybutyltriphenylphosphonium bromide and 40 mL of tetrahydrofuran (THF). The slurry was cooled to −40° C. in a dry ice/acetone bath and treated with 28.5 mL (28.5 mmol) of 1.0M lithium hexamethylsilizaide/THF solution. The orange solution was allowed to warm to 25° C. The reaction mixture was cooled to −20° C. and treated with 1.40 mL (1.65 g, 13.3 mmol) of 3-fluorobenzaldehyde and then allowed to warm to 25° C. After 20 hour, the reaction mixture was diluted with methyl t-butyl ether (MTBE) and aqueous saturated sodium bicarbonate solution. The layers were separated. The aqueous phase was acidified with 4% aqueous hydrochloric acid to pH 2 and extracted with MTBE (1×40 mL). The organic phase was washed with brine (1×30 mL), dried over sodium sulfate and concentrated. The 6-(3-fluorophenyl)hex-5-enoic acid was used as is. A 500 mL Parr shaker reaction vessel was charged with the ethyl 6-(3-fluorophenyl)hex-5-enoic acid isolated above, 10 mL of ethylacetate and 30 mL of ethanol. This mixture was treated with 0.24 g of 10% palladium on charcoal and placed under an atmosphere of 58 psig of hydrogen gas in a Parr shaker apparatus. After hydrogen was no longer taken up, the reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to give crude 6-(3-fluorophenyl)hexanoic acid contaminated with triphenylphosphine oxide by-product. The product was taken up into MTBE and purified by extraction into aqueous saturated sodium bicarbonate solution (5×30 mL), acidification with 4% aqueous hydrochloric acid to pH 2 and extraction back into MTBE. The residual phosphine oxide was removed by adding 1 part hexanes to 2 parts MTBE and running through a plug of silica gel. The product was obtained after concentration to 1.37 g of 6-(3-fluorophenyl)hexanoic acid as a clear liquid. 1H NMR (d6-DMSO): δ 11.9, bs, 1H (COOH); δ 7.19, q, 1H, (arylH meta to F); δ 6.9, m, 3H (other arylH's); δ 2.47, t, 2H, (CH2 α to aryl); δ 2.08, t, 2H (CH2 α to COOH); δ 1.44, m, 4H (CH2's β to aryl and COOH); δ 1.17, p, 2H (CH2 in middle of chain). 13C NMR (d6-DMSO): 174.42, 162.2 (d), 145.2, 130.0, 124.35, 114.9, 112.23, 34.61, 33.56, 30.33, 28.09, 24.25. Example 6—Preparation of 3-(4-t-Butylphenyl)propanoic acid (Compound 6) A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with 7.76 g (47.8 mmol) of 4-t-butylbenzaldehyde, 5.28 g (50.7 mmol) of malonic acid and 2.2 mL (2.2 g, 27.2 mmol) of pyridine. The slurry was heated to 80° C., at which temperature a clear yellow solution formed. After stirring for 2 hr, the reaction mixture was cooled to 25° C. The resulting solid was isolated by filtration, rinsing with water (2×30 mL) and 2:1 methyl t-butyl ether (MTBE)/hexanes (2×30 mL). A total of 3.1 g of 4-t-butylcinnamic acid was isolated. A 500 mL Parr shaker reaction vessel was charged with 3.10 g (15.2 mmol) of 4-t-butylcinnamic acid, 20 mL of ethyl acetate and 10 mL of ethanol. This mixture was treated with 0.15 g of 10% palladium on charcoal and placed under an atmosphere of 51 psig of hydrogen gas in a Parr shaker apparatus. A total of 14 psig of hydrogen was taken up in 16 hours. The reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to a white solid, 3-(4-t-butylphenyl)propanoic acid (3.07 g). 1H NMR (d6-DMSO): δ 12.2, bs, 1H (COOH); δ 7.16, d, 2H, (arylH's); δ 7.01, d, 2H (aryH's); δ 2.65, t, 2H, (CH2 α to aryl); δ 2.38, t, 2H (CH2 α to COOH); δ 1.13, s, 9H (t-Bu). 13C NMR (d6-DMSO): 174, 148, 137, 127.8, 125.9, 35, 33, 31.2, 29.5. Example 7—Preparation of 3-(4-n-Butylphenyl)propanoic acid (Compound 7) A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with 7.76 g (47.8 mmol) of 4-n-butylbenzaldehyde, 5.28 g (50.7 mmol) of malonic acid and 2.2 mL (2.2 g, 27.2 mmol) of pyridine. The slurry was heated to 80° C., at which temperature a clear yellow solution formed. After stirring for 2 hr, the reaction mixture was cooled to 25° C. The resulting solid was isolated by filtration, rinsing with water (2×30 mL) and 2:1 methyl t-butyl ether (MTBE)/hexanes (2×30 mL). A total of 3.1 g of 4-n-butylcinnamic acid was isolated. A 500 mL Parr shaker reaction vessel was charged with 3.10 g (15.2 mmol) of 4-n-butylcinnamic acid, 20 mL of ethyl acetate and 10 mL of ethanol. This mixture was treated with 0.15 g of 10% palladium on charcoal and placed under an atmosphere of 51 psig of hydrogen gas in a Parr shaker apparatus. A total of 14 psig of hydrogen was taken up in 16 hours. The reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to a white solid, 3-(4-n-butylphenyl)propanoic acid (3.07 g). 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.00, d, 2H, (arylH's); δ 6.96, d, 2H (aryH's); δ 2.65, t, 2H, (CH2 β to COOH); δ 2.39, m, 4H (CH2 α to COOH and CH2 α to aryl); δ 1.38, p, 2H (CH2β to aryl); δ 1.16, hex, 2H (CH2's γ to aryl); δ 0.77, t, 3H (CH3). 13C NMR (d6-DMSO): 173.79, 139.8, 137.98, 128.15, 128.03, 35.30, 34.40, 33.18, 29.94, 21.72, 13.74. Example 8—Preparation of 3-(4-n-Propylphenyl)propanoic acid (Compound 8) A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with 7.76 g (47.8 mmol) of 4-n-propylbenzaldehyde, 5.28 g (50.7 mmol) of malonic acid and 2.2 mL (2.2 g, 27.2 mmol) of pyridine. The slurry was heated to 80° C., at which temperature a clear yellow solution formed. After stirring for 2 hr, the reaction mixture was cooled to 25° C. The resulting solid was isolated by filtration, rinsing with water (2×30 mL) and 2:1 methyl t-butyl ether (MTBE)/hexanes (2×30 mL). A total of 3.1 g of 4-n-Propylcinnamic acid was isolated. A 500 mL Parr shaker reaction vessel was charged with 3.10 g (15.2 mmol) of 4-n-Propylcinnamic acid, 20 mL of ethyl acetate and 10 mL of ethanol. This mixture was treated with 0.15 g of 10% palladium on charcoal and placed under an atmosphere of 51 psig of hydrogen gas in a Parr shaker apparatus. A total of 14 psig of hydrogen was taken up in 16 hours. The reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to a white solid, 3-(4-n-Propylphenyl)propanoic acid (3.07 g). 1H NMR (d6-DMSO): δ 12.1, bs, 1H (COOH); δ 7.09, d, 2H, (arylH's); δ 7.05, d, 2H (arylH's); δ 2.75, t, 2H, (CH2 β to COOH); δ 2.47, m, 4H (CH2 α to COOH and CH2 α to aryl); δ 1.52, hex, 2H (CH2 β to aryl); δ 0.85, t, 3H (CH3). 13C NMR (d6-DMSO): 173.76, 139.64, 138.01, 128.20, 128.02, 36.86, 35.28, 29.93, 24.11, 13.63. Example 9—Preparation of 3-(4-n-Propoxyphenyl)propanoic acid (Compound 9) A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with 7.76 g (47.8 mmol) of 4-n-Propoxybenzaldehyde, 5.28 g (50.7 mmol) of malonic acid and 2.2 mL (2.2 g, 27.2 mmol) of pyridine. The slurry was heated to 80° C., at which temperature a clear yellow solution formed. After stirring for 2 hr, the reaction mixture was cooled to 25° C. The resulting solid was isolated by filtration, rinsing with water (2×30 mL) and 2:1 methyl t-butyl ether (MTBE)/hexanes (2×30 mL). A total of 3.1 g of 4-n-Propoxycinnamic acid was isolated. A 500 mL Parr shaker reaction vessel was charged with 3.10 g (15.2 mmol) of 4-t-butylcinnamic acid, 20 mL of ethyl acetate and 10 mL of ethanol. This mixture was treated with 0.15 g of 10% palladium on charcoal and placed under an atmosphere of 51 psig of hydrogen gas in a Parr shaker apparatus. A total of 14 psig of hydrogen was taken up in 16 hours. The reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to a white solid, 3-(4-n-Propoxyphenyl)propanoic acid (3.07 g). 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.00, d, 2H, (arylH's meta to O); δ 6.70, d, 2H (arylH's ortho to O); δ 3.76, t, 2H, (OCH2); δ 2.63, t, 2H, (CH2 α to aryl); δ 2.37, t, 2H (CH2 α to COOH); δ 1.59, hex, 2H (CH2 β to O); δ 0.85, t, 3H (CH3). 13C NMR (d6-DMSO): 173.76, 156.97, 132.59, 129.13, 114.22, 68.81, 35.55, 29.48, 22.05, 10.38. Example 10—Preparation of 3-(4-Isopropoxyphenyl)propanoic acid (Compound 10) A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with 7.76 g (47.8 mmol) of 4-Isopropoxybenzaldehyde, 5.28 g (50.7 mmol) of malonic acid and 2.2 mL (2.2 g, 27.2 mmol) of pyridine. The slurry was heated to 80° C., at which temperature a clear yellow solution formed. After stirring for 2 hr, the reaction mixture was cooled to 25° C. The resulting solid was isolated by filtration, rinsing with water (2×30 mL) and 2:1 methyl t-butyl ether (MTBE)/hexanes (2×30 mL). A total of 3.1 g of 4-Isopropoxycinnamic acid was isolated. A 500 mL Parr shaker reaction vessel was charged with 3.10 g (15.2 mmol) of 4-t-butylcinnamic acid, 20 mL of ethyl acetate and 10 mL of ethanol. This mixture was treated with 0.15 g of 10% palladium on charcoal and placed under an atmosphere of 51 psig of hydrogen gas in a Parr shaker apparatus. A total of 14 psig of hydrogen was taken up in 16 hours. The reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to a white solid, 3-(4-4-Isopropoxyphenyl)propanoic acid (3.07 g). 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.00, d, 2H, (arylH's meta to O); δ 6.70, d, 2H (arylH's ortho to O); δ 4.43, hept, 1H, (OCH); δ 2.63, t, 2H, (CH2 α to aryl); δ 2.38, t, 2H (CH2 α to COOH); δ 1.13, d, 6H (CH3's). 