专利摘要:
polypeptide having albumin and leptin binding domain, its use in the treatment of diseases, as well as a pharmaceutical composition comprising it. The present invention relates to polypeptides constructed which have an albumin binding domain and a first peptide hormone domain selected from a leptin, a leptin analogue or an active fragment thereof. the present invention further provides a pharmaceutical composition comprising said polypeptides and the use thereof in the treatment of diseases and disorders, including lipodystrophy, dyslipidemia, hyperlipidemia, obesity, hypothalamic amenorrhea, alzheimer's disease, leptin deficiency, fatty liver disease, diabetes ( including type i and type ii), non-alcoholic steatohepatitis (nash), non-alcoholic fatty liver disease (dhgna), metabolic syndrome x and huntington's disease.
公开号:BR112013007388B1
申请号:R112013007388-8
申请日:2011-09-28
公开日:2022-01-04
发明作者:Soumitra S. Ghosh;Mary Erickson;David C. Litzinger;Zijian Guo;Jonathan David Roth
申请人:Amylin Pharmaceuticals, Llc;Astrazeneca Pharmaceuticals Lp;
IPC主号:
专利说明:

REFERENCE TO RELATED ORDERS
[0001] The present application claims priority to US Application No. 61/387,402 filed September 28, 2010 and US Application No. 61/422,091 filed December 10, 2010, the disclosures of which are incorporated herein by reference. SEQUENCE LISTING
[0002] The present application contains a sequence listing, which was sent in ASCII format via EFS-Web and is incorporated herein by reference in its entirety. This ASCII copy, created on October 27, 2011, is called 1317WO1.txt and is 159,244 bytes in size. BACKGROUND OF THE INVENTION
[0003] The invention provides new compounds which have demonstrated biological activity. The compounds also demonstrate surprising and significant improvement in physical properties such as solubilization and stability.
[0004] The compounds of the invention are based on leptin sequences described in U.S. Application No. 61/387,402 and US Application No. 61/422,091. The compounds are surprisingly highly soluble and do not demonstrate a propensity to aggregate, unlike naturally occurring leptins. The physical properties of the compounds facilitate the preparation of pharmaceutically acceptable soluble formulations and compositions, also provided by the invention. Conditions amenable to such treatment include lipodystrophy, dyslipidemia, hyperlipidemia, excessive weight, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin deficiency, fatty liver disease, diabetes (including type I and type II), nonalcoholic steatohepatitis (Non Alcoholic Steato Hepatitis - NASH), Non-Alcoholic Fatty Liver Disease (NAFLD), Metabolic Syndrome X and Huntington's Disease or combinations thereof.
[0005] There remains a need to develop polypeptides useful in the diseases, conditions and metabolic disorders described above. Accordingly, it is an object of the present invention to provide new polypeptides useful for treating the above conditions and methods for producing and using the same.
[0006] Each Patent, Patent Application and Publication cited herein is incorporated herein by reference in its entirety and for all intents and purposes. BRIEF SUMMARY OF THE INVENTION
[0007] Chimeric polypeptide compounds having leptin biological activity in addition to enhanced physical properties are provided. The compounds are chimeric polypeptides which are based on a wild-type seal leptin polypeptide in which at least a 1-30 amino acid contiguous region of a wild-type seal leptin sequence has been replaced by a 1-amino acid contiguous region. 30 amino acids of a mature human leptin sequence.
[0008] In a first aspect, a chimeric polypeptide is provided, as described herein.
[0009] In another aspect, a method is provided for treating a disease or disorder in an individual in need of treatment. The method includes administering a chimeric polypeptide as described herein to the subject.
[00010] In yet another aspect, a pharmaceutical composition is provided which includes a chimeric polypeptide described herein in combination with a pharmaceutically acceptable excipient.
[00011] In yet another aspect, there are provided polynucleotides encoding the chimeric polypeptide and its intermediates, expression vectors carrying such polynucleotides, host cells expressing such polynucleotides, and means for their expression, synthesis, post-translational modification and isolation. BRIEF DESCRIPTION OF THE DRAWINGS
[00012] Figs. 1A-1C depict the effects of daily administration of the indicated chimeric polypeptides described herein on food intake and body weight change (% corrected for vehicle) when administered to female C57/B6 mice as described in Example 4. Figure 1A : food intake. Figure 1B: change in body weight (% vehicle-corrected). Figure 1C: dose-response curve.
[00013] Figs. 2A-2C depict the effects of daily administration of the indicated chimeric polypeptides described herein on food intake and body weight change (% corrected for vehicle) when administered to female C57/B6 mice as described in Example 5. Figure 2A : food intake. Figure 2B: change in body weight (% vehicle-corrected). Figure 2C: dose-response curve. DETAILED DESCRIPTION OF THE INVENTION I. Definitions
[00014] "Obesity" and "overweight" refer to mammals having a weight greater than normally expected and can be determined, for example, by physical appearance, body mass index (BMI) as known in the art, waist-to-hip circumference, skinfolds, waist circumference and the like. The Center for Disease Control and Prevention (CDC) defines overweight as an adult human being who has a BMI of 25 to 29.9 and defines obesity as an adult human being who has a BMI of 30 or greater. There are additional measures for determining obesity. For example, the CDC states that a person with a waist-to-hip ratio greater than 1.0 is overweight.
[00015] "Lean body mass" refers to the fat-free mass of the body, that is, total body weight minus body fat weight is lean body mass. Lean body mass can be measured using methods such as hydrostatic weighing, computerized cameras, dual energy X-ray absorptiometry, skin calipers, magnetic resonance imaging (MRI) and bioelectric impedance analysis (Bioelectric Impedance Analysis). - BIA), as known in the art.
[00016] "Mammal" refers to warm-blooded animals that generally have skin or fur, that give birth to their offspring, and that feed their offspring with milk. Mammals include humans, companion animals (eg, dogs, cats), farm animals (eg, cows, horses, sheep, pigs, goats), wild animals, and the like. In one embodiment, the mammal is a female. In one embodiment, the mammal is a female human. In one embodiment, the mammal is a dog or cat. In one embodiment, the mammal is a diabetic mammal, e.g., a human with type 2 diabetes. In one embodiment, the mammal is an obese diabetic mammal, e.g., an obese mammal with type 2 diabetes. The term " individual", in the context of the methods described herein, refers to a mammal.
[00017] "Fragment" in the context of polypeptides refers here, in the usual chemical sense, to a portion of a polypeptide. For example, a fragment can result from N-terminal deletion or C-terminal deletion of one or more residues of a parent polypeptide and/or a fragment can result from the internal deletion of one or more residues of a parent polypeptide. "Fragment", in the context of an antibody, refers to a portion of an antibody that can be linked to a biologically active molecule to modulate solubility, distribution within an individual, and so on. For example, leptin A200 described herein is a conjugate of an antibody Fc fragment with a leptin, as known in the art. See, for example, WO 98/28427 and US2007/002084. The term "parent" in the context of polypeptides refers, in the usual sense, to a polypeptide that serves as a reference structure before modification, for example, insertion, deletion and/or substitution.
[00018] "Analog", as used herein in the context of polypeptides, refers to a compound that has amino acid insertions, deletions and/or substitutions relative to a parent compound. An analog may have superior stability, solubility, efficacy, half-life and the like. In some embodiments, an analogue is a compound that has at least 50%, for example 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or even more sequence identity with the parent compound.
[00019] "Identity", "sequence identity" and the like, in the context of comparing two or more nucleic acid or polypeptide sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (i.e. about 50% identity, preferably 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90 %, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more of identity over a specified region when compared and aligned for maximum match over a window of comparison or designated region), as measured using a sequence comparison algorithm as known in the art, for example BLAST or BLAST 2.0. This definition includes sequences that have deletions and/or additions, as well as those that have substitutions, as well as naturally occurring variants, for example, polymorphic or allelic variants and man-made variants. In preferred algorithms, counting is done by gaps and the like, as known in the art. For sequence comparison, typically, a sequence acts as a reference sequence against which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequence coordinates are assigned if necessary, and sequence algorithm program parameters are assigned. Preferably, default program parameters can be used or alternative parameters can be assigned. The sequence comparison algorithm then calculates the percentage of sequence identities for the test sequences relative to the reference sequence based on the program parameters. Optimal alignment of sequences for comparison can be performed, for example, by the local homology algorithm of Smith & Waterman, Adv. Appl. Math., 2: 482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol., 48: 443 (1970), by the similarity search method of Pearson & Lipman, Proc. Nat'l. academy Sci. USA 85: 2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA and TFASTA in the Wisconsin Genetics software package, Genetics Computer Group, 575 Science Dr., Madison, Wis) or manual alignment and visual inspection. See, for example, Current Protocols in Molecular Biology (Ausubel et al., Eds. 1995 supplement)). Preferred examples of algorithms that are suitable for determining percent sequence identity and sequence similarity include the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al., Nucl. Acids Res., 25: 3389-3402 (1977) and Altschul et al., J. Mol. Biol. 215: 403-410 (1990). BLAST and BLAST 2.0 are used, as known in the art, to determine percent sequence identity for the nucleic acids and proteins of the present invention. Software for performing BLAST analysis is publicly available through the National Center for Biotechnology Information website. This algorithm first involves identifying High Scoring Sequence Pairs (HSPs) by identifying short words of length W in the query string, which match or satisfy some threshold T-score of positive value when aligned with a word. of the same length in a database string. T is referred to as the neighbor word score threshold (Altschul et al., Id.). These initial neighbor word "hits" act as seeds to initiate searches to find longer HSPs containing them. The word "hits" are extended in both directions along each sequence, as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for example, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for non-matching residues; always < 0). For amino acid sequences, a score matrix is used to calculate the cumulative score. Extension of word hits in each direction is stopped when: the cumulative alignment score drops by the amount X from its maximum reached value; the cumulative score goes to zero or below because one or more negative score residue alignments accumulate or the end of any sequence is reached. The W, T and X parameters in the BLAST algorithm determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) defaults to a word length (W) of 11, an expectation (E) of 10, M = 5, N = -4, and a comparison of both strands. For amino acid sequences, the BLASTP program defaults to a word length of 3 and expectation (E) of 10 and the BLOSUM62 scoring matrix (see Henikoff & Henikoff, Proc. Nat. Acad. Sci. USA, 89: 10915 (1989)) alignments (B) of 50, expectation (E) of 10, M = 5, N = -4 and a comparison of both strands.
[00020] The term "about", in the context of a numerical value, refers to +/- 10% of the numerical value, unless expressly stated otherwise.
[00021] The terms "peptide" and "polypeptide", in the context of components of the chimeric polypeptides described herein, are synonymous.