13C NMR (d6-DMSO): 173.78, 155.69, 132.50, 129.18, 115.44, 68.98, 35.50, 29.47, 21.85. Example 11—Preparation of 3-(4-n-Butoxyphenyl)propanoic acid (Compound 11) A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with 7.76 g (47.8 mmol) of 4-n-Butoxybenzaldehyde, 5.28 g (50.7 mmol) of malonic acid and 2.2 mL (2.2 g, 27.2 mmol) of pyridine. The slurry was heated to 80° C., at which temperature a clear yellow solution formed. After stirring for 2 hr, the reaction mixture was cooled to 25° C. The resulting solid was isolated by filtration, rinsing with water (2×30 mL) and 2:1 methyl t-butyl ether (MTBE)/hexanes (2×30 mL). A total of 3.1 g of 4-n-Butoxycinnamic acid was isolated. A 500 mL Parr shaker reaction vessel was charged with 3.10 g (15.2 mmol) of 4-n-Butoxycinnamic acid, 20 mL of ethyl acetate and 10 mL of ethanol. This mixture was treated with 0.15 g of 10% palladium on charcoal and placed under an atmosphere of 51 psig of hydrogen gas in a Parr shaker apparatus. A total of 14 psig of hydrogen was taken up in 16 hours. The reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to a white solid, 3-(4-n-Butoxyphenyl)propanoic acid (3.07 g). 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.00, d, 2H, (arylH's meta to O); δ 6.70, d, 2H (arylH's ortho to O); δ 3.79, t, 2H, (OCH2); δ 2.62, t, 2H, (CH2 α to aryl); δ 2.35, t, 2H (CH2 α to COOH); δ 1.55, p, 2H (CH2 β to O); δ 1.30, hex, 2H (CH2 β to COOH); δ 0.80, t, 3H (CH3). 13C NMR (d6-DMSO): 173.77, 156.98, 132.58, 129.12, 114.21, 66.98, 35.56, 30.77, 29.48, 18.73, 13.67. Example 12—Preparation of 3-(3-Phenoxyphenyl)propanoic acid (Compound 12) A 125 mL Ehrlenmayer flask equipped with a magnetic stirrer bar was charged with 7.76 g (47.8 mmol) of 3-Phenoxybenzaldehyde, 5.28 g (50.7 mmol) of malonic acid and 2.2 mL (2.2 g, 27.2 mmol) of pyridine. The slurry was heated to 80° C., at which temperature a clear yellow solution formed. After stirring for 2 hr, the reaction mixture was cooled to 25° C. The resulting solid was isolated by filtration, rinsing with water (2×30 mL) and 2:1 methyl t-butyl ether (MTBE)/hexanes (2×30 mL). A total of 3.1 g of 3-Phenoxycinnamic acid was isolated. A 500 mL Parr shaker reaction vessel was charged with 3.10 g (15.2 mmol) of 3-Phenoxycinnamic acid, 20 mL of ethyl acetate and 10 mL of ethanol. This mixture was treated with 0.15 g of 10% palladium on charcoal and placed under an atmosphere of 51 psig of hydrogen gas in a Parr shaker apparatus. A total of 14 psig of hydrogen was taken up in 16 hours. The reaction mixture was removed from the Parr shaker apparatus after dissipating the hydrogen gas, filtered through a Celite pad to remove the catalyst and concentrated to a white solid, 3-(3-Phenoxyphenyl)propanoic acid (3.07 g). 1H NMR (d6-DMSO): δ 12.1, bs, 1H (COOH); δ 7.37, t, 2H, (arylH's meta to O on unsubstituted phenyl); δ 7.27, t, 1H, (arylH meta to O on substituted phenyl); δ 7.12, t, 1H, (arylH para to O on unsubstituted phenyl); δ 6.99, m, 3H (arlyH's); δ 6.89, s, 1H (arylH ortho to both O and CH2); δ 6.79, dd, 1H (arylH's ortho to O on substituted phenyl); δ 2.79, t, 2H, (CH2 α to aryl); δ 2.51, t, 2H (CH2 α to COOH). 13C NMR (d6-DMSO): 173.62, 156.65, 156.49, 143.22, 129.97, 129.80, 123.41, 123.27, 118.61, 118.44, 116.13, 34.98, 30.10. Example 13—Preparation of 3-(3-Ethoxyphenyl)propanoic acid (Compound 13) A 75 mL mini-block tube equipped with a magnetic stirrer bar was charged with 6.16 g (50.4 mmol) of 3-hydroxybenzaldehyde, 4.40 mL (8.58 g, 55.0 mmol) of ethyl iodide 30 mL of dimethylformamide and 6.05 g (57.1 mmol) of sodium carbonate. The slurry was heated to 50° C. After 40 hours the reaction was only 50% complete so another 3 ml (5.85 g, 37.4 mmol) ethyl iodide was added. After 60 more hours another 3 mL (5.85 g, 37.4 mmol) ethyl iodide and 3 g (28.5 mmol) of sodium carbonate were added. The reaction mixture was cooled to 25° C. and diluted with methyl t-butyl ether (MTBE) and water. The organic layer was decanted off. The aqueous phase was rinsed with MTBE, again decanting off the organic layer. The combined organic layers were washed with 2N aqueous sodium hydroxide (3×30 mL) and brine (1×30 mL), dried over sodium sulfate and concentrated to give 3-ethoxybenzaldehyde which was used as (following the above procedure for 3-(4-t-butylphenyl)propanoic acid (Example 6) to prepare 3-(3-ethoxyphenyl)propanoic acid (1.31 g) as an off-white solid. 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.04, t, 1H, (arylH meta to OEt); δ 6.6, m, 3H (other arylH's); δ 3.85, q, 2H, (OCH2); 2.65, t, 2H, (CH2 α to aryl); δ 2.39, t, 2H (CH2 α to COOH); δ 1.18, t, 3H (CH3). 13C NMR (d6-DMSO): 173.73, 158.49, 142.41, 129.25, 120.30, 114.40, 111.79, 62.72, 35.12, 30.35, 14.66. Example 14—Preparation of 3-(3-Isopropoxyphenyl)propanoic acid (Compound 14) A 75 mL mini-block tube equipped with a magnetic stirrer bar was charged with 6.16 g (50.4 mmol) of 3-hydroxybenzaldehyde, 4.40 mL (8.58 g, 55.0 mmol) of isopropyl iodide 30 mL of dimethylformamide and 6.05 g (57.1 mmol) of sodium carbonate. The slurry was heated to 50° C. After 40 hours the reaction was only 50% complete so another 3 ml (5.85 g, 37.4 mmol) isopropyl iodide was added. After 60 more hours another 3 mL (5.85 g, 37.4 mmol) isopropyl iodide and 3 g (28.5 mmol) of sodium carbonate were added. The reaction mixture was cooled to 25° C. and diluted with methyl t-butyl ether (MTBE) and water. The organic layer was decanted off. The aqueous phase was rinsed with MTBE, again decanting off the organic layer. The combined organic layers were washed with 2N aqueous sodium hydroxide (3×30 mL) and brine (1×30 mL), dried over sodium sulfate and concentrated to give 3-Isopropoxybenzaldehyde which was used as (following the above procedure for 3-(4-t-butylphenyl)propanoic acid (Example 6) to prepare 3-(3-Isopropoxyphenyl)propanoic acid (1.31 g) as an off-white solid. 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.03, t, 1H, (arylH meta to O-i-Pr); δ 6.6, m, 3H (other arylH's); δ 4.45, hept, 1H, (OCH); 2.65, t, 2H, (CH2 α to aryl); δ 2.38, t, 2H (CH2 α to COOH); δ 1.12, d, 6H (CH3's). 13C NMR (d6-DMSO): 173.70, 157.40, 142.46, 129.26, 120.17, 115.54, 112.93, 68.77, 35.09, 30.31, 21.84. Example 15—Preparation of 3-(3-n-Butoxyphenyl)propanoic acid (Compound 15) A 75 mL mini-block tube equipped with a magnetic stirrer bar was charged with 6.16 g (50.4 mmol) of 3-hydroxybenzaldehyde, 4.40 mL (8.58 g, 55.0 mmol) of n-butyl iodide 30 mL of dimethylformamide and 6.05 g (57.1 mmol) of sodium carbonate. The slurry was heated to 50° C. After 40 hours the reaction was only 50% complete so another 3 ml (5.85 g, 37.4 mmol) n-butyl iodide was added. After 60 more hours another 3 mL (5.85 g, 37.4 mmol) n-butyl iodide and 3 g (28.5 mmol) of sodium carbonate were added. The reaction mixture was cooled to 25° C. and diluted with methyl t-butyl ether (MTBE) and water. The organic layer was decanted off. The aqueous phase was rinsed with MTBE, again decanting off the organic layer. The combined organic layers were washed with 2N aqueous sodium hydroxide (3×30 mL) and brine (1×30 mL), dried over sodium sulfate and concentrated to give 3-n-Butoxybenzaldehyde which was used as (following the above procedure for 3-(4-t-Butylphenyl)propanoic acid (Example 6) to prepare 3-(3-n-Butoxyphenyl)propanoic acid (1.31 g) as an off-white solid. 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.04, t, 1H, (arylH meta to O-i-Pr); δ 6.6, m, 3H (other aryH's); δ 3.82, t, 2H, (OCH2); 2.65, t, 2H, (CH2 α to aryl); δ 2.38, t, 2H (CH2 α to COOH); δ 1.56, p, 2H (CH2 β to O); δ 1.30, hex, 2H (CH2 β to COOH); δ 0.81, t, 3H (CH3). 13C NMR (d6-DMSO): 173.75, 158.69, 142.44, 129.23, 120.28, 114.40, 111.84, 66.87, 35.15, 30.78, 30.36, 18.74, 13.68. Example 16—Preparation of 3-(3-n-Propoxyphenyl)propanoic acid (Compound 16) A 75 mL mini-block tube equipped with a magnetic stirrer bar was charged with 6.16 g (50.4 mmol) of 3-hydroxybenzaldehyde, 4.40 mL (8.58 g, 55.0 mmol) of n-propyl iodide 30 mL of dimethylformamide and 6.05 g (57.1 mmol) of sodium carbonate. The slurry was heated to 50° C. After 40 hours the reaction was only 50% complete so another 3 ml (5.85 g, 37.4 mmol) n-propyl iodide was added. After 60 more hours another 3 mL (5.85 g, 37.4 mmol) n-propyl iodide and 3 g (28.5 mmol) of sodium carbonate were added. The reaction mixture was cooled to 25° C. and diluted with methyl t-butyl ether (MTBE) and water. The organic layer was decanted off. The aqueous phase was rinsed with MTBE, again decanting off the organic layer. The combined organic layers were washed with 2N aqueous sodium hydroxide (3×30 mL) and brine (1×30 mL), dried over sodium sulfate and concentrated to give 3-n-Propylbenzaldehyde which was used as (following the above procedure for 3-(4-t-butylphenyl)propanoic acid (Example 6) to prepare 3-(3-n-Propylphenyl)propanoic acid (1.31 g) as an off-white solid. 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.04, t, 1H, (arylH meta to O-i-Pr); δ 6.6, m, 3H (other aryH's); δ 3.77, t, 2H, (OCH2); 2.66, t, 2H, (CH2 α to aryl); δ 2.39, t, 2H (CH2 α to COOH); δ 1.59, hex, 2H (CH2 β to O); δ 0.85, t, 3H (CH3). 