[00022] Leptins. "Leptins" and "a leptin" mean: leptins, active leptin fragments, leptin analogues and leptin derivatives; and a leptin, an active leptin fragment, a leptin analogue and a leptin derivative; respectively. Accordingly, unless otherwise indicated, reference to "leptins" is intended to mean leptins, active leptin fragments, leptin analogues and leptin derivatives as described herein. Similarly, unless otherwise indicated, reference to "a leptin" is intended to encompass a leptin, an active leptin fragment, a leptin analogue and a leptin derivative, as described herein. Such exemplary leptins which may be employed in the design, preparation and use of the chimeric polypeptides described in the present invention include those which trigger one or more biological responses known in the art to be elicited when leptins are administered to volunteers (see, for example, Applications US Patent Published Nos US 2007/0020284 and US 2008/0207512 , US Patent Nos 6,309,853 , 7,183,254 and US PCT Published Application Nos WO 96/005309 , WO 98/28427 and WO 2009/064298 ), such as: reduced food intake, reduced body weight, reduced weight gain, induction of satiety, reduced caloric availability, reduced caloric efficiency, reduced metabolic plateau, increased insulin sensitivity, decreased hyperlipidemia, correction of dyslipidemia, reduction of hypertriglyceridemia, improvement of obesity, improvement of overweight, improvement of diabetes mellitus (including type I diabetes, type II diabetes and diabetes mellitus es gestation), improvement of insulin resistance, improvement of lipodystrophy conditions associated therewith, as well as other biological responses known in the art to be triggered upon administration of a leptin (see, for example, U;S. Published Nos. US 2007/0020284 and US 2008/0207512 , U.S. Patent Nos. 6,309,853 , 7,183,254 and U.S. PCT Applications Published Nos WO 96/005309 , WO 98/28427 and WO 2009/064298 ).
[00023] Leptins include, but are not limited to, the compounds described in US Patent Nos. US 5,594,101, US 5,851,995, US 5,691,309, US 5,580,954, US 5,554,727, US 5,552,523, US 5,559. 208 , US 5,756,461 , US A 6,309,853 , US Patent Application Published No. US 2007/0020284 and PCT Applications Published Nos WO 96/23517 , WO 96/005309 , WO 98/28427 , WO 2004/039832 , WO 98 /55139, WO 98/12224 and WO 97/02004, each of which is incorporated herein in full and for all intents and purposes. Methods for assaying leptin activities and biological responses in vitro and in vivo, including satiety inhibiting activity, food intake inhibiting activity, and weight loss activity, are known in the art and are described herein and also in the mentioned references. above and other references cited here.
[00024] leptins, leptin analogs, leptin fragments and active derivatives include the following representative leptin: leptin Mature Murine: VPIQKVQDDTKTLIKTIVTRINDISHT-Xaa SVSSKQKVTGLDFIPGLHPILTLS KMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLPQ ASGLETLESLGGVLEASGYSTEVVALSRLQGSLQDMLQQLDLSPGC, wherein Xaa at position 28 is Q or absent (SEQ ID NO : 1). Mature murine leptin form 1: VPIQKVQDDTKTLIKTIVTRINDISHTQSVSAKQRVTGLDFIPGLHPILSL SKMDQTLAVYQQVLTSLPSQNVLQIANDLENLRDLLHLLAFSKSCSLP QTSGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDVSPEC (SEQ ID NO: 2). Mature murine leptin form 2: VPIQKVQDDTKTLIKTIVTRINDISHTSVSAKQRVTGLDFIPGLHPILSLS KMDQTLAVYQQVLTSLPSQNVLQIANDLENLRDLLHLLAFSKSCSLPQ TSGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDVSPEC (SEQ ID NO: 3). Mature murine leptins with N-terminal methionine: MVPIQKVQDDTKTLIKTIVTRINDISHT-Xaa- SVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQILTSMPSRNVIQIS NDLENLRDLLHVLAFSKSCHLPQASGLETLESLGGVLEASGYSTEVV ALSRLQGSLQDMLQQLDLSPGC, where Xaa at position 29 is either NO or 4Q. Mature murine leptin form 1 with N-terminal methionine: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSAKQRVTGLDFIPGLHPIL SLSKMDQTLAVYQQVLTSLPSQNVLQIANDLENLRDLLHLLAFSKSCS LPQTSGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDVSP EC (SEQ ID NO: 5). Mature murine leptin form 2 with N-terminal methionine: MVPIQKVQDDTKTLIKTIVTRINDISHTSVSAKQRVTGLDFIPGLHPILSL SKMDQTLAVYQQVLTSLPSQNVLQIANDLENLRDLLHLLAFSKSCSLP QTSGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDVSPEC (SEQ ID NO: 6). Porcine Mature Leptin: VPIWRVQDDTKTLIKTIVTRISDISHMQSVSSKQRVTGLDFIPGLHPVL SLSKMDQTLAIYQQILTSLPSRNVIQISNDLENLRDLLHLLASSKSCPLP QARALETLESLGGVLEASLYSTEVVALSRLQGALQDMLRQLDLSPGC (SEQ ID NO: 7). Mature porcine leptin with N-terminal methionine: MVPIWRVQDDTKTLIKTIVTRISDISHMQSVSSKQRVTGLDFIPGLHPV LSLSKMDQTLAIYQQILTSLPSRNVIQISNDLENLRDLLHLLASSKSCPL PQARALETLESLGGVLEASLYSTEVVALSRLQGALQDMLRQLDLSPG C (SEQ ID NO: 8). Mature bovine leptin: VPICKVQDDTKTLIKTIVTRINDISHT-Xaa- SVSSKQRVTGLDFIPGLHPLLSL SKMDQTLAIYQQILTSLPSRNVVQISNDLENLRDLLHLLAASKSCPLPQ VRALESLESLGVVLEASLYSTEVVALSRLQGSLQDMLRQLDLSPGC, where Xaa at position 28 is either Q or is absent (SEQ ID NO: 9). N-terminal methionine mature bovine leptin: MVPICKVQDDTKTLIKTIVTRINDISHT-Xaa- SVSSKQRVTGLDFIPGLHPL LSLSKMDQTLAIYQQILTSLPSRNVVQISNDLENLRDLLHLLAASKSCP LPQVRALESLESLGVVLEASLYSTEVVALSRLQGSLQDMLRQLDLSP GC, where Xaa at position 29 is either NO ID: 10 (SEQ). full length human leptin unprocessed (i.e., including signal sequence 21 residues of N-terminal): MHWGTLCGFLWLWPYLFYVQAVPIQKVQDDTKTLIKTIVTRINDISHT QSVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQILTSMPSRNVIQI SNDLENLRDLLHVLAFSKSCHLPWASGLETLDSLGGVLEASGYSTEV VALSRLQGSLQ DMLWQLDLSPGC (SEQ ID NO: 11) mature human leptin (with signal sequence N-terminal 21 amino acids removed): VPIQKVQDDTKTLIKTIVTRINDISH-Xaa-Xaa- SVSSKQKVTGLDFIPGLHPILT LSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHL PWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSP GC, where: Xaa at position 27 is T or A ; and Xaa at position 28 is Q or is absent (SEQ ID NO: 12). Mature human leptin with N-terminal methionine: MVPIQKVQDDTKTLIKTIVTRINDISH-Xaa-Xaa- SVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSP GC, where: Xaa at position 28 is; and Xaa at position 29 is Q or is absent (SEQ ID NO: 13). Rhesus Mature Leptin: VPIQKVQSDTKTLIKTIVTRINDISHTQSVSSKQRVTGLDFIPGLHPVLT LSQMDQTLAIYQQILINLPSRNVIQISNDLENLRDLLHLLAFSKSCHLPL ASGLETLESLGDVLEASLYSTEVVALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO: 14). Mature Rhesus leptin with N-terminal methionine: MVPIQKVQSDTKTLIKTIVTRINDISHTQSVSSKQRVTGLDFIPGLHPVL TLSQMDQTLAIYQQILINLPSRNVIQISNDLENLRDLLHLLAFSKSCHLP LASGLETLESLGDVLEASLYSTEVVALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO: 15). Mature mouse leptin: VPIHKVQDDTKTLIKTIVTRINDISHTQSVSARQRVTGLDFIPGLHPILSL SKMDQTLAVYQQILTSLPSQNVLQIAHDLENLRDLLHLLAFSKSCSLP QTRGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDLSPEC (SEQ ID NO: 16). N-terminal methionine mature rat leptin: MVPIHKVQDDTKTLIKTIVTRINDISHTQSVSARQRVTGLDFIPGLHPIL SLSKMDQTLAVYQQILTSLPSQNVLQIAHDLENLRDLLHLLAFSKSCSL PQTRGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDLSPE C (SEQ ID NO: 17). Mature platypus leptin: The sequence of mature platypus leptin follows: ISIEKIQADTKTLTKTIITRIIQLSTQNGVSTDQRVSGLDFIPGNQQFQNL ADMDQTLAVYQQILSSLPMPDRTQISNDLENLRSLFALLATLKNCPFT RSDGLDTMEIWGGIVEESLYSTEVVTLDRLRKSLKNIEKQLDHIQG (SEQ ID NO: 18). Leptin full length platypus unprocessed (i.e., include signal sequence 21 residues of N-terminal): Following a sequence of leptin platypus total length, including an N-terminal signal sequence of 21 residues MRCILLYGFLCVWQHLYYSHPISIEKIQADTKTLTKTIITRIIQLSTQNGV STDQRVSGLDFIPGNQQFQNLADMDQTLAVYQQILSSLPMPDRTQIS NDLENLRSLFALLATLKNCPFTRSDGLDTMEIWGGIVEESLYSTEVVT LDRLRKSLKNIEKQLDHIQG ( SEQ ID NO: 19). Mature human leptin form 1: VPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPG C (SEQ ID NO: 20). Mature human leptin form 2: VPIQKVQDDTKTLIKTIVTRINDISHAQSVSSKQKVTGLDFIPGLHPILTL SKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPG C (SEQ ID NO: 21). Mature human leptin form 3: VPIQKVQDDTKTLIKTIVTRINDISHTSVSSKQKVTGLDFIPGLHPILTLS KMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLPW ASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO: 22). Mature human leptin form 4: VPIQKVQDDTKTLIKTIVTRINDISHASVSSKQKVTGLDFIPGLHPILTLS KMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLPW ASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO: 23). N-terminal methionine mature human leptin form 1 (also known as Metreleptin or A100): MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQ IDLS PGC (2SE). N-terminal methionine mature human leptin form 2: MVPIQKVQDDTKTLIKTIVTRINDISHAQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLS PGC (SEQ ID NO: 25). N-terminal methionine mature human leptin form 3: MVPIQKVQDDTKTLIKTIVTRINDISHTSVSSKQKVTGLDFIPGLHPILTL SKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPG C (SEQ ID NO: 26). N-terminal methionine mature human leptin form 4: MVPIQKVQDDTKTLIKTIVTRINDISHASVSSKQKVTGLDFIPGLHPILTL SKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPG C (SEQ ID NO: 27). Seal Leptin: PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 28). Seal leptin with amino acids 71-92 replaced by metreleptin amino acids 73-94 (helix 3), respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDM2SEVRTL: IDQRNPGC (IDLRQLD: NOPGRNC). Seal leptin with amino acids 30 and 71-92 replaced by metreleptin amino acids 32 and 73-94 (helix 3), respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSQRL (NOLRQQRNCQ0RNCID). N-terminal methionine seal leptin: MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 31). Seal leptin with N-terminal methionine and with amino acids 7192 substituted for metreleptin amino acids 73-94 (helix 3), respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSQ2 RARGSDTIKGLGNVLRASVHSTEVVALSQ2 ). Leptin seal with N-terminal methionine and amino acids 30 and 71-92 replaced by amino acids 32 and 73-94 (helix 3) metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 33). Leptin A200: Leptin A200 is a product of Fc antibody condensation with leptin, as known in the art. See, for example, Lo et al., 2005, Protein Eng. Design & Selection, 18: 1-10. The A200 amino acid sequence is as follows: MDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNST YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP QVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKT TPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGKVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFI PGLHPILTLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLA FSKSCHLPWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDML WQLDLSPGC (SEQ ID NO: 34) Leptin A300: Leptin A300 metreleptin with W101Q and W139Q substitutions (1Met N-terminal counted as residue 1): MVPIQKVQDDTKTLI KTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQI LTSM PSRNVIQISNDLENLRDLLHVLAFSKSCHLPQASGLETLDSLGGVLEA SGYSTEVVALSRLQGSLQDMLQQLDLSPGC (SEQ ID NO: 35).
[00025] A400 Leptin: Leptin is metreleptin A400 with the serine residue at position 78 replaced by a cysteine residue, as shown below: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDF IPGLHPILTLSKMDQTLAVYQQILTSMPSRNVIQICNDLENLRDLLHVLA FSKSCHLPWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDML WQLDLSPGC (SEQ ID NO: 36); to which a 20 kilodalton (kDa) PEG moiety was attached via the cysteine residue at position 78.
[00026] Leptin A500: Research by a number of investigators, including the inventors, has focused on the aggregation effects of residue replacement on leptin. See, for example, Ricci et al., 2006. “Mutational Approach to Improve Physical Stability of Protein Therapeutics Susceptible to Aggregation: Role of Altered Conformation in Irreversible Precipitation”, Book Chapter. in: Misbehaving Proteins: Protein (Mis)Folding, Aggregation, and Stability, Murphy RM, Tsai AM, Eds., New York. Springer. pages 331-350, which is incorporated herein by reference and for all purposes. Consequently, leptin A500 with the following sequence was used in certain compounds and methods described here: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLEFIPGLHPILT LSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHL PQASGLETLESLGGVLEASGYSTEVVALSRLQGSLQDMLQQLDLSP GC (SEQ ID NO: 37).
[00027] Variants A100 Leptin: Leptin A100 variants following with the following amino acid substitutions: D41E, H98S, W101Q, D109E, G113E, M137I, W139Q and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLEFIPGLHPILT LSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQASGLETLESLGEVLEASGYSTEVVALSRLQGSLQDILQQLDLSPE C (SEQ ID NO: 38). H98S, W101Q, A102T, G113E, M137I, W139Q and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCQSL PQASGLETLQDSLGGVLEASGYSTEVVALSRLQGSLQ: NO. H98S, W101Q, G113E, M137I, W139Q and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDIL (SEQLDLSPE C4QQLDLSPE: NO). W101Q, G113E, M137I, W139Q and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDILQQLDLSPE ID:4SEQ1). H98S, W101Q, M137I, W139Q and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQASGLETLSGGVLEASGYSTEVVALSRLQGSLQDILQQLDLS PEC42. W101Q, G113E, M137I, W139Q, L143V and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDILQSEQLDV (ID3QGSLQDILQSE3V). H98S, W101Q, A102T, M137I, W139Q and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQTSGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDIL4QQLD NOPEC: H98S, W101Q, D109E, G113E and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQASGLETLESLGEVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPE C (SEQ ID). W101Q, M137I, W139Q and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDILQQLDLSP EC (SEQ ID NO: 46). W101Q, M137I, W139Q, L143V and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDILQQLDVSP EC (SEQ). H98S, W101Q, A102T, M137I, W139Q, L143V and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQTSGLETLDSLGGVLEASGYSTEVVALSRLQGSLQ: NO. H98S, W101Q, A102T, G113E and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQTSGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPE C (SEQ ID). W101Q, G113E and W139Q: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDMLQQLDLSP GC (SEQ ID NO: 50). W101Q, G113E, W139Q and G146E: MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDMLQQLDLSP EC (SEQ ID NO: 51). I. Chimeric Polypeptides
[00028] In one aspect of the present description, a series of chimeric polypeptides are described. These chimeric polypeptides are based on a wild-type seal leptin polypeptide in which at least one contiguous 1-30 amino acid region of a wild-type seal leptin sequence has been replaced by a contiguous 1-30 amino acid region of a mature human leptin sequence. A wild-type seal leptin sequence includes the wild-type seal leptin sequence (SEQ ID NO: 28) and the seal leptin sequence with an N-terminal wild-type methionine (SEQ ID NO: 31). A mature human leptin sequence useful for chimerization with wild-type seal leptin, as provided herein, includes the sequences described above below: mature human leptins (SEQ ID NO: 12), N-terminal methionine mature human leptins (SEQ ID NO: 13), mature human leptin form 1 (SEQ ID NO: 20), mature human leptin form 2 (SEQ ID NO: 21), mature human leptin form 3 (SEQ ID NO: 22), form 4 mature human leptin (SEQ ID NO: 23), N-terminal methionine mature human leptin form 1 (Metreleptin or A100, SEQ ID NO: 24), N-terminal methionine mature human leptin form 2 (SEQ ID NO : 25), mature human leptin form 3 with N-terminal methionine (SEQ ID NO: 26), mature human leptin form 4 with N-terminal methionine (SEQ ID NO: 27), A200 (SEQ ID NO: 34), A300 (SEQ ID NO: 35), A400 (SEQ ID NO: 36), A500 (SEQ ID NO: 37) and variants of A100 (SEQ ID NO: 38-51). In some embodiments, a series of chimeric polypeptides are described in which at least one contiguous 1-30 amino acid region of a wild-type seal leptin sequence (SEQ ID NO. 28 or SEQ ID NO: 31) has been substituted. by a contiguous region of 1-30 amino acids of A100 (SEQ ID NO. 24).
[00029] In any of the chimeric polypeptides described, a contiguous region of 1-30 amino acids may comprise any naturally occurring or non-naturally occurring amino acid. Any combination of amino acids can be used without restriction. That is, two or more amino acids in a contiguous region can be replaced by a naturally occurring amino acid, a non-naturally occurring amino acid, a conservative substitution, a non-conservative substitution, or any combination thereof.
[00030] The chimeric polypeptides described here have demonstrated biological activity in addition to enhanced physical properties. For example, human-seal chimeric polypeptides show leptin activity in vitro and in vivo. The chimeric polypeptides also show enhanced stability and solubility compared to the mature human leptin polypeptides which are used to derive the sequences, as shown by the Examples.
[00031] The term "leptin activity" includes leptin binding activity and leptin functional activity. Those skilled in the art will recognize leptin analog compounds with leptin activity using assays suitable for measuring leptin binding or leptin functional activity. Leptin analog compounds can have an IC50 of about 200 nM or less, about 100 nM or less, or about 50 nM or less, or about 5 nM or less, or about 1 nM or less, in a binding assay. leptin, such as the one described here. The term "IC50" refers, in the common sense, to half the maximum inhibitory concentration of a compound that inhibits a biological or biochemical function. Accordingly, in the context of receptor binding studies, IC50 refers to the concentration of test compound which compensates for half of a known binder of a specified receptor. Leptin analog compounds can have an EC50 of about 20 nM or less, about 10 nM or less, about 5 nM or less, about 1 nM or less, or about 0.1 nM or less, in a functional assay of leptin, such as the one described here. The term "EC50" refers, in the common sense, to the effective concentration of a compound which induces half the response between a baseline response and the maximal response, as is known in the art. A. Chimeric Polypeptides That Incorporate Human Helix 1
[00032] The Helix 1 region of a mature human leptin polypeptide spans a contiguous region of 20 amino acids. Helix 1 and Helix 3 are antisense helices that form part of the Binding Site II of leptin to its receptor. This site interacts with the Cytokine Receptor Homology (CRH) domain of the leptin receptor and is believed to be the major receptor binding site, but not involved in receptor activation. See, for example, Peelman et al., 2004, J. Biol. Chem. 279: 41038.
[00033] In one aspect, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with a helix 1 sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 3-22 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 5-24 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 52: Leptin seal with 3-22 amino acids substituted by amino acids 5-24 (Helix 1) metreleptin, respectively: PIQKVQDDTKTLIKTIVTRINDISPPQGVCSRPRVAGLDFIP RVQSVRTLSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLA SAKSCPVPRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLR QLDRNPGC (SEQ ID NO: 52).
[00034] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 3-22 of SEQ ID NO: 31 have been replaced with a contiguous region spanning amino acids at positions 5-24 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 53: Leptin seal with N-terminal methionine and amino acids 3-22 replaced by amino acids 5-24 (Helix 1) metreleptin, respectively: MPIQKVQDDTKTLIKTIVTRINDISPPQGVCSRPRVAGLDFIPRVQSVR TLSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 53). B. Chimeric Polypeptides That Incorporate Human Helix 2
[00035] The Helix 2 region of a mature human leptin polypeptide spans a region of 16 contiguous amino acids. This helix is embedded in the 4 helix bundle, as described in the original crystal structure article by Zhang et al. (Nature 1997 387: 206).
[00036] In one aspect, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with a helix 2 sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 50-65 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 52-67 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 54: Leptin seal with amino acids 50-65 replaced by amino acids 52-67 (helix 2) metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SKMDQTLAVYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 54 ).
[00037] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 50-65 of SEQ ID NO: 31 have been replaced with a contiguous region spanning the amino acids at positions 52-67 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 55: Leptin seal with N-terminal methionine and amino acids 50-65 replaced by amino acids 52-67 (helix 2) metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSKMDQTLAVYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 55). C. Chimeric Polypeptides That Incorporate Human Helix 3
[00038] The helix 3 region of a mature human leptin polypeptide spans a contiguous region of 22 amino acids. Helix 3 and Helix 1 are antisense helices that form part of the Binding Site II of leptin to its receptor. This site interacts with the Cytokine Receptor Homology (CRH) domain of the leptin receptor and is believed to be the major receptor binding site, but not involved in receptor activation. See, for example, Peelman et al., 2004, J. Biol. Chem. 279: 41038.
[00039] In one aspect, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with a helix 3 sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 71-92 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 73-94 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 29: Leptin seal with amino acids 71-92 replaced by amino acids 73-94 (helix 3) metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 29 ).
[00040] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 71-92 of SEQ ID NO: 31 have been replaced with a contiguous region spanning amino acids at positions 73-94 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 32: Leptin seal with N-terminal methionine and amino acids 71-92 replaced by amino acids 73-94 (helix 3) metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 32). D. Chimeric Polypeptides That Incorporate a Human Helix 4
[00041] The Helix 4 region of a mature human leptin polypeptide spans a contiguous region of 22 amino acids. Helix 4 is believed to form part of the Binding Site I and Binding Site III of leptin, both of which are important for receptor activation. See, for example, Peelman et al., 2004, J. Biol. Chem. 279: 41038.
[00042] In one aspect, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with a helix 4 sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 120-141 of SEQ ID NO: 28 have been replaced by a contiguous region spanning the amino acids at positions 122-143 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 56: Leptin seal with amino acids replaced by amino acids 120-141 122-143 (helix 4) metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 56).
[00043] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 120-141 of SEQ ID NO: 31 have been replaced with a contiguous region spanning the amino acids at positions 122143 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 57: Leptin seal with N-terminal methionine and amino acids 120-141 replaced by amino acids 122-143 (helix 4) metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 57). E. Chimeric Polypeptides That Incorporate the Human AB Loop
[00044] The AB loop region of a mature human leptin polypeptide spans a contiguous region of 27 amino acids. The AB loop is believed to form part of Binding Site III as well as a small portion of Binding Site I of leptin. See, for example, Peelman et al., 2004, J. Biol. Chem. 279: 41038. This region also contains the absolutely conserved motif GLDFIP (SEQ ID NO: 164).