13C NMR (d6-DMSO): 173.74, 158.67, 142.42, 129.25, 120.29, 114.42, 111.85, 68.68, 35.12, 30.34, 22.05, 10.40. Example 17—Preparation of 3-(3-Isobutoxyphenyl)propanoic acid (Compound 17) A 75 mL mini-block tube equipped with a magnetic stirrer bar was charged with 6.16 g (50.4 mmol) of 3-hydroxybenzaldehyde, 4.40 mL (8.58 g, 55.0 mmol) of isobutyl iodide 30 mL of dimethylformamide and 6.05 g (57.1 mmol) of sodium carbonate. The slurry was heated to 50° C. After 40 hours the reaction was only 50% complete so another 3 ml (5.85 g, 37.4 mmol) isobutyl iodide was added. After 60 more hours another 3 mL (5.85 g, 37.4 mmol) isobutyl iodide and 3 g (28.5 mmol) of sodium carbonate were added. The reaction mixture was cooled to 25° C. and diluted with methyl t-butyl ether (MTBE) and water. The organic layer was decanted off. The aqueous phase was rinsed with MTBE, again decanting off the organic layer. The combined organic layers were washed with 2N aqueous sodium hydroxide (3×30 mL) and brine (1×30 mL), dried over sodium sulfate and concentrated to give 3-Isobutoxybenzaldehyde which was used as (following the above procedure for 3-(4-t-butylphenyl)propanoic acid (Example 6) to prepare 3-(3-Isobutoxyphenyl)propanoic acid (1.31 g) as an off-white solid. 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.03, t, 1H, (arylH meta to O-i-Pr); δ 6.6, m, 3H (other aryH's); δ 3.59, d, 2H, (OCH2); 2.65, t, 2H, (CH2 α to aryl); δ 2.38, t, 2H (CH2 α to COOH); δ 1.86, n, 1H, (CH); δ 0.84, d, 6H (CH3's). 13C NMR (d6-DMSO): 173.74, 158.78, 142.43, 129.24, 120.31, 114.42, 111.92, 73.53, 35.13, 30.35, 27.71, 19.06. Example 18—Preparation of 3-(4-Isobutoxyphenyl)propanoic acid (Compound 18) A 75 mL mini-block tube equipped with a magnetic stirrer bar was charged with 6.16 g (50.4 mmol) of 4-hydroxybenzaldehyde, 4.40 mL (8.58 g, 55.0 mmol) of isobutyl iodide 30 mL of dimethylformamide and 6.05 g (57.1 mmol) of sodium carbonate. The slurry was heated to 50° C. After 40 hours the reaction was only 50% complete so another 3 ml (5.85 g, 37.4 mmol) isobutyl iodide was added. After 60 more hours another 3 mL (5.85 g, 37.4 mmol) isobutyl iodide and 3 g (28.5 mmol) of sodium carbonate were added. The reaction mixture was cooled to 25° C. and diluted with methyl t-butyl ether (MTBE) and water. The organic layer was decanted off. The aqueous phase was rinsed with MTBE, again decanting off the organic layer. The combined organic layers were washed with 2N aqueous sodium hydroxide (3×30 mL) and brine (1×30 mL), dried over sodium sulfate and concentrated to give 4-Isobutoxybenzaldehyde which was used as (following the above procedure for 3-(4-t-butylphenyl)propanoic acid (Example 6) to prepare 3-(4-Isobutoxyphenyl)propanoic acid (1.31 g) as an off-white solid. 1H NMR (d6-DMSO): δ 12.0, bs, 1H (COOH); δ 7.00, d, 2H, (arylH's meta to O); δ 6.70, d, 2H (arylH's ortho to O); δ 3.60, d, 2H, (OCH2); 2.65, t, 2H, (CH2 α to aryl); δ 2.38, t, 2H (CH2 α to COOH); δ 1.89, n, 1H, (CH); δ 0.87, d, 6H (CH3's). 13C NMR (d6-DMSO): 173.76, 157.06, 132.61, 129.12, 114.26, 73.67, 35.56, 29.47, 27.68, 19.04. Example 19—Preparation of 4-(4-Ethylphenyl)butanoic acid (Compound 19) A 250 mL 3-neck round bottom flask equipped with a thermometer and a magnetic stirring bar was charged with 4.01 g (61.3 mmol) of zinc dust and 35 mL of dimethylformamide (DMF) under a nitrogen atmosphere. The slurry was treated with 0.56 g (2.2 mmol) of iodine. The red disappeared in 90 seconds. The reaction mixture was treated with 6.00 mL (8.18 g, 42.0 mmol) of ethyl 4-bromobutyrate and heated to 80° C. for 4 hour. The reaction mixture was cooled to 30° C. and treated with 4.98 g (21.5 mmol) of 4-iodoethylbenzene and 0.48 g (0.9 mmol) of dichlorobis(triphenylphosphine)nickel(II). The reaction mixture was heated to 45° C. for 80 hours. The cooled reaction mixture was treated with aqueous 4% hydrochloric acid to quench the excess zinc. The mixture was extracted with methyl t-butyl ether (MTBE) (1×60 mL). The organic phase was washed with brine (1×30 mL), dried over sodium sulfate and concentrated. The crude ethyl 4-(4-ethylphenyl)butyrate was taken up in ethanol, treated with 20 mL of 2N aqueous sodium hydroxide, and heated to reflux. After 4 hours the reaction mixture was cooled to 25° C. and washed with MTBE (2×30 mL). The aqueous phase was acidified with aqueous 4% hydrochloric acid. A solid was isolated by filtration to give 1.99 g of 4-(4-ethylphenyl)butanoic acid. 1H NMR (d6-DMSO): δ 11.9, bs, 1H (COOH); δ 6.98, d, 2H, (arylH's); δ 6.95, d, 2H (aryH's); δ 2.41, m, 4H, (CH2's α to aryl); δ 2.07, t, 2H (CH2 α to COOH); δ 1.64, m, 2H (CH2 β to both aryl and COOH); δ 1.03, t, 3H (CH3). 13C NMR (d6-DMSO): 174.23, 141.08, 138.67, 128.20, 127.65, 33.97, 33.03, 27.73, 26.35, 15.65. Example 20—Preparation of 4-(4-Isopropylphenyl)butanoic acid (Compound 20) A 250 mL 3-neck round bottom flask equipped with a thermometer and a magnetic stirring bar was charged with 4.01 g (61.3 mmol) of zinc dust and 35 mL of dimethylformamide (DMF) under a nitrogen atmosphere. The slurry was treated with 0.56 g (2.2 mmol) of iodine. The red disappeared in 90 seconds. The reaction mixture was treated with 6.00 mL (8.18 g, 42.0 mmol) of ethyl 4-bromobutyrate and heated to 80° C. for 4 hour. The reaction mixture was cooled to 30° C. and treated with 4.98 g (21.5 mmol) of 4-iodoisopropylbenzene and 0.48 g (0.9 mmol) of dichlorobis(triphenylphosphine)nickel(II). The reaction mixture was heated to 45° C. for 80 hours. The cooled reaction mixture was treated with aqueous 4% hydrochloric acid to quench the excess zinc. The mixture was extracted with methyl t-butyl ether (MTBE) (1×60 mL). The organic phase was washed with brine (1×30 mL), dried over sodium sulfate and concentrated. The crude ethyl 4-(4-Isopropylphenyl)butyrate was taken up in ethanol, treated with 20 mL of 2N aqueous sodium hydroxide, and heated to reflux. After 4 hours the reaction mixture was cooled to 25° C. and washed with MTBE (2×30 mL). The aqueous phase was acidified with aqueous 4% hydrochloric acid. A solid was isolated by filtration to give 1.99 g of 4-(4-Isopropylphenyl)butanoic acid. 1H NMR (d6-DMSO): δ 11.9, bs, 1H (COOH); δ 7.01, d, 2H, (arylH's); δ 6.96, d, 2H (aryH's); δ 2.70, hept, 1H, (CH) δ 2.40, t, 2H, (CH2 α to aryl); δ 2.07, t, 2H (CH2 α to COOH); δ 1.63, p, 2H (CH2 β to both aryl and COOH); δ 1.04, d, 6H (CH3's). 13C NMR (d6-DMSO): 174.23, 145.75, 138.81, 128.18, 126.15, 33.97, 33.07, 32.99, 26.33, 23.93. Example 21—Preparation of 5-(4-Ethylphenyl)pentanoic acid (Compound 21) A 250 mL 3-neck round bottom flask equipped with a thermometer and a magnetic stirring bar was charged with 4.01 g (61.3 mmol) of zinc dust and 35 mL of dimethylformamide (DMF) under a nitrogen atmosphere. The slurry was treated with 0.56 g (2.2 mmol) of iodine. The red disappeared in 90 seconds. The reaction mixture was treated with 6.00 mL (8.18 g, 42.0 mmol) of ethyl 4-bromopentanoate and heated to 80° C. for 4 hour. The reaction mixture was cooled to 30° C. and treated with 4.98 g (21.5 mmol) of 4-iodoethylbenzene and 0.48 g (0.9 mmol) of dichlorobis(triphenylphosphine)nickel(II). The reaction mixture was heated to 45° C. for 80 hours. The cooled reaction mixture was treated with aqueous 4% hydrochloric acid to quench the excess zinc. The mixture was extracted with methyl t-butyl ether (MTBE) (1×60 mL). The organic phase was washed with brine (1×30 mL), dried over sodium sulfate and concentrated. The crude ethyl 4-(4-ethylphenyl)pentanoate was taken up in ethanol, treated with 20 mL of 2N aqueous sodium hydroxide, and heated to reflux. After 4 hours the reaction mixture was cooled to 25° C. and washed with MTBE (2×30 mL). The aqueous phase was acidified with aqueous 4% hydrochloric acid. A solid was isolated by filtration to give 1.99 g of 4-(4-ethylphenyl)pentanoic acid. 1H NMR (d6-DMSO): δ 11.9, bs, 1H (COOH); δ 6.98, d, 2H, (arylH's); δ 6.95, d, 2H (aryH's); δ 2.42, m, 4H, (CH2's α to aryl); δ 2.09, t, 2H (CH2 α to COOH); δ 1.4, m, 2H (CH2's β to aryl and COOH); δ 1.03, t, 3H (CH3). 13C NMR (d6-DMSO): 174.38, 140.90, 139.11, 128.15, 127.57, 34.39, 33.49, 30.45, 27.73, 24.09, 15.66. Example 22—Oral Delivery of Insulin to Male Sprague-Dawley Rats Insulin stock solution (15 mg/ml) (Human zinc insulin, Calbiochem-Novabiochem Corp., La Jolla, Calif.) was prepared with deionized water. Oral dosing compositions containing 200 mg/kg of delivery agent compound and 0.5 mg/kg of insulin in aqueous solution were prepared with the delivery agent compound shown in Table 3 below. Either the sodium salt of the delivery agent compound was used or the free acid was converted to the sodium salt with one equivalent of sodium hydroxide. The dosing solution was administered to fasted male Sprague-Dawley rats by oral gavage with an average weight of about 225-250 grams. Blood glucose levels were then determined by glucometer (One Touch Ultra®, LifeScan, Inc.) and compared to vehicle control (1 ml/kg of water). Samples were collected prior to dosing (time 0) and at 15, 30, 45 and 60 minutes after dosing. The % glucose reduction values in Table 3 are values found at the C minimum, and are an average % reduction with respect to the number of times the experiment was run for each delivery agent. TABLE 3 Percent Change in Glucose after delivery agent & insulin administration Insulin 200 mg/kg Delivery Agent Compound; 0.5 mg/kg Insulin Delivery Delivery Agent % Glucose Agent % Glucose Compound Reduction Compound Reduction 1 −43.6 36 −16.0 3 −6.6 37 −13.8 4 −38.3 38 −54.6 5 −12.3 38 −24.0 6 −50.4 39 −63.3 6 −59.8 39 −39.5 6 −47.4 39 −31.6 6 −53.1 40 −40.8 7 −8.9 41 −43.9 8 −6.2 41 −33.3 9 −30.5 42 −36.5 9 −25.8 43 −24.1 10 −49.4 44 −53.4 10 −61.7 44 −34.6 11 −8.5 44 −33.3 12 −24.8 45 −12.2 13 −40.2 46 −12.0 14 −42.9 47 −29.2 15 −8.5 48 −2.4 16 −22.1 49 −32.0 17 −12.6 50 −46.1 18 −43.6 50 −42.9 19 −31.0 51 −29.3 20 −23.2 52 −18.2 21 −14.6 53 −50.6 23 −13.6 53 −35.5 25 −53.8 53 −56.9 25 −45.3 54 −18.0 25 −34.2 55 −45.2 25 −20.2 55 −42.7 26 −6.6 55 −36.0 27 −10.8 55 −48.4 27 −10.8 56 −21.8 28 −57.3 57 −26.5 28 −50.2 58 −40.2 28 −53.7 59 −52.0 28 −53.8 59 −31.2 28 −39.2 60 −36.7 29 −22.5 61 −41.0 32 −13.8 61 −20.5 33 −20.9 62 −20.5 33 −22.7 62 −26.4 34 −27.2 63 −4.5 35 −18.2 63 −13 64 −45.5 69 13.1 64 −29.6 70 −5.5 65 −30.7 71 −14.1 66 −19.5 72 −13.1 67 −8.6 73 −37.3 68 −36.1 16597053 novo nordisk north america operations a/s USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open Mar 31st, 2022 02:17PM Mar 31st, 2022 02:17PM Novo Nordisk Health Care Pharmaceuticals & Biotechnology