[00045] In one aspect, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with an AB loop sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 23-49 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 25-51 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 58: Leptin seal with amino acids replaced by amino acids 23-49 25-51 (AB loop) of metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 58 ).
[00046] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 23-49 of SEQ ID NO: 31 have been replaced with a contiguous region spanning the amino acids at positions 25-51 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 59: Leptin seal with N-terminal methionine and amino acids 23-49 replaced by amino acids 25-51 (AB loop) of metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 59). F. Chimeric Polypeptides That Incorporate Human Loop 3-4
[00047] The Loop 3-4 region of a mature human leptin polypeptide spans a contiguous region of 27 amino acids. The 3-4 loop is believed to contain a part of the Binding Site III of leptin to its receptor. See, for example, Peelman et al., 2004, J. Biol. Chem. 279: 41038.
[00048] In one aspect, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with a 3-4 loop sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 93-119 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 95-121 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 60: Leptin seal with 93-119 amino acids replaced by amino acids 95-121 (3-4 loop) of metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 60).
[00049] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 93-119 of SEQ ID NO: 31 have been replaced with a contiguous region spanning the amino acids at positions 95-121 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprising the sequence described in SEQ ID NO: 61: Leptin seal with N-terminal methionine and amino acids 93-119 replaced by amino acids 95-121 (loop 34) metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 61). G. Combined Chimeric Polypeptides
[00050] In another aspect of the present description, a series of combined chimeric polypeptides are described. These combined chimeric polypeptides are based on a wild-type seal leptin polypeptide wherein two or more contiguous 1-30 amino acid regions of a wild-type seal leptin sequence (e.g., SEQ ID NO: 28 or SEQ ID NO: 31) have been replaced, in each region, by a contiguous region of 1-30 amino acids from a mature human leptin sequence. Combined chimeric polypeptides can be chimeric to demonstrate enhanced physical properties compared to mature human leptin polypeptides which are used to derive the sequences while retaining the biological activity of human leptin.
[00051] In some embodiments, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with a helix 1 sequence and a helix 3 sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 3-22 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 5-24 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 71-92 of SEQ ID NO: 28 have been replaced by a region contiguous spanning amino acids at positions 73-94 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 62: Seal leptin with amino acids 3-22 replaced by metreleptin amino acids 5-24 (helix 1) and amino acids 71-92 replaced by amino acids 73-94 ( helix 3) of metreleptin, respectively:PIQKVQDDTKTLIKTIVTRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 62)
[00052] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 3-22 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 5-24 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 71-92 of SEQ ID NO:31 have been replaced by a contiguous region spanning amino acids at positions 73-94 of A100 (SEQ ID NO:24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 63: Seal leptin with N-terminal methionine and with amino acids 3-22 substituted by amino acids 5-24 (helix 1) of metreleptin and amino acids 72-93 substituted by metreleptin amino acids 73-94 (helix 3), respectively: MPIQKVQDDTKTLIKTIVTRINDISPPQGVCSRPRVAGLDFIPRVQSVR TLSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 63)
[00053] In some embodiments, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with an incorporated helix 3 sequence and an AB loop sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 71-92 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 73-94 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 23-49 of SEQ ID NO: 28 have been replaced by a region contiguous spanning amino acids at positions 25-51 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 64: Seal leptin with amino acids 71-92 replaced by metreleptin amino acids 73-94 (helix 3) and with amino acids 23-49 replaced by amino acids 25-51 (AB loop) of metreleptin, respectively:PIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPRA RGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 64)
[00054] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 71-92 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 73-94 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 23-49 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 25-51 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 65: Seal leptin with N-terminal methionine and with amino acids 71-92 replaced by amino acids 73-94 (helix 3) of metreleptin and with amino acids 23-49 substituted by metreleptin amino acids 25-51 (AB loop), respectively: MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 65)
[00055] In some embodiments, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with an incorporated helix 3 sequence and a 3-4 loop sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 71-92 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 73-94 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 93-119 of SEQ ID NO: 28 have been replaced by a region contiguous sequence spanning amino acids at positions 95-121 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 66: Seal leptin with amino acids 71-92 replaced by metreleptin amino acids 73-94 (helix 3) and with amino acids 93-119 replaced by amino acids 95-121 (loop 34) of metreleptin, respectively:PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 66)
[00056] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 71-92 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 73-94 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 93-119 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 95-121 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 67: N-terminal methionine seal leptin, with amino acids 71-92 replaced by metreleptin amino acids 73-94 (helix 3) and with amino acids 93-119 substituted by metreleptin amino acids 95-121 (loop 3-4), respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 67)
[00057] In some embodiments, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with an incorporated AB loop sequence and a helix 4 sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 23-49 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 25-51 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 120-141 of SEQ ID NO: 28 have been substituted with a region contiguous sequence spanning amino acids at positions 122-143 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 68: Seal leptin with amino acids 23-49 replaced by metreleptin amino acids 25-51 (AB loop) and with amino acids 120-141 replaced by amino acids 122-143 (helix 4) of metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPGC (SEQ ID NO: 68)
[00058] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 23-49 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 25-51 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 120-141 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 122-143 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 69: Seal leptin with N-terminal methionine, with amino acids 23-49 replaced by amino acids 25-51 (AB loop) of metreleptin and with amino acids 120-141 substituted by metreleptin amino acids 122-143 (helix 4), respectively: MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 69)
[00059] In some embodiments, the present disclosure relates to chimeric polypeptides that are based on wild-type seal leptin with an incorporated AB loop sequence and a 3-4 loop sequence incorporated from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 23-49 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 25-51 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 93-119 of SEQ ID NO: 28 have been replaced by a region contiguous sequence spanning amino acids at positions 95-121 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 70: Seal leptin with amino acids 23-49 replaced by metreleptin amino acids 25-51 (AB loop) and with amino acids 93-119 replaced by amino acids 95-121 (loop 34) of metreleptin, respectively:PIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLPW ASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 70)
[00060] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 23-49 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 25-51 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 93-119 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 95-121 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 71: Seal leptin with N-terminal methionine, with amino acids 23-49 replaced by amino acids 25-51 (AB loop) of metreleptin and with amino acids 93-119 substituted by metreleptin amino acids 95-121 (loop 3-4), respectively: MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 71).
[00061] In some embodiments, the present disclosure pertains to chimeric polypeptides that are based on wild-type seal leptin with an incorporated AB loop sequence, an incorporated 3-4 loop sequence, and an incorporated helix 3 sequence. from a mature human leptin. In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide (SEQ ID NO: 28), wherein the contiguous region spanning amino acids at positions 23-49 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids at positions 25-51 of A100 (SEQ ID NO: 24), the contiguous region spanning amino acids at positions 93-119 of SEQ ID NO: 28 have been replaced by a region contiguous region spanning amino acids at positions 95-121 of A100 (SEQ ID NO: 24) and the contiguous region spanning amino acids at positions 71-92 of SEQ ID NO: 28 have been replaced by a contiguous region spanning amino acids in positions 73-94 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 72: Seal leptin with amino acids 23-49 replaced by amino acids 25-51 (AB loop) of metreleptin, with amino acids 93-119 replaced by amino acids 95-121 (loop 34) of metreleptin and with amino acids 71-92 replaced by amino acids 73-94 (helix 3) of metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLPPGW ASGLETLDSLGGVLEASGYSTEVVALSRLKAAL
[00062] In some embodiments, a chimeric polypeptide comprises the amino acid sequence of a wild-type seal leptin polypeptide with an N-terminal methionine (SEQ ID NO: 31), wherein the contiguous region spanning the amino acids at positions 23-49 of SEQ ID NO: 31 have been replaced by a contiguous region spanning amino acids at positions 25-51 of A100 (SEQ ID NO: 24), the contiguous region spanning amino acids at positions 93-119 of SEQ ID NO:31 have been replaced with a contiguous region spanning amino acids at positions 95-121 of A100 (SEQ ID NO:24) and the contiguous region spanning amino acids at positions 71-92 of SEQ ID NO:28 have been substituted by a contiguous region spanning amino acids at positions 73-94 of A100 (SEQ ID NO: 24). In some embodiments, a chimeric polypeptide comprises the sequence described in SEQ ID NO: 73: N-terminal methionine seal leptin, with amino acids 23-49 replaced by metreleptin amino acids 25-51 (AB loop), with amino acids 93-119 replaced by amino acids 95-121 (3-4 loop) of metreleptin and replaced by amino acids 71-92 amino acids 73-94 (helix 3) metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 73).
[00063] In some embodiments, the chimeric polypeptides provided by the invention contain a Cys to Ser substitution at position 30 of the wild-type seal polypeptide sequence. According to some embodiments, the following are provided chimeric polypeptides: Leptin seal 30 and with amino acids replaced by amino acids 3-22 and 5-24 32 (first helix) demetreleptina respectively: PIQKVQDDTKTLIKTIVTRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 74) .Leptina seal with N-terminal methionine and amino acids 30 and replaced by amino acids 3-22 and 5-24 32 (helix 1) metreleptin, respectively: MPIQKVQDDTKTLIKTIVTRINDISPPQGVSSRPRVAGLDFIPRVQSVR TLSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 75) with seal .Leptina amino acids 30 and 50-65 replaced by metreleptin amino acids 32 and 52-67 (helix 2), respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SKMDQTLAVYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDM7Ideptin IDPN6). with N-terminal methionine and with amino acids 30 and 50-65 replaced by metreleptin amino acids 32 and 52-67 (helix 2), respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSKMDQTLAVYQQILTSLQSRSVVQIANDLANLRALLRLQLASAKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLQ (NO CDM LRQRNQ 7KA ID). Seal leptin with amino acids 30 and 71-92 replaced by metreleptin amino acids 32 and 73-94 (helix 3), respectively: VIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLLD: IDQRNQ: NOLRQRNC: ID Leptin seal with N-terminal methionine and amino acids 30 and 71-92 replaced by amino acids 32 and 73-94 (helix 3) metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 33). Leptin seal with amino acids 120-141 replaced by 30 and 32 and amino acids 122-143 (helix 4) metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 78). Leptin seal with N-terminal methionine and amino acids 30 to amino acids 120-141 replaced by 32 and 122143 (Helix 4) metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 79). Seal leptin with amino acids 30 and 93-119 replaced by amino acids 32 and 95-121 (loop 3-4) of metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILLSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSEQLD: NOLRQQ8RNKA IDDLDSLGGVLEASGYSTEVVALSEQLD. Leptin seal with N-terminal methionine and amino acids 93-119 30 and replaced by amino acids 32 and 95 121 (loop 3-4) of metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 81). Leptin seal 30 amino acid substituted with amino acid 32, amino acids 3-22 replaced by 524 amino acids (Helix 1) metreleptin and amino acids 71-92 replaced by amino acids 73-94 (helix 3) metreleptin, respectively: PIQKVQDDTKTLIKTIVTRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 82) N-terminal methionine seal leptin and amino acid 30 substituted for amino acid 32, amino acids 3-22 substituted for amino acids 5-24 (helix 1) of metreleptin and amino acids 72-93 substituted for amino acids 73-94 ( helix 3) metreleptin, respectively: MPIQKVQDDTKTLIKTIVTRINDISPPQGVSSRPRVAGLDFIPRVQSVR TLSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 83) seal Leptin amino acid 30 with substituted amino acid 32, amino acid 71-92 substituted for amino acids 73-94 (helix 3) and metreleptin aminoác amino acids 93-119 95-121 substituted by (3-4 loop) of metreleptin, respectively: PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 84) Leptin seal with N-terminal methionine, 30 amino acid substituted for amino acid 32, amino acids 71-92 replaced by amino acids 73-94 (helix 3) metreleptin and replaced by amino acids 93-119 95-121 amino acids (the loop 3-4) of metreleptin, respectively: MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 85).
[00064] Other Modalities. It should be understood that each of the polypeptides described herein are also considered to include (optionally) a methionine at the N-terminus lattice with the first naturally occurring amino acid thereof. For example, mature metreleptin (leptin A100) consists of human leptin to which an N-terminal methionine has been added as described in SEQ ID NO: 24. Similarly, a methionine residue can be included at the N-terminus of any of the following. amino acid sequences and Formulas described herein in general.
[00065] In some embodiments, chimeric polypeptide analogs are provided. A chimeric polypeptide analog may have at least 80%, for example, 80%, 85%, 90%, 95%, 98% or even greater sequence identity to a parent chimeric polypeptide. In some embodiments, the parent chimeric polypeptide is a polypeptide shown in SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO : 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 68, SEQ ID NO: 69, SEQ ID NO: 70 , SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, SEQ ID NO: 74, SEQ ID NO: 75, SEQ ID NO: 76, SEQ ID NO: 77, SEQ ID NO: 78, SEQ ID NO: 79, SEQ ID NO: 80, ID NO: 81, SEQ ID NO: 82, SEQ ID NO: 83, SEQ ID NO: 84 or SEQ ID NO: 85. Accordingly, in some embodiments, chimeric polypeptide analog may have at least 80%, e.g. 80%, 85%, 90%, 95%, 98% or even greater sequence identity to any chimeric polypeptide selected from the group consisting of SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32 and SEQ ID NO: 33. In some embodiments , a chimeric polypeptide analog may have at least 80%, for example, 80%, 85%, 90%, 95%, 98% or even greater sequence identity to the chimeric polypeptide shown in SEQ ID NO: 33. In In some embodiments, a chimeric polypeptide analog may have at least 80%, for example, 80%, 85%, 90%, 95%, 98%, or even greater sequence identity to the chimeric polypeptide shown in SEQ ID NO: 29 , SEQ ID NO: 30, SEQ ID NO: 32, or SEQ ID NO: 33. In some embodiments, a chimeric polypeptide analog may have at least 90% sequence identity to the chimeric polypeptide shown in SEQ ID NO: 33 .
[00066] Additionally, chimeric polypeptide analogs can be designed, prepared and used according to the invention, in which 1,2, 3, 4, 5, 6, 7, 8.9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or even 21 amino acids of a leptin selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33 , SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO : 68, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, SEQ ID NO: 74, SEQ ID NO: 75, SEQ ID NO: 76 , SEQ ID NO: 77, SEQ ID NO: 78, SEQ ID NO: 79, SEQ ID NO: 80, ID NO: 81, SEQ ID NO: 82, SEQ ID NO: 83, SEQ ID NO: 84, and SEQ ID NO: 85 are substituted with another amino acid, such as a conservative amino acid or a non-conservative amino acid, or are otherwise altered. As is customary in the art, the term "conservative", in the context of amino acid substitutions, refers to substitution which maintains properties of charge type (e.g., anionic, cationic, neutral, polar and the like), hydrophobicity or hydrophilicity, density (e.g. van der Waals contacts and the like) and/or functionality (e.g. hydroxyl, amine, sulfhydryl and the like). The term "non-conservative" refers to an amino acid substitution that is non-conservative.
[00067] In another aspect, the invention provides chimeric polypeptide analogs comprising at least a 1-30 amino acid contiguous region of a mature human leptin analog sequence that contains at least one amino acid substitution at a position where divergence is observed in a corresponding position on a leptin of another species.
[00068] As understood in the art, for example, murine leptins, rat leptins, bovine leptins, porcine leptins and rhesus leptins such as those described herein are each substantially homologous to human leptins; in particular, the mature forms of these leptins are substantially homologous to the mature leptins, and yet particularly close to the N-terminal portion of the protein. Analogs of such leptins, such as human mature letpin form 1 (SEQ ID NO: 20) and metreleptin (SEQ ID NO: 24) can be prepared such as, for example, by substituting or altering amino acid residues in one or more positions in such sequences where divergence is observed in a corresponding mature mouse, rat, bovine, porcine or rhesus leptin. For example, mature human leptins (eg, SEQ ID NO: 20) induce biological responses, eg in mice, rats and monkeys). See, for example, WO 98/28427, WO 2009/064298, US2007/0020284, US2008/0207512 and Murakami et al., 1995, Biochem. Biophys. Res. 209: 944-952. By virtue of the fact that mature human leptins have biological activity, for example, in such species, leptin analogues can be designed and prepared in which one or more amino acids at positions which are divergent from the corresponding position(s)( s) in a leptin of one or more such species are substituted for the corresponding amino acid(s) at such divergent positions.
[00069] For example, using a mature human leptin protein according to SEQ ID NO: 20 wherein the first amino acid is valine and the amino acid at position 146 is cysteine, one or more of the amino acids at positions 32, 35, 50, 64, 68, 71, 74, 77, 89, 97, 100, 101, 105, 106, 107, 108, 111, 118, 136, 138, 142 and 145 for amino acid(s) ) corresponding(s) found at the corresponding position(s) in SEQ ID NO: 2, in order to design and prepare leptin analogues comprised of the chimeric polypeptides according to the invention. Additionally, one can also substitute another amino acid, such as a conservative amino acid or a non-conservative amino acid, at one or more of positions 32, 35, 50, 64, 68, 71, 74, 77, 89, 97, 100, 101 , 105, 106, 107, 108, 111, 118, 136, 138, 142 and 145, for example, of SEQ ID NO: 20 in order to design and prepare leptin analogues comprised of the chimeric polypeptides according to the invention.
[00070] Further leptin analogs can be prepared based on the sequence of the mouse mature leptin protein (SEQ ID NO: 16). See, for example, WO 98/28427, US2007/0020284 and Murakami et al., 1995, Id., incorporated herein by reference in full and for all intents and purposes. Mature mouse leptin differs from mature human leptin form 1 (SEQ ID NO: 20) at the following positions: 4, 32, 33, 35, 50, 68, 71, 74, 77, 78, 89, 97, 100, 101 , 102, 105, 106, 107, 108, 111, 118, 136, 138 and 145. Consequently, at one or more of such positions in SEQ ID NO: 20, one may substitute the amino acid(s) found (s) at the corresponding position(s) found in mature rat leptin (SEQ ID NO: 16) in order to design and prepare leptin analogues comprised of the chimeric polypeptides according to the invention. Additionally, one can also substitute another amino acid, such as a conservative amino acid or a non-conservative amino acid, at one or more of positions 4, 32, 33, 35, 50, 68, 71, 74, 77, 78, 89,97 , 100, 101, 102, 105, 106, 107, 108, 111, 118, 136, 138 and 145, for example, of SEQ ID NO: 20, in order to design and prepare leptin analogues comprised of the chimeric polypeptides according to with the invention.
[00071] The positions of mature rat leptin (SEQ ID NO: 16) and mature murine leptin form 1 (SEQ ID NO: 2) which diverge from mature human leptin form 1 (SEQ ID NO: 20) are as follows: 4, 32, 33, 35, 50, 64, 68, 71, 74, 77, 78, 89, 97, 100, 102, 105, 106, 107, 108, 111, 118, 136, 20 138 , 142 and 145. Accordingly, at one or more of such positions in SEQ ID NO: 20, one may substitute the found amino acid(s) in the corresponding position(s) found (s) in the rat mature leptin sequence (SEQ ID NO: 16) or murine mature leptin form 1 sequence (SEQ ID NO: 2), in order to design and prepare leptin analogs comprised of the chimeric polypeptides according to with the invention. Additionally, one can also substitute another amino acid, such as a conservative amino acid or a non-conservative amino acid, at one or more of positions 4, 32, 33, 35, 50, 64, 68, 71, 74, 77, 78, 89 , 97, 100, 102, 105, 106, 107, 108, 111, 118, 136, 138, 142 and 145, in order to design and prepare leptin analogues comprised of the chimeric polypeptides according to the invention.
[00072] In addition, amino acids found in mature rhesus monkey leptin (SEQ ID NO: 14) which diverge from form 1 of mature human leptin (SEQ ID NO: 20) are (with amino acid residues indicated in parentheses in the abbreviation one letter amino acid): 8 (S), 35 (R), 48 (V), 53 (Q), 60 (1), 66 (1), 67 (N), 68 ((L), 89 ( L), 100 (L), 108 (E), 112 (D) and 118 (L) Since mature human leptins trigger biological responses in monkeys, a leptin, such as form 1 of mature human leptin (SEQ ID NO: 20) with one or more of the rhesus monkey divergent amino acids substituted for another amino acid, such as the amino acids in parentheses, can be employed in the design, preparation and use of leptin analogs comprised of the chimeric polypeptides according to the invention. It should be noted that certain rhesus diverging amino acids are also those found, for example, in form 1 of mature murine leptin above (positions 35, 68, 89, 100 and 112). Thus, leptin analogs can be prepared in which one or more amino acids at positions 4, 8, 32, 33, 35, 48, 50, 53, 60, 64, 66, 67, 68, 71, 74, 77, 78 , 89, 97, 100, 102, 105, 106, 107, 108, 111, 112, 118, 136, 138, 142 and 145, e.g. of human mature leptin form 1 (SEQ ID NO: 20), are substituted for the corresponding amino acid(s) at said position(s) in rhesus monkey or murine leptins (e.g. SEQ ID NO: 2 and/or SEQ ID NO: 14 ).
[00073] In accordance with the invention, chimeric polypeptide analogs can be designed and prepared to comprise contiguous amino acid regions of human leptin analogs. In some embodiments, the invention provides chimeric polypeptide analogs based on a wild-type seal leptin polypeptide in which at least one contiguous 1-30 amino acid region of a wild-type seal leptin sequence has been replaced by a region contiguous 1-30 amino acid sequence of a mature human leptin analogue sequence and wherein the mature human leptin nucleic acid sequence contains at least one amino acid substitution at a position where divergence is observed at a corresponding position in one leptin from another species. Chimeric polypeptide analogs comprising two or more contiguous 1-30 amino acid regions of a mature human leptin analog sequence are also contemplated.
[00074] Chimeric polypeptides to which a chemical moiety is attached are polypeptide derivatives. Derivatization of chimeric polypeptides through linkage of one or more chemical moieties has been found to confer some advantage under certain circumstances, such as increased stability and circulation time of the therapeutic protein and decreased immunogenicity and propensity, for example, to generate antibodies. neutralizing agents and/or incidence of injection site reactions. See, for example, WO 98/28427, US2007/0020284, US Patent No. 4,179,337, Davis et al., issued December 18, 1979. For a review, see Abuchowski et al., in ENZYMES AS DRUGS (JS Holcerberg and J.