nyse:nvo Novo Nordisk Feb 7th, 2006 12:00AM Aug 9th, 2001 12:00AM https://www.uspto.gov?id=US06994261-20060207 Support for a cartridge for transferring an electronically readable item of information from the cartridge to an electronic circuit A support for a medication cartridge provided with one or more electronically readable items of information carrying areas is disclosed. The support disclosed has a number of closely spaced mutually electrically insulated conductors embedded in an electrically insulating material. The support is useful in assisting reading the item of information stored on the outside of the medication containing cartridge. 6994261 1. A support for a cartridge with one or more electronically readable items of information carrying areas, wherein the support for the cartridge is at least partially constituted by one or more electrically connecting supports, each comprising a number of closely spaced mutually electrically insulated conductors embedding in an electrically insulating material that stretches from one supporting surfaces of the cartridge to a contact area for receiving and transferring the information, when the cartridge is position in the support and wherein each of the electrically connecting supports is constituted by alternating layers of electrically conducting material of maximum thickness Tcl and electrically insulating material of maximum thickness Til, respectively. 2. A support for a cartridge provided with one or more electronically readable items of information carrying area, wherein the support is at least partially constituted by one or more electrically connecting supports, each comprising a number of closely spaced mutually electrically insulated conductors embedded in an electrically insulating material that stretches from one of the supporting surfaces of the cartridge to a contact area for receiving and transferring the information, when the cartridge is positioned in the support and wherein he cartridge has an axis of symmetry and the contact area comprises a group of identical and regularly spaced electrically conducting pad of width Wcp in the direction of adjacent pads, the adjacent pads being separated by an electrically insulating area of width Diacp and the following relations between said distances are fulfilled: Diacp>2*Til+Tcl. 3. A support for a cartridge provided with one or more electronically readable items of information carrying area, wherein the support is at least partially constituted by one or more electrically connecting supports, each comprising a number of closely spaced mutually electrically insulated conductors embedded in an electrically insulating material that stretches from one of the supporting surfaces of the cartridge to a contact area for receiving and transferring the information, when the cartridge is positioned in the support, wherein the cartridge has an axial direction of symmetry and the cartridge is provided with a plurality of rectangular, essentially parallel, identically sized information carrying areas of height Hica in the direction of the circumference of the axis of symmetry, the information carrying areas being spaced with equal mutual distance Dica along the periphery of the cartridge in the direction of a circumference of the axis of symmetry and the supporting means comprising two rectangular, essentially parallel, identical electrically connecting supports of height Hctm in the direction perpendicular to the axis of symmetry of the cartridge, separated by an electrically insulating volume of width Dctm between the two electrically connecting supports and satisfying the following relationship between the distances: Hica<Dctm<2*Hica+Dica, and Hctm<Dica<2*Hctm+Dctm. 3 CROSS-REFERENCE TO RELATED APPLICATIONS The present application claims priority on U.S. provisional application No. 60/229,106, filed on Aug. 30, 2000. TECHNICAL FIELD OF THE INVENTION The invention relates to the electronic marking of cartridges or the like. The invention relates specifically to: A support for a cartridge provided with one or more electronically readable information carrying areas. The invention furthermore relates to: The use of a composite material, comprising a number of closely spaced mutually electrically insulated conductors embedded in an electrically insulating material. DESCRIPTION OF RELATED ART The following account of the prior art relates to one of the areas of application of the present invention, the electronic marking of medication cartridges. The marking of medication cartridges to be able to electronically read details of its contents is of increasing importance, i.a. to ensure a safe and convenient use of the medication in connection with a patient's self-treatment of a disease such as diabetes. In order for the user to feel secure about handling the medication it is important that errors in his or hers use of the drug are avoided. One remedy to avoid errors is an intelligent marking of the contents (i.e. drug, concentration, relevant dose, last day of use, etc.) of the medication cartridge. This includes a visually intelligible marking of the cartridge for reading by a user as well as an electronically readable code for use by a processing unit of the medication delivery device, the processing unit being used for monitoring and controlling the actual delivery of the medication to the user and for making a record of the drug administration history, etc. In other words a certain area of the medication cartridge must be reserved to hosting an increasing amount of information. The information on a medication cartridge must be provided in a safe and simple way that is easily transferred from the cartridge to an electronic processing circuit. U.S. Pat. No. 5,954,700 discloses a cartridge for containing a fluid and for use with an electronic delivery device that includes a cartridge housing for holding the fluid, and an information providing source. The information-providing source may be a set of wires and contacts, or contact bands that provide the predetermined information to an electronic delivery device by producing a binary code. To implement a specific binary code requires an individual customisation of the cartridge as regards the contacts or contact bands and the wires connecting them to a positive or negative voltage. Our co-pending application “Electronic marking of a medication cartridge” discloses a method of marking a medication cartridge that is simple and improves safety in reading. The method provides the information redundantly and implements in one of its embodiments a given binary code by adding insulating areas to a conducting foil (connected to a supply voltage) and transferring the information to a processing unit by means of a support based on the principles outlined in the present patent application. Another remedy for avoiding errors and for making the user feel comfortable with the handling is that the drug contained in the medication cartridge is visible from outside, so that the user is able to check the color, the uniformity, whether impurities are present, etc. For this reason, as large a part of the surface of the medication cartridge as possible should be free of labels and other opaque items that limit a user's view of the contents. Further, there is a general trend to miniaturization of electronic devices including medication delivery devices, so that they are easy to carry and discreet in use. DISCLOSURE OF THE INVENTION Thus there is a need for a way of transferring densely coded information on the, typically curved, surface of a medication cartridge to an electronic circuit connected to a processing circuit, said information being provided in binary form on the cartridge by a mixture of patterns of electrically conducting and electrically insulating wires, areas or patches or the like. The object of the present invention is to provide means, which are capable of securely transferring information with an increased density from a cartridge to an electronic circuit, and which are flexible and may be customized to a variety of physical designs. This is achieved according to the invention in that the support for the cartridge is at least partially constituted by one or more electrically connecting supports, each comprising a number of closely spaced mutually electrically insulated conductors embedded in an electrically insulating material that stretches from one of the supporting surfaces of the cartridge to a contact area for receiving and transferring the information, when said cartridge is