[00075] Polypeptide derivatives may constitute polypeptides to which a chemical modification has been made to one or more of their amino acid side groups, α-carbon atoms, terminal amino group or terminal carboxylic acid group. A chemical modification includes, but is not limited to, linking one or more chemical moieties, creating new bonds, and removing one or more chemical moieties. Modifications to amino acid side chain groups include, without limitation, alkylation, acylation, ester formation, amide formation, maleimide coupling, acylation of lysine ε-amino groups, N-alkylation of arginine, histidine or lysine, alkylation of glutamic or aspartic carboxylic acid groups and deamidation of glutamine or asparagine. Amino terminus modifications include, without limitation, desamino, N-lower alkyl, di-N-lower alkyl and N-acyl modifications. Amino terminus modifications include, without limitation, desamino, N-lower alkyl, N-di-lower alkyl and N-acyl modifications, such as alkyl, branched alkyl, alkylaryl-acyl. Modifications of the terminal carboxy group include, but are not limited to, amide, lower alkyl amide, dialkyl amide, arylamide, alkylarylamide and lower alkyl ester modifications. Lower alkyl is C1-C4 alkyl. In addition, one or more side groups or end groups may be protected by protecting groups known to those skilled in synthetic chemistry. The α-carbon of an amino acid can be mono- or dimethylated.
[00076] Such derivatives include polypeptides conjugated to one or more water-soluble polymer molecules, such as polyethylene glycol ("PEG") or fatty acid chains of various lengths (e.g., stearyl, palmitoyl, octanoyl), by the addition of polyamino acids such as poly-his, poly-arg, poly-lys and poly-ala or by adding small molecule substituents, including short alkyl and restricted alkyl (e.g. branched, cyclic, fused, adamantyl) and aromatic groups . In some embodiments, the water-soluble polymeric molecules will have a molecular weight ranging from about 500 Daltons to about 60,000 Daltons.
[00077] Such polymeric conjugations may occur alone at the N- or C-terminus or side chains of amino acid residues within the sequence of a chimeric polypeptide, as described herein. Alternatively, there may be multiple derivatization sites along the chimeric polypeptide amino acid sequence. Substitution of one or more amino acids for lysine, aspartic acid, glutamic acid or cysteine can provide additional sites for derivatization. See, for example, U.S. Patent Nos. 5,824,784 and 5,824,778. In some embodiments, a chimeric polypeptide may be conjugated to one, two or three polymeric molecules.
[00078] In some embodiments, the water-soluble polymer molecules are attached to an amino, carboxyl, or thiol group and may be attached at the N or C terminus or on the side chains of lysine, aspartic acid, glutamic acid, or cysteine. Alternatively, water-soluble polymeric molecules can be linked with diamine and dicarboxylic groups. In some embodiments, a chimeric polypeptide is conjugated to one, two, or three PEG molecules through an epsilon amino group of a lysine amino acid.
[00079] Polypeptide derivatives also include polypeptides with chemical changes at one or more amino acid residues. Such chemical changes include amidation, glycosylation, acylation, sulfation, phosphorylation, acetylation, and cyclization. Chemical changes can occur particularly at the N- or C-terminus or side chains of amino acid residues within a leptin sequence. In one embodiment, the C-terminus of these peptides may have a free -OH or -NH 2 group. In another embodiment, the N-terminus may be capped with an isobutyloxycarbonyl group, an isopropyloxycarbonyl group, an n-butyloxycarbonyl group, an ethoxycarbonyl group, an isocaproyl ("isocap") group, an octanyl group, a octylglycine group (denoted as "G(Oct)" or "octylGly"), an 8-amino-octanic acid group, a dansyl and/or Fmoc group. In some embodiments, cyclization may be through disulfide bridge formation. Alternatively, there may be multiple sites of chemical change along the amino acid sequence of the polypeptide.
[00080] In certain embodiments, chimeric polypeptides are chemically altered to include a Bolton-Hunter group. Bolton-Hunter reagents are known in the art ("Radioimmunoassay and Related Methods", AE Bolton and WM Hunter, Chapter 26 of the HANDBOOK OF EXPERIMENTAL IMMUNOLOGY, VOLUME I, IMMUNOCHEMISTRY, edited by DM Weir, Blackwell Scientific Publications, 1986) and may be used to introduce neutrally bonded tyrosine-like moieties through terminal oc-amino groups or 8-amino groups of lysine. In some embodiments, the N-terminal end of a polypeptide is modified with a Bolton-Hunter group. In some embodiments, an internal lysine residue is modified with a Bolton-Hunter group. In some embodiments, there may be multiple Bolton-Hunter modification sites along the amino acid sequence of the polypeptide. Bolton-Hunter reagents used for polypeptide modification are commercially available and may include, but are not limited to, water-soluble Bolton-Hunter reagent, sulfo-succinimidyl-3-[4-hydrophenyl]propionate (Pierce Biotechnology, Inc., Rockford , IL) and Bolton-Hunter reagent 2, N-succinimidyl-3-(4-hydroxy-3-iodo-phenyl)propionate (Wako Pure Chemical Industries, Ltd., Japan, catalog 199-09341). An exemplary Bolton-Hunter group conjugated through an amide bond to a polypeptide is illustrated below, where the dashed line passes through the amide bond.

[00081] Polypeptides can be iodinated (such as radiolabeled with 125 I) before or after Bolton-Hunter modification.
[00082] Derivatives of polypeptides may include one or more modifications of a "non-essential" amino acid residue. In the context of the invention, a "non-essential" amino acid residue is a residue that can be altered, e.g., derivatized, without substantially eliminating or reducing the activity (e.g., agonist activity) of the chimeric polypeptide. The chimeric polypeptides of the invention may include derivatizations of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acid residues; of these, one or more amino acid residues may be non-essential amino acid residues. Additionally, the polypeptides of the invention can be derivatized so that they include additions of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids without eliminating or substantially reducing the activity of the polypeptide. Additionally, such non-essential amino acid residues may be substituted for an amino acid residue that is amenable to derivatization, as described herein.
[00083] As used herein, "amino acid", "amino acid residue" and the like refer to natural amino acids, unnatural amino acids and modified amino acids. Unless otherwise indicated, any reference to an amino acid in general or specifically by name includes reference to both the D and L stereoisomers if its structure permits these stereoisomeric forms. Natural amino acids include alanine (Ala), arginine (Arg), asparagine (Asn), aspartic acid (Asp), cysteine (Cys), glutamine (Gln), glutamic acid (Glu), glycine (Gly), histidine (His), isoleucine (Ile), leucine (Leu), lysine (Lys), methionine (Met), phenylalanine (Phe), proline (Pro), serine (Ser), threonine (Thr), tryptophan (Trp), tyrosine (Tyr) and valine (Val). Unnatural amino acids include, but are not limited to, homolysine, homoarginine, homoserine, azetidine carboxylic acid, 2-aminoadipic acid, 3-aminoadipic acid, beta-alanine, aminopropionic acid, 2-aminobutyric acid, 4-aminobutyric acid, 6-aminobutyric acid -aminocaproic acid, 2-aminoheptanoic acid, 2-aminoisobutyric acid, 3-aminoisobutyric acid, 2-aminopimelic acid, tert-butylglycine, 2,4-diaminoisobutyric acid, desmosine, 2,2'-diaminopimelic acid, 2,3 acid -diaminopropionic acid, N-ethylglycine, N-ethylasparagine, homoproline, hydroxylysine, allohydroxylysine, 3-hydroxyproline, 4-hydroxyproline, isodesmosine, alloisoleucine, N-methylalanine, N-methylglycine, N-methylisoleucine, N-methylpentylglycine , N-methylvaline, naphthalanine, norvaline, norleucine, ornithine, pentylglycine, pipecolic acid and thioproline. Additional unnatural amino acids include modified amino acid residues that are chemically blocked, reversibly or irreversibly, or chemically modified about their N-terminal amino group or their side chain groups, e.g. D and L N-methylated amino acids or residues where side chain functional groups are chemically modified to another functional group. For example, modified amino acids include methionine sulfoxide; methionine sulfone, aspartic acid (beta-methyl ester), an amino acid modified from aspartic acid; N-ethylglycine; a modified glycine amino acid; or alanine carboxamide; a modified alanine amino acid. Additional residues that can be incorporated are described in Sandberg et al., J. Med. Chem. Chem. 41: 2481-91, 1998.
[00084] As mentioned above, suitable chemical moieties for such derivatization of the chimeric polypeptides include, for example, various water-soluble polymers. Preferably, for therapeutic use of the final product preparation, the polymer will be pharmaceutically acceptable. Those skilled in the art will be able to select the desired polymer based on considerations such as whether the polymer/protein conjugate will be used therapeutically and, if so, the desired dosage, circulation time, resistance to proteolysis, and other considerations. For chimeric polypeptides, the effectiveness of derivatization can be verified by administering the derivatized leptin or derivatized polypeptide, in the desired form (i.e., by means of an osmotic pump or, more preferably, by injection or infusion, or formulated for delivery). oral, pulmonary or nasal, for example) and observing biological effects and biological responses, as described here.
[00085] Such a water-soluble polymer may be selected from the group consisting, for example, of polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxy methyl cellulose, dextran, polyvinyl alcohol, polyvinylpyrrolidone, poly-1,3-dioxolane, poly-1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyamino acids (homopolymers or random copolymers) and dextran or poly(n-vinyl pyrrolidone) / polyethylene glycol, propylene glycol homopolymers, polypropylene oxide/oxide copolymers of ethylene, polyoxyethylated polyols and polyvinyl alcohol. Polyethylene glycol propionaldehyde may have manufacturing advantages because of its stability in water. Also, succinate, styrene and hydroxyethyl starch can be used.
[00086] Derivatives of chimeric polypeptides according to the invention can be prepared by linking polyamino acids or branch point amino acids to the leptin moiety. For example, the polyamino acid may be an additional carrier protein, such as an Fc moiety, which may also serve to increase the circulating half-life of the leptin or chimeric polypeptide. Additionally, such polyamino acids may be selected from the group consisting of serum albumin (such as human serum albumin), an additional antibody or portion thereof (e.g., the Fc region), or other polyamino acids, e.g., polylysines. As indicated below, the polyamino acid binding location may be at the N-terminus of the leptin moiety or C-terminus or other sites therebetween and may also be attached by a chemical "linker" moiety to the leptin, such as a linker. peptide or a non-peptide linker.
[00087] The polymer can be of any molecular weight and can be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about 2 kilodaltons (kDa) and about 100 kDa (the term "about" indicates that, in polyethylene glycol preparations, some molecules will weigh more, some less, than the molecular weight indicated) for ease of handling and fabrication. In certain embodiments, the polyethylene glycol is between about 2 kDa and about 60 kDa. In certain embodiments, the polyethylene glycol is between about 2 kDa and about 40 kDa. In certain embodiments, the polyethylene glycol is between about 5 kDa and about 40 kDa. In certain embodiments, the polyethylene glycol is between about 10 kDa and about 40 kDa. In certain embodiments, the polyethylene glycol is between about 5 kDa and about 30 kDa. In certain embodiments, the polyethylene glycol is from about 5 kDa to about 20 kDa. In certain embodiments, the polyethylene glycol is between about 10 kDa and about 20 kDa. Other sizes may be used, depending on the desired therapeutic profile (e.g., duration of sustained release desired, solubility characteristics, effects, if any, on biological activity, ease of handling, degree or lack of antigenicity, and others. known effects of polyethylene glycol linked to a leptin and/or a chimeric polypeptide of the invention). Additional considerations that may influence the choice of a PEG of a particular molecular weight that can be linked to a leptin to generate a leptin derivative in accordance with the invention include the extent to which PEG of such molecular weight can: reduce aggregation and/or increase the solubility of leptin and/or the chimeric polypeptide when present in a pharmaceutically acceptable composition or formulation or when exposed to physiological fluids or tissues when administered to a subject (e.g., by injection); and alleviating the incidence of injection site reactions caused by the administration of leptin or the chimeric polypeptide when administered to a subject by injection; alleviating the generation of neutralizing antibodies that can be raised against the leptin or chimeric polypeptide as a result of administering such leptin or chimeric polypeptide to a subject and so on.
[00088] The number of polymer molecules thus bound may vary and those skilled in the art will be able to verify the resulting effect on function. One can mono-derivatize or provide a di-, tri-, tetra- or some combination of derivatization, with the same or different chemical moieties (e.g. polymers such as polyethylene glycols of different weights). The ratio of polymer molecules to leptin molecules or chimeric polypeptide molecules to be derivatized will vary, as will their concentrations in the reaction mixture. In general, the optimal ratio, in terms of reaction efficiency, where there is no excess of unreacted leptin (or chimeric polypeptide, as the case may be) or polymer will be determined by factors such as the desired degree of derivatization (e.g., mono -, di-, tri-, etc.), the molecular weight of the selected polymer, whether the polymer is branched or unbranched, and the reaction conditions.
[00089] The chemical moieties should be linked to leptin and/or chimeric polypeptide considering the effects on functional or antigenic domains of leptin and/or chimeric polypeptide. There are a number of bonding methods available to those skilled in the art. For example, EP 0 401 384, incorporated herein by reference (Coupling of PEG to G-CSF), see also Malik et al., 1992, Exp. Hematol. 20: 1028-1035 (which reports pegylation of GM-CSF using tresyl chloride). For example, polyethylene glycol can be covalently linked through amino acid residues via a reactive group, such as a free amino or carboxyl group. Reactive groups are those to which an activated polyethylene glycol molecule can be attached. Amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residue. Those having a free carboxyl group may include aspartic acid residues, glutamic acid residues and the C-terminal amino acid residue. Sulphydryl groups can also be used as a reactive group for linking the polyethylene glycol molecule(s). Preferred for therapeutic purposes is attachment to an amino group, such as attachment to the lysine group or N-terminus. Fixation at residues important for receptor binding should be avoided if receptor binding is desired.
[00090] It may be desired to specifically design and prepare a chemically modified N-terminus leptin for use in preparing chimeric polypeptides of the invention. Using polyethylene glycol as an illustration of the present compositions one can select, from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the ratio of polyethylene glycol molecules to leptin or chimeric polypeptide molecules, as appropriate, in the reaction mixture, the type of pegylation reaction to be performed and the method for obtaining the selected N-terminally pegylated protein. The method of obtaining the N-terminally pegylated preparation (i.e., separating this portion from other monopegylated portions, if necessary) may be by purifying the N-terminally pegylated material from a population of PEGylated protein molecules. Selective N-terminal chemical modification can be performed through reductive alkylation, which exploits the differential reactivity of different types of primary amino groups (lysine versus the N-terminus) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a polymer containing carbonyl group is obtained. For example, one can selectively pegylate the N-terminal protein by carrying out the reaction at a pH which allows one to take advantage of the pKa differences between the ε-amino group of lysine residues and that of the α-amino group of the N-terminal residue. of the protein. Through such selective derivatization, binding of a water-soluble polymer to a protein is controlled: conjugation with the polymer occurs predominantly at the N-terminus of the protein and there is no significant modification of other reactive groups, such as the amino groups of the chain. lysine side. Using reductive alkylation, the water-soluble polymer can be of the type described above and should have a single reactive aldehyde for coupling to the protein. Polyethylene glycol propionaldehyde, which contains a single reactive aldehyde, can be used. III. Design and Production Methods
[00091] Design of constructs. The chimeric polypeptides described herein can be designed at the amino acid level. These sequences can then be re-translated using a variety of software products known in the art so that the nucleotide sequence is optimized for the intended expression host, e.g. protein based expression, codon optimization, restriction site content. For example, the nucleotide sequence can be optimized for E. coli based protein expression and restriction site content. Based on the nucleotide sequence of interest, overlapping oligonucleotides can be provided for various PCR steps, as is known in the art. These oligonucleotides can be used in multiple PCR reactions under conditions well known in the art for the construction of cDNA encoding the protein of interest. An example is 1X Amplitaq buffer, 1.3 mM MgCh, 200 μm dNTPs, 4 U of Amplitaq Gold, 0.2 μM of each primer (AmpliTaq Gold, ABI), with cycling parameters: (94°C: 30s , 58°C: 1 min, 72°C: 1 min), 35 cycles.
[00092] Restriction sites may be added to the ends of PCR products for use in vector binding, as known in the art. Specific sites can include NdeI and XhoI, so the cDNA can then be in the proper reading frame in a pET45b expression vector (Novagen). When using these sites, any N-terminal His Tags that are in this vector can be removed, as the translation start site would then be downstream of the tag. Once expression constructs are terminated, verification can be conducted via sequencing using, for example, the T7 promoter primer, T7 terminator primer, and standard ABI BigDye Term v3.1 protocols, as known in the art. Sequence information can be obtained, for example, from an ABI 3730 DNA Analyzer and can be analyzed using Vector NTI v. 19 (Invitrogen). Expression constructs can be designed in a modular fashion, so that linker sequences can be easily cut and altered, as is known in the art.
[00093] Protease recognition sites, known in the art or described herein, can be incorporated into constructs useful for the design, construction, manipulation and production of the chimeric recombinant polypeptides described herein.
[00094] General Production Methods. The chimeric polypeptides described herein can be prepared using biological, chemical and/or recombinant DNA methods that are known in the art. Exemplary methods are described herein and in US Patent No. 6,872,700 , WO 2007/139941 , WO 2007/140284 , WO 2008/082274 , WO 2009/011544 , and US Publication No. 5 2007/0238669 , the descriptions of which are incorporated herein by reference in their entirety and for all intents and purposes. Other methods for preparing the compounds are presented here.
[00095] The chimeric polypeptides described herein can be prepared using standard solid phase peptide synthesis techniques, such as an automated or semi-automated peptide synthesizer. The chimeric polypeptide can be produced by non-biological peptide synthesis using amino acids and/or amino acid derivatives having protected reactive side chains, non-biological peptide synthesis comprising stepwise coupling of amino acids and/or the amino acid derivatives to form a polypeptide according to the first aspect having protected reactive side chains, removing protecting groups from the reactive side chains of the polypeptide, and folding the polypeptide in aqueous solution. Thus, normal amino acids (eg, glycine, alanine, phenylalanine, isoleucine, leucine, and valine) and pre-protected amino acid derivatives are used to sequentially build a polypeptide sequence in solution or on a solid support in an organic solvent. When a complete polypeptide sequence is constructed, the protecting groups are removed and the polypeptide is allowed to fold in an aqueous solution.
[00096] Typically, using these techniques, an alpha-N-carbamoyl amino acid and an amino acid protected from an amino acid attached to the peptide chain growing on a resin are coupled at room temperature in an inert solvent (e.g., dimethylformamide, N-methylpyrrolidinone, methylene chloride and the like) in the presence of coupling agents (e.g. dicyclohexylcarbodiimide, 1-hydroxybenzotriazole and the like) in the presence of a base (e.g. diisopropylethylamine and the like). The alpha-N-carbamoyl protecting group is removed from the resulting peptide-resin using a reagent (e.g. trifluoroacetic acid, piperidine, and the like) and the coupling reaction repeated with the next N-protected amino acid to be added to the peptide chain. . Suitable N-protecting groups are well known in the art, such as t-butyloxycarbonyl (tBoc) fluorenylmethoxycarbonyl (Fmoc) and the like. Solvents, amino acid derivatives and 4-amino methylbenzhydryl resin used in the peptide synthesizer are available from Applied Biosystems Inc. (Foster City, California).
[00097] For chemical synthesis, solid phase peptide synthesis can be used for the chimeric polypeptides since, in general, solid phase synthesis is a simple approach with excellent scalability to commercial scale and is generally compatible with relatively long manipulated polypeptides. Solid phase peptide synthesis can be performed with an automated peptide synthesizer (Model 430A, Applied Biosystems Inc., Foster City, CA) using the NMP/HOBt system (Option 1) and tBoc or Fmoc chemistry (see User Manual). from Applied Biosystems for ABI 430A Peptide Synthesizer A, Version 1.3B, July 1, 1988, section 6, pages 49-70, Applied Biosystems, Inc., Foster City, California) with capping. Boc-peptide-resins can be cleaved with HF (-5°C to 0°C, 1 hour). The peptide can be extracted from the resin by alternating water with acetic acid and the lyophilized filtrates. Fmoc-peptide resins can be cleaved according to standard methods (eg, Introduction to Cleavage Techniques, Applied Biosystems, Inc., 1990, pages 6-12). Peptides can also be assembled using an Advanced Chem Synthesizer Tech (Model MPS 350, Louisville, Kentucky).
[00098] The compounds described herein may also be prepared using recombinant DNA techniques, using methods known in the art, such as Sambrook et al., 1989, Molecular Cloning: A Laboratory Manual, 2ndEd., Cold Spring Harbor. Non-peptidic compounds can be prepared by methods known in the art. For example, phosphate-containing amino acids and peptides containing such amino acids can be prepared using methods known in the art, as described in Bartlett et al., 1986, Biorg. Chem., 14: 356-377. IV. Methods of use and treatment of diseases
[00099] Indications. A variety of diseases and disorders are believed to be beneficially treated by the polypeptide compounds and methods described herein.
[000100] Obesity and overweight. Obesity and its associated diseases, including being overweight, are common and serious public health problems in the United States and around the world. Obesity in the upper body is the main known risk factor for type 2 diabetes mellitus and is a strong risk factor for cardiovascular disease. Obesity is a recognized risk factor for hypertension, atherosclerosis, congestive heart failure, stroke, gallbladder disease, osteoarthritis, sleep apnea, reproductive disorders such as polycystic ovary syndrome, breast, prostate and colon cancer. and increased incidence of complications from general anesthesia. See, for example, Kopelman, 2000, Nature 404: 635-43.
[000101] Obesity shortens life expectancy and carries a serious risk of the co-morbidities listed above, as well as disorders such as infections, varicose veins, acanthosis nigricans, eczema, exercise intolerance, insulin resistance, hypercholesterolemia, hypertension, cholelithiasis, orthopedic injuries and thromboembolic disease. See, for example, Rissanen et al., 1990, Br. Med. J., 301: 835-7. Obesity is also a risk factor for the group of conditions known as insulin resistance syndrome or "Syndrome X" and metabolic syndrome. The worldwide medical cost of obesity and associated disorders is enormous.