positioned in said support. When each of said one or more electrically connecting supports is constituted by alternating layers of electrically conducting material of maximum thickness Tcl and electrically insulating material of maximum thickness Til, respectively, it is ensured that a simple and flexible solution is provided. By controlling the thicknesses of the two layer types, the maximum density of information may be controlled. When said support is made of elastic materials, it may be ensured that the support conforms to the shape of the cartridge when the cartridge is positioned in the support with a certain minimum pressure. I.e. it makes the support even more flexible and relaxes the tolerances to its conformity with the cartridge and with the contact area (e.g. pads on a printed circuit board (PCB) for connecting to a processing circuit on the PCB). In a preferred embodiment said one or more electrically connecting supports are made of elastomeric materials. In a preferred embodiment said electrically conducting material consists of silicone rubber with a concentration of carbon black sufficient for electrical conduction. When said cartridge has an axial direction of symmetry, and said information carrying areas are located preferably in one axial end of the cartridge, it is ensured that the main part of the cartridge is not covered by the electronically readable information and may be held free for optical inspection of the contents of the cartridge (in case of a transparent cartridge). By using a fine layer pitch in the electrically connecting supports, the electronically readable information may be densely written and thus be concentrated to an end of the cartridge that is used for a lid or cover, in which case the whole effective volume available for housing the medication may be open for inspection. When said cartridge has an axial direction of symmetry, and said information carrying areas are located preferably in an axial direction of the cartridge covering only a limited angular sector, it is ensured that the whole length of the cartridge in its axial direction may be used for coding information and at the same time it is possible to inspect the contents of the cartridge over the full axial length of the cartridge. In a preferred embodiment said support comprises one electrically connecting support preferably stretching in an axial direction of the cartridge. When said support comprises two or more electrically connecting supports each stretching preferably in an axial direction of the cartridge and being located side by side along the radial periphery of the cartridge, it is ensured that the electronically readable information may be written several times in information carrying areas distributed along the radial periphery of the cartridge and that two or more of the areas may be simultaneously read. The information in each area may be different, in which case the information capacity is increased. Alternatively, the information in each area may be redundantly provided by coding the same information in all areas or by alternatingly coding its binary true and inverted forms. The redundancy may be used to implement checks to improve reading security. When the surface of the support facing towards the cartridge, including the one or more electrically connecting supports, in an axial cross section correspond to the surface of the cartridge, it is ensured that the cartridge is received and supported in an optimal way if the support is made of a relatively inelastic material. When the surface of the support facing towards the cartridge, including the one or more electrically connecting supports, in an axial cross section essentially correspond to the surface of the cartridge, when said cartridge is positioned in said support, it is ensured that the cartridge is received and supported in an optimal way if the support is made of a relatively elastic material. When said cartridge has an axial direction of symmetry, and said contact area consists of groups of identical and regularly spaced electrically conducting pads of width Wcp in the direction of adjacent pads, adjacent pads being separated by an electrically insulating area of width Diacp, and the following relations between said distances are fulfilled: Diacp>2*Tcl, and Wcp>Til+Tcl, it is ensured that the electrical states of adjacent (possibly abutted) predefined positions are not transferred to the same pad (Diacp>2*Tcl), and that at least one conducting layer contacts any given pad (Wcp>Til+Tcl). When said cartridge has an axial direction of symmetry, and said cartridge is provided with a multitude of rectangular, essentially parallel, identically sized information carrying areas of height Hica in the direction of a circumference of said axis of symmetry, said information carrying areas being spaced with equal mutual distance Dica along the periphery of the cartridge in the direction of a circumference of said axis of symmetry, and said supporting means comprise two rectangular, essentially parallel, identical electrically connecting supports of height Hctm in the direction perpendicular to the axis of symmetry of the cartridge, separated by an electrically insulating volume of width Dctm between the two electrically connecting supports, and the following relations between said distances are fulfilled: Hica<Dctm<2*Hica+Dica, and Hctm<Dica<2*Hctm+Dctm, it is ensured that the cartridge cannot be positioned in such a way that a given information carrying area has contact to two electrically connecting supports at the same time (Hica<Dctm). It is further ensured that the cartridge cannot be positioned in such a way that a given electrically connecting support has contact to two information carrying areas at the same time (Hctm<Dica). It is further ensured that the cartridge cannot be positioned in such a way that the electrically connecting supports fall entirely between two information carrying areas, in which case they would not have contact to any of the information carrying areas of the cartridge (Dica<2*Hctm+Dctm). It is further ensured that the cartridge cannot be positioned in such a way that two adjacent information carrying areas fall entirely between the electrically connecting supports, in which case the latter might not have contact to any of the information carrying areas of the cartridge (Dctm<2*Hica+Dica). When said information carrying areas of height Hica each consist of electrically conducting and electrically insulating rectangular patches provided at said predefined positions on said cartridge according to a binary representation of said item of information, said patches having a width Wpda abut each other, and the sum of the maximum thicknesses Tcl and Til of said alternating layers of electrically conducting and electrically insulating materials, respectively, constituting said electrically connecting supports, is less than the width Wpda of said patches, thus fulfilling the following relation between said distances: Wpda>Til+Tcl, it is ensured that each patch has contact to at least one of the conducting layers of an electrically connecting support when the cartridge is properly placed in the support. The use of a composite material, comprising a number of closely spaced mutually electrically insulated conductors embedded in an electrically insulating material is furthermore provided by the present invention. When said composite material is used at least for the partial support of a cartridge and for the transfer of an electronically readable information from information carrying areas on said cartridge to a contact area, a secure transfer of information with an increased density from a cartridge to an electronic circuit is provided. When said closely spaced mutually electrically insulated conductors embedded in an electrically insulating material are constituted by alternating layers of electrically conducting material and electrically insulating material, respectively, it is ensured that that a simple and flexible solution is provided. In a preferred embodiment said alternating layers of electrically conducting material and electrically insulating material, respectively are made of elastomeric materials. BRIEF DESCRIPTION OF THE DRAWINGS The invention will be explained more fully below in connection with a preferred embodiment and with reference to the drawings in which: FIG. 1 shows a cartridge containing an electrically readable information in the form of patterns of patches in the axial direction of the cartridge and a support according to the invention comprising one electrically connecting support for transferring the information to an electronic circuit, FIG. 2 shows a cartridge containing an electrically readable information in the form of patterns of patches in the axial direction of the cartridge and a support according to the invention comprising two electrically connecting supports for transferring the information to an electronic circuit, FIG. 3 shows a cartridge containing an electrically readable information in the form of ring patterns and a support according to the invention comprising one electrically connecting support for transferring the information to an electronic circuit, FIGS. 4.a–4.e show various ways of placing information carrying areas for holding electronically readable information on a cartridge, FIGS. 5.a–5.e show various ways of laying out the electrically conducting and electrically insulating areas in predefined positions within an information carrying area, implementing a binary representation of an item of information in its true and inverted form, FIGS. 6.a–6.c show various geometries of an electrically connecting support according to the invention, FIGS. 7.a and 7.b show an example of a cartridge and a support according to the invention comprising three electrically connecting supports made of elastic materials, and FIG. 8 shows geometries involved in reading an item of information provided a multitude of times along the periphery of a cartridge with a rotational symmetry by means of two electrically connecting supports. The figures are schematic and simplified for clarity, and they just show details, which are essential to the understanding of the invention, while other details are left out. In general, the reference numerals of a given drawing start with the number of that drawing, i.e. in FIG. 1, reference numerals typically have a 1 as the most significant digit (e.g. 1, 11, 125, 1250). This means on the other hand that functionally identical features occurring in different drawings have different reference numerals. DETAILED DESCRIPTION OF EMBODIMENTS FIGS. 1–3 show a cartridge containing an electrically readable information in the form of patterns of electrically conducting and electrically insulating areas and a support according to the invention comprising one or more electrically connecting supports for transferring the information to an electronic circuit. A support according to the invention has the combined function of receiving and mechanically supporting a part of the cartridge provided with information carrying areas AND of transferring the information from the these information carrying areas to an electronic circuit for further processing. With reference to FIGS. 1, 2 and 3 (having reference numerals starting with 1, 2 and 3, respectively), the cartridge 10, 20, 30, respectively, is only partially shown, as indicated by the ‘broken’ outline in the right-hand part of the cartridge. The cartridge possesses a rotational symmetry as indicated by the arrow 11, 21, 31, symbolizing the axis of symmetry. A label 12, 22, 32 containing information carrying areas laid out in the axial direction of the cartridge, is located on the outer surface at one axial end of the cartridge, where a lid 13, 23, 33, optionally in the form of a piston when the cartridge is a replaceable medication cartridge for a medication delivery device, provides a closure of the cartridge. The label 12, 22, 32 comprises an electrically conducting foil 120, 220, 320 having information carrying areas 121–127, 221–227, 325 extending in the axial direction of the cartridge. In FIGS. 1 and 2, a multitude of information carrying areas (121–127, 221–227, respectively, plus the ones situated on the hidden part of the surface) are evenly distributed on the surface of the cartridge in a radial direction (i.e. along the whole periphery encircling the axial direction of the cartridge). Each information carrying area, comprising patterns of electrically conducting and electrically insulating patches, thus only covers a limited radial sector of the surface. In FIG. 3, on the other hand, only one information carrying area 325 is indicated. This extends, however, along the whole periphery of the cartridge (i.e. the item of information is represented by closed rings 3251–3260 of electrically conducting and electrically insulating areas). Each of the information carrying areas 121–127, 221–227, 325 contains an item of information in the form of patterns of electrically conducting and electrically insulating areas. Each pattern represents an item of information in binary form. Each bit of information is represented by an electrically characteristic layer in a predefined position in the information carrying area. A binary one in a specific predefined position may be represented by an electrically conducting layer covering that predefined position, and a binary zero in a specific predefined position may be represented by an electrically insulating layer covering that predefined position. Alternatively, binary one may be represented by an insulating layer and binary zero by a conducting layer. Because the foils 120, 220, 320 in FIGS. 1–3 containing the information carrying areas 121–127, 221–227, 325 are electrically conducting, it is only necessary to apply an electrically insulating layer (e.g. a paint) to the predefined positions representing one of the a binary states (in this embodiment ‘zero’). In FIGS. 1–3, the cartridge is shown in a position just above the support 15, 25, 35, respectively, which, again for illustrative purposes, is shown just above a PCB with electronic components and connecting wires 16, 26, 36 containing pads 163, 263, 264, 363 with electrical connections, symbolically indicated by an arrow 162, 262, 362, to a processing unit 161, 261, 361, e.g. a micro-processor. The support consists of one or more electrically connecting supports 151, 251, 252, 351 embedded in an electrically insulating material 155, 255, 355. The electrically connecting supports comprise alternating layers of electrically conducting 1511, 2511, 3511 and electrically insulating 1512, 2512, 3512 layers of an elastomeric material, e.g. silicone rubber with the electrically conducting layer having a concentration of carbon black sufficient for electrical conduction. Each electrically conducting layer is electrically insulated from all other electrically conducting layers, so that each electrically conducting layer in effect represents an insulated conductor. By controlling the layer thicknesses, the maximum ‘density of information’ in the axial direction may be controlled. In the embodiments of FIGS. 1–3, the supports 15, 25, 35, including the electrically connecting supports 151, 251, 252, 351, are shown to be adapted to receive the curved shape of the part of the cartridge, where the information carrying areas 121–127, 221–227, 325 are located, by shaping them equivalently. This makes possible the use of non-elastic materials for the support, if convenient. In an operating configuration, the support is placed (and optionally fastened) on the PCB 160, 260, 360 so that electrical contact between the electrically connecting supports 151, 251, 252, 351 and the pads 163, 263, 264, 363 is ensured. The cartridge is positioned on the support so that electrical contact between one (FIGS. 1, 3) or two (FIG. 2) of the information carrying areas in their full axial lengths (i.e. involving all patches of a given information carrying area representing bits of information) and the electrically connecting supports is ensured. The geometrical dimensions of the patches, layers and pads and mutual distance between adjacent information carrying areas on the cartridge and corresponding electrically connecting supports are discussed below with reference to FIGS. 6 and 8. By applying a specific electric potential to the electrically conducting foil 120, 220, 320, this potential will be transferred from those predefined areas containing a conductive layer (i.e. in the present embodiment those predefined areas not being covered by an insulating layer) to the corresponding pads on the PCB. Via the connecting circuitry, a direct measure of the pattern of binary states of the information carrying area connected to the pads by a given electrically connecting support is presented on the inputs of the processing unit, possibly by appropriately terminating the inputs with pull-up or pull-down circuitry depending on the potential applied to the electrically conducting foil and the definition of the binary states. A specific part of the foil may be preferably reserved to the application of the electric potential (e.g. an area of the foil circumfering the cartridge and not occupied by information carrying areas, in FIG. 3 e.g. the part of the foil 320 not covered by information bits in predefined positions 321–330). The support 15, 25, 35 is only shown as having an axial length corresponding to the axial length of the corresponding information carrying areas (e.g. 125 in FIG. 1) but it may of course extend in both axial directions if appropriate for the application in question. Likewise the support is shown to cover a certain radial sector (less than 90 degrees), but it may of course cover any radial sector, including 360 degrees, if appropriate. In a preferred embodiment, the sector covered by the support is less than 180 degrees allowing a direct ‘vertical’ placement of the cartridge in the support (in opposition to the case of a 360 degrees support, where the cartridge has to be axially inserted). In FIGS. 1–3, the label 12, 22, 32 containing information carrying areas 121–127, 221–227, 325 is placed in one axial end of the cartridge 10, 20, 30 covering only the space occupied by the axial extent of the lid/piston 13, 23, 33 to ensure that a full view of the contents of the cartridge is available for inspection. Of course it might be located in any convenient position along the surface of the cartridge. Similarly, in FIGS. 1–3, the information carrying areas extend in the axial direction of the cartridge. They might as well extend in a radial direction (as discussed in connection with FIGS. 4 and 5) or in a direction there between (e.g. forming one or more helixes on the surface of the cartridge), if convenient, as long as the support, including the electrically connecting support(s), is adapted thereto. The electrical connections, schematically indicated by an arrow 162, 262, 362, connecting the pads 163, 263, 264, 363 with the processing unit 161, 261, 361 may be a one to one parallel set of electrical connections between each pad and a corresponding input on the processor 161, 261, 361, but it may also comprise a multiplexing or coding unit to reduce the number of necessary inputs to the processing unit. FIG. 1 shows a cartridge containing an electrically readable information in the form of patterns of patches in the axial direction of the cartridge and a support according to the invention comprising one electrically connecting support for transferring the information to an electronic circuit. The binary information contained in each of the information carrying areas 121, 122, 123, 124, 125, 126, 127 is the same as schematically indicated in the information carrying areas 125 and 126 in that the patterns of electrically conducting patches, exemplified by 1250, 1260 (no filling), and electrically insulating patches, exemplified by 1251, 1261 (hatched), are identical. The embodiment in FIG. 1 benefits from the rotational symmetry of the cartridge 10 and the label 12 with identical information carrying areas 121–127 equally distributed on the label along the periphery of the cartridge in that it only requires the user to position the cartridge properly in a radial direction (possibly involving a slight rotation of the cartridge around its axis of symmetry) to ensure that an electrical contact between one of the information carrying areas 121–127 and the electrically connecting support 151 is present (since the positioning in an axial direction 11 may be mechanically ensured by receiving means for the cartridge). The control of the cartridge being correctly positioned may be in the hands of the processing unit 161, which, if necessary, may indicate to the user via a display (not shown) or a voice interface that a corrective action is required, and which may block further use of the device, if the cartridge is not correctly positioned. FIG. 2 shows a cartridge containing an electrically readable information in the form of patterns of patches in the axial direction of the cartridge and a support according to the invention comprising two electrically connecting supports for transferring the information to an electronic circuit. In the embodiment in FIG. 2, the support 25 comprises two electrically connecting supports 251, 252 for simultaneously reading two items of information from two information carrying areas (e.g. 225, 226) on the cartridge 20. In FIG. 2 the evenly distributed information carrying areas 221–227 contain an item of information in a true binary form alternating with the information in its inverted form as indicated by the schematically illustrated patterns of electrically conducting 2261 and insulating 2251 patches in information carrying areas 225 and 226, respectively, one pattern being the inverse of the other. Apart from the advantages of the rotational symmetry as described above in connection with FIG. 1, the embodiment of FIG. 2 has the advantage of reading the information in a binary true and inverted form, which allows the safety in reading to be improved. Instead of providing the information in its true and inverted forms, the same binary representation of the item of information may be provided in all information carrying areas (as in FIG. 1) and read twice, which also allows an improved safety in reading. In the embodiment of FIG. 2, the electrically conducting ‘end’-patches 2250, 2260 may be used for connecting a power supply voltage. FIG. 3 shows a cartridge containing an electrically readable information in the form of ring patterns and a support according to the invention comprising one electrically connecting support for transferring the information to an electronic circuit. In the embodiment in FIG. 3, the support 35 comprises only one electrically connecting support 351 for reading an item of information from an information carrying area 325 on the cartridge 30. The information carrying area 325 extends along the whole periphery of the cartridge 30. A binary representation of the item of information is implemented by closed rings 3251–3260 of electrically conducting and electrically insulating areas in predefined positions. This embodiment has the advantage of having a full rotational symmetry so that the cartridge 30 may be (radially) arbitrarily oriented in the support. FIGS. 4.a–4.e show various ways of placing information carrying areas for holding electronically readable information on a cartridge. FIGS. 4.a–4.d show a cartridge 40 with an axis of rotational symmetry 41 and information carrying areas located at one axial end of the cartridge. FIG. 4.a shows two information carrying areas 401, 402 positioned side by side in a radial direction on the surface of the cartridge 40 (i.e. along the periphery perpendicular to the axis of symmetry). Each information carrying area covers only a limited radial sector of the surface. FIG. 4.b shows two information carrying areas 403, 404 positioned side by side in the axial direction 41 on the surface of the cartridge 40 (i.e. along the periphery parallel to the axis of symmetry). Each information carrying area 403, 404 covers only a limited radial sector of the surface. FIG. 4.c shows two information carrying areas 405, 406 positioned side by side in the axial direction on the surface of the cartridge 40 (i.e. along the periphery parallel to the axis of symmetry 41). Each information carrying area 405, 406 encircles the entire radial periphery of the cartridge. FIG. 4.d shows information carrying areas 410, 411, 412, 413, 414 positioned side by side, evenly distributed in a radial direction on the surface of the cartridge 40 (i.e. along the periphery perpendicular to the axis of symmetry). Each information carrying area covers only a limited radial sector of the surface. Information carrying areas 410, 411, 412, 413, 414 plus identical ones situated on the hidden part of the surface are evenly distributed on the surface of the cartridge in a radial direction, i.e. extending along the whole periphery encircling the axial direction of the cartridge. FIG. 4.e shows an information carrying area 415 extending along the major part of the axial length of the cartridge 40. The information carrying area is located within in a surface area 420 corresponding to a radial sector 421. In FIG. 4.e a single information carrying area is shown within the surface area 420. There might as well, however, be several information carrying areas located side by side in axial (cf. FIG. 4.b) or radial (cf. FIG. 4.a) direction. FIGS. 5.a–5.e show various ways of laying out the electrically conducting and electrically insulating areas in predefined positions within an information carrying area, implementing a binary representation of an item of information in its true and inverted form. In each of FIGS. 5.a–5.e two information carrying areas containing an item of information in a true and inverted binary form, respectively, are schematically shown. Each information carrying area has a rectangular shape defining a longitudinal direction as the direction defined by its longest side. A direction is also defined by the direction perpendicular to the face between two neighboring predefined positions each containing a specific bit of information. FIG. 5.a shows an embodiment with two information carrying areas 50, 51 located side by side in a direction perpendicular to the direction 505 defined by adjacent predefined positions. Each individual bit of information is implemented as a patch of electrically conducting 511 (no filling) or electrically insulating 501 (hatched) material located at a specific predefined position of the information carrying area. Neighboring patches abut each other. The structure of information carrying areas 50, 51 may e.g. be used in FIGS. 4.a. and 4.d. FIG. 5.b shows an embodiment with two information carrying areas 52, 53 located side by side in a direction perpendicular to the direction 525 defined by adjacent predefined positions. Each individual bit of information is implemented as a patch of electrically conducting 531 (no filling) or electrically insulating 521 (hatched) material located at a specific predefined position of the information carrying area. Neighboring patches are separated by a an ‘empty’ space 520, 530 of width equal to the width of each of the information carrying patches 521, 531. The ‘empty’ space may consist of an electrically conducting or insulating layer (as long as the pads on the PCB (cf. FIGS. 1–3) are correspondingly laid out). The structure of information carrying areas 52, 53 may e.g. be used in FIGS. 4.a. and 4.d. FIG. 5.c shows an embodiment with two information carrying areas 54, 55 located side by side in a direction 545 defined by adjacent predefined positions. Each individual bit of information is implemented as a patch of electrically conducting 551 (no filling) or electrically insulating 541 (hatched) material located at a specific predefined position of the information carrying area. Neighboring patches abut each other. The structure of information carrying areas 54, 55 may e.g. be used in FIGS. 4.a. and 4.d. FIG. 5.d shows an embodiment with two information carrying areas 56, 57 located side by side in a direction 565 defined by adjacent predefined positions. Each individual bit of information is implemented as a patch of electrically conducting 562, 571 (no filling) or electrically insulating 561, 572 (hatched) material located at a specific predefined position of the information carrying area. Neighboring patches abut each other. The structure of information carrying areas 56, 57 may e.g. be used in FIGS. 4.b. and 4.c. FIG. 5.e shows an embodiment with two information carrying areas 58, 59 located side by side in a direction perpendicular to the direction 585 defined by adjacent predefined positions. Each individual bit of information is implemented as a patch of electrically conducting 591 (no filling) or electrically insulating 581 (hatched) material located at a specific predefined position of the information carrying area. Neighboring patches abut each other. The structure of information carrying areas 58, 59 may e.g. be used in FIGS. 4.b. and 4.c. FIGS. 6.a–6.c show various geometries of an electrically connecting support according to the invention. Common for FIGS. 6.a–6.c is that the layer thicknesses 630, 640 of the electrically insulating and conducting layers, respectively, are exaggerated compared to the dimensions of the patches 61 on the information carrying areas and the pads 62 on the PCB. FIG. 6.a shows an embodiment of an electrically connecting support 60, where the thickness Til 630 of the insulating layer 63 is larger than the thickness Tcl 640 of the conducting layer 64. The patches 61 of the information carrying area are shown to be of equal width Wpda 610 and to abut each other. The pads 62 on the PCB are shown to have equal width Wcp 620 and to be evenly distributed with a distance Diacp 621 between each pad. FIG. 6.b shows an embodiment of an electrically connecting support 60, where the thickness Til of the insulating layer 63 is smaller than the thickness Tcl of the conducting layer 64. FIG. 6.c shows an embodiment of an electrically connecting support 60, where the thickness Til of the insulating layer 63 equals the thickness Tcl of the conducting layer 64. The relation Diacp>2*Tcl makes sure that the electrical states of adjacent information carrying patches on the cartridge are not transferred to the same pad in the contact area under the assumption that the border between adjacent patches is located at a position ‘corresponding to midway between two pads’. The fulfillment of the relation Wcp>Til+Tcl ensures that at least one conducting layer contacts any given pad. Correspondingly, the fulfillment of the relation Wpda>Til+Tcl ensures that each patch has contact to at least one of the conducting layers of an electrically connecting support, when the cartridge is properly placed in the support. In FIGS. 6.a–6.c, the information carrying patches on the cartridge are shown as abutted. This need not be the case. They may have any width Wpda as long as the relation Wpda>Til+Tcl is fulfilled to ensure that at least one conducting layer contacts any given information carrying patch. The relations reflect the minimum distances of pads and patches and between pads and thus for given layer thicknesses determine the information density (minimum width per bit) FIGS. 7.a–7.b show an example of a cartridge and a support according to the invention comprising three electrically connecting supports made of elastic materials. FIG. 7.a shows a cartridge 71 having an axis of rotational symmetry 72 being positioned just above a support 70 comprising three individual electrically connecting supports 701, 702, 703 ready for receiving the cartridge. The cartridge is provided with information carrying areas positioned on the cartridge along its radial periphery with a spacing corresponding to the geometry of the electrically connecting supports 701, 702, 703. The space between the electrically connecting supports may be filled with an isolating material (e.g. silicone rubber), not shown. In FIG. 7.b the cartridge 71 is positioned in the support 70 and fixed with a slight downwards pressure indicated by the arrow 73. The support including the electrically connecting supports 701, 702, 703 is made of elastic materials so that it conforms to the shape of the cartridge over the axial length of the support, when the cartridge is placed in the support. The three items of information that may be simultaneously read may be identical, in which case the redundancy may be used to improve the safety in reading (by a simple majority test or by more advanced error correcting techniques), or they may be different, in which case a larger amount of information may be read from the cartridge. FIG. 8 shows geometries involved in reading an item of information provided a multitude of times along the periphery of a cartridge with a rotational symmetry by means of two electrically connecting supports. In FIG. 8, the electrically connecting supports 81, 82 are shown in a position where they read information from information carrying areas 830, 840, respectively, and transfer the information to groups of pads 83, 84, respectively, on a PCB. The information carrying areas 810, 820, 830, 840, 850, 860 on a label 80 carry an item of information alternatingly in a binary true and inverted form as indicated by the schematically shown individual patches of equal width Wpda 89. The patches are either electrically conducting 8102 (no filling) or electrically insulating 8101 (hatched). The following geometric relations between the information carrying areas positioned on a cartridge and the electrically connecting supports 81, 82 of a support according to the invention for the cartridge: Hica<Dctm<2*Hica+Dica, and Hctm<Dica<2*Hctm+Dctm, where Hica=Height 87 of information carrying areas Dica=Distance 88 between information carrying areas Hctm=Height 85 of electrically connecting supports Dctm=Distance 86 between electrically connecting supports. Hica<Dctm ensures that the cartridge cannot be positioned in such a way that a given information carrying area has contact to two electrically connecting supports at the same time. Hctm<Dica ensures that the cartridge cannot be positioned in such a way that a given electrically connecting support has contact to two information carrying areas at the same time. Dica<2*Hctm+Dctm ensures that the cartridge cannot be positioned in such a way that the electrically connecting supports fall entirely between two information carrying areas, in which case they would not have contact to any of the information carrying areas of the cartridge. Dctm<2*Hica+Dica ensures that the cartridge cannot be positioned in such a way that two adjacent information carrying areas fall entirely between the electrically connecting supports, in which case the latter might not have contact to any of the information carrying areas of the cartridge. In a preferred embodiment, the following relation is fulfilled (in addition to the above mentioned relations between Dctm, Hctm, Dica, Hica), Dctm+Hctm=Dica+Hica, which ensures that the electrically connecting supports 81, 82 will have contact to two of the information carrying areas irrespective of the radial orientation of the cartridge in the support. Some preferred embodiments have been shown in the foregoing, but it should be stressed that the invention is not limited to these, but may be embodied in other ways within the subject-matter defined in the following claims. The invention may for example be applied to the electronic marking of cartridges for other purposes than medical, e.g. film cartridges, various cassettes containing media holding digital data or analog signals (e.g. representing software, data, film or music), etc., that are used in an electronic ‘environment’ (e.g. in a camera, computer, recorder, player, viewer, etc.). 09925792 novo nirdisk a/s USA B2 Utility Patent Grant (with pre-grant publication) issued on or after January 2, 2001. Open 235/487 Mar 31st, 2022 02:17PM Mar 31st, 2022 02:17PM Novo Nordisk Health Care Pharmaceuticals & Biotechnology

Request a demo to view additional historical data, and much more.

Make fast
queries

No longer rely on engineers to access data; build and share queries with intuitive tools, and derive insights in real time.

Bookmark queries with your team

Save and share custom queries using a private bookmarking feature, accessible to your whole team.

Be first to know with alerts

Scan data points across millions of companies and receive emails when relevant metrics cross critical thresholds.

Visualize data for quick insights

Create custom keyword-based word clouds, charts, and advanced visualizations to quickly analyze the data.

Map competitor locations

Analyze competitor presence and growth over time by overlaying store locations with the Nearby Competitor feature.

Add widgets to your dashboards

Access existing maps, charts, word clouds, and other visualizations to understand your data quickly. Or build custom widgets to view data just the way you want it.