[000102] The pathogenesis of obesity is believed to be multi-factorial. One problem is that, in obese individuals, nutrient availability and energy expenditure do not come into balance until there is excess adipose tissue. The central nervous system (CNS) controls energy balance and coordinates a variety of behavioral, autonomic and endocrine activities appropriate to the animal's metabolic state. The mechanisms or systems that control these activities are widely distributed throughout the forebrain (eg, hypothalamus), hindbrain (eg, brainstem), and spinal cord. Ultimately, metabolic (i.e., fuel availability) and cognitive (i.e., learned preferences) information from these systems are integrated and the decision to engage in appetite (food-seeking) and consumption (ingestion) behaviors is made. activated (search for food and start ingestion) or deactivated (end of meal). The hypothalamus is thought to be primarily responsible for integrating these signals and then issuing commands to the brainstem. Brainstem nuclei control elements of the consumption motor control system (eg, muscles responsible for mastication and swallowing). As such, these nuclei in the CNS have literally been referred to as the "ultimate common pathway" for ingestion behavior.
[000103] Neuroanatomical and pharmacological evidence supports that energy and nutritional homeostasis signals integrate in the forebrain nuclei and that the consumption motor control system resides in brainstem nuclei, probably in regions around the trigeminal motor nucleus. There is extensive reciprocal connection between the hypothalamus and the brain stem. A variety of CNS-targeted anti-obesity therapeutics (eg, small molecules and peptides) predominantly focus on forebrain substrates residing in the hypothalamus and/or on hindbrain substrates residing in the brainstem.
[000104] Obesity remains a poorly treatable, chronic, essentially intractable metabolic disorder. Consequently, there is a need for new therapies useful in reducing weight and/or maintaining weight in an individual. Such therapies would have a profound beneficial effect on the individual's health. Methods and therapies employing the chimeric peptides described herein, alone or in combination with other anti-obesity agents (see, for example, WO 2009064298 and US 20080207512 ) can confer such beneficial effects.
[000105] Leptin Deficiency. Leptin deficiency has been shown to result in obesity. One form of leptin deficiency is congenital leptin deficiency, a rare genetic disorder. See Montaque et al., 1997, Nature 387: 903-908. Severe leptin deficiency may be a result of uncontrolled insulin-deficient diabetes mellitus that results from destruction of insulin-secreting β-cells. It is postulated that a lack of insulin leads to the synthesis and storage of triglycerides in adipose tissue, which prevents weight gain and, in turn, significantly reduces plasma leptin levels as leptin is synthesized in the tissue. adipose. These and other leptin deficiencies and diseases and disorders resulting from such deficiencies can be treated with leptin replacement therapy, such as through daily injections of leptin or leptin agonists. The chimeric polypeptides described herein may provide a more convenient and advantageous therapeutic treatment of such diseases and disorders.
[000106] Diabetes and Cardiovascular Disease. Diabetes mellitus is recognized as a complex chronic disease in which 60% to 70% of all case deaths among diabetic patients are a result of cardiovascular complications. Diabetes is considered not only a risk equivalent to coronary heart disease, but is also identified as an independent prognosis of adverse events, including recurrent myocardial infarction, congestive heart failure, and death after a cardiovascular incident. It would be expected that the adoption of stricter glucose control and aggressive treatment for cardiovascular risk factors would reduce the risk of coronary heart disease complications and improve overall survival in diabetic patients. However, diabetic patients are two to three times more likely to have an acute myocardial infarction than non-diabetic and diabetic patients live 8-13 years shorter than non-diabetic patients.
[000107] Understanding the high-risk nature of diabetes/acute myocardial infarction, the American College of Cardiology/American Heart Association ("ACC/AHA") clinical practice guidelines for the management of inpatients with unstable angina or myocardial infarction non-ST-elevation myocardium (collectively referred to as "ACS") has recently recognized that hospitalized diabetic patients are a special population that requires aggressive management of hyperglycemia. Specifically, the guidelines state that glucose-lowering therapy for hospitalized ACS/diabetic patients should be directed toward achieving preprandial glucose levels of less than 10 mg/dL, a maximum daily goal of 180 mg/dL, and a post-discharge hemoglobin A1c of less than 7%.
[000108] In a national sample of elderly patients with ACS, an increase in 30-day mortality in diabetic patients was shown to correspond to patients having higher glucose values at the time of hospital admission. See "Diabetic Coronary Artery Disease & Intervention", Coronary Therapeutics 2002, Oak Brook, IL, September 20, 2002. There is increasing evidence to support that hyperglycemia, rather than transient elevated glucose, upon hospital admission is related to adverse events. serious. Although the optimal measure for hyperglycemia and vascular risk in patients is not readily known, it appears that the mean glucose value during hospitalization is more predictive of mortality. In a separate study of patients with ACS in more than 40 hospitals in the United States, persistent hyperglycemia, as opposed to random glucose values, upon hospital admission, was found to be more predictive of in-hospital mortality. See Acute Coronary Syndrome Summit: A State of the Art Approach, Kansas City, MO, September 21, 2002. Compared with glucose values at admission, a logistic regression model of glycemic control throughout hospitalization was more prognostic of mortality. There was an almost twice as high risk of mortality during hospitalization for every 10 mg/dL increase in glucose above 120 mg/dL. In a small group of consecutive ACS/diabetic patients, there was a gradual increase in mortality within one year, with increasing glucose levels at the time of hospital admission. In the hospital setting, ACC/AHA guidelines suggest initiation of aggressive insulin therapy to achieve lower blood glucose during hospitalization.
[000109] It has been reported that leptin may have direct benefits for the treatment of diabetes, particularly in type I diabetes and type II diabetes, with or without the presence of obesity, and more particularly in conditions of low leptin in the serum. Leptin replacement has been reported to reduce or prevent hyperinsulinemia, hyperglycemia and insulin resistance in several animal models of type 1 and 2 diabetes, with or without associated obesity. For example, elevated plasma leptin levels generated through pharmacological administration of leptin or with adenoviral gene therapy reduced hyperglycemia and associated increases in plasma glucagon levels in STZ-induced diabetes despite persistently low insulin levels.
[000110] Lipid Regulation Diseases. As known in the art, lipodystrophy is characterized by abnormal or degenerative conditions of the body's adipose tissue. Dyslipidemia is a disruption in the normal lipid component in the blood. It is believed that prolonged elevation of insulin levels can lead to dyslipidemia. Hyperlipidemia is the presence of high or abnormal levels of lipids and/or lipoproteins in the blood. Hypothalamic amenorrhea is a condition in which menstruation stops for several months because of a problem involving the hypothalamus. Leptin replacement therapy in women with hypothalamic amenorrhea has been found to improve thyroid, reproductive and growth hormone axes and bone formation markers without causing adverse effects. See, for example, Oral et al., N Engl J Med. 2004, 351: 959-962, 987-997. Fatty liver disease, for example non-alcoholic fatty liver disease (NAFLD), refers to a broad spectrum of liver disease ranging from simple fatty liver (steatosis) to non-alcoholic steatohepatitis (NASH) to cirrhosis ( irreversible, advanced liver scarring). All stages of NAFLD have in common the accumulation of fat (infiltration of fat) in the liver cells (hepatocytes). Leptin is believed to be one of the key regulators for inflammation and fibrosis progression in several chronic liver diseases, including NASH. See, for example, Ikejima et al., Hepatology Res. 33: 151-154.
[000111] Additionally, without wishing to be bound by any theory, it is believed that relative insulin deficiency in type 2 diabetes, glucose toxicity, and increased hepatic free fatty acid load through increased distribution from intra-abdominal adipose tissue through the portal vein, are implicated as possible causes of fatty liver disorders. In fact, it has been hypothesized that eating behavior is the key factor that determines the metabolic syndrome of obesity with its various complications, including NASH. Consequently, treatments aimed at reducing food intake and increasing the number of small meals, as has been demonstrated in patients with type 2 diabetes, can effectively treat and prevent NASH. Drugs that promote insulin secretion and weight loss and delay gastric emptying are also effective in improving glucose tolerance and therefore may improve fatty liver with concomitant hyperinsulinemia. Thus, the use of a leptin, a leptin analogue, for example metreleptin or an active fragment thereof, may be suitable as a treatment modality for this condition. Accordingly, chimeric polypeptides described herein that include a leptin, a leptin analogue or an active fragment thereof may be useful in the treatment of fatty liver diseases.
[000112] Alzheimer's disease. Alzheimer's Disease (AD), as known in the art, is associated with plaques and tangles in the brain, which include dysregulation of β-beta protein. It is believed that brain lipids are closely involved in β-beta-related pathogenic pathways and that an important modulator of lipid homeostasis is leptin. Consequently, leptin can modulate bidirectional β-beta kinesis, reducing its levels extracellularly. In fact, chronic administration of leptin to AD-transgenic animals has been shown to reduce β-beta load in the brain, substantiating its therapeutic potential. See Fewlass et al., 2004, FASEB J., 18: 1870-1878. Additionally, type 2 diabetes mellitus and AD share epidemiological and biochemical characteristics in that both are characterized by aggregates of insoluble proteins with a fibrillar -amylin conformation in pancreatic islets of type 2 DM and Ap in AD brain. Without wishing to be bound by any theory, it is believed that similar toxic mechanisms can characterize type 2 DM and AD. See Lim et al., FEBS Lett., 582: 2188-2194.
[000113] Metabolic Syndrome X. Metabolic Syndrome X is characterized by insulin resistance, dyslipidemia, hypertension and visceral adipose tissue distribution and plays a key role in the pathophysiology of type 2 diabetes. It has also been found to be strongly correlated with NASH, fibrosis and cirrhosis of the liver. Consequently, chimeric polypeptides described herein may be useful in the treatment of metabolic syndrome X.
[000114] Huntington's Disease. Huntington's disease is an autosomal dominant neurodegenerative disease. Characteristics of the disease include motor disorders, dementia, psychiatric problems, and unintentional weight loss. The chimeric polypeptides described herein may be useful in the treatment of Huntington's disease.
[000115] Accordingly, in one aspect, there is provided a method for treating a disease or disorder in an individual. The individual is in need of treatment for the disease or disorder. The disease or disorder may be lipodystrophy, dyslipidemia, hyperlipidemia, overweight, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin deficiency, fatty liver disease, and diabetes (including Type I and Type II). Other diseases and disorders that can be treated by the compounds and methods described herein include non-alcoholic steatohepatitis (NASH) and non-alcoholic liver disease (NAFLD), metabolic syndrome X and Huntington's disease. The method of treatment includes administering to the subject a chimeric polypeptide as described herein in an amount effective to treat the disease or disorder. Essay
[000116] Methods for producing and assaying chimeric polypeptides described herein are generally available to those skilled in the art. In addition, specific methods are described herein, as well as in the patent publications and other references cited herein, which are incorporated by reference for their additional purpose.
[000117] Food Intake. Without wishing to be bound by any theory, it is believed that food intake is useful in assessing the utility of a compound as described here. For example, a number of metabolic pathologies are known to be related to food intake (eg, diabetes, obesity). Accordingly, an initial screen may be conducted to determine the extent to which food intake is modulated by the administration of compounds described herein and an initial positive screen may be useful in subsequent development of a compound.
[000118] In vitro assays. Without wishing to be bound by any theory or mechanism of action, it is believed that there are correlations between the results of in vitro assays (eg, receptor) and the utility of agents for the treatment of diseases and metabolic disorders. Consequently, in vitro assays (eg, cell-based assays) are useful as a screening strategy for potential metabolic agents, as described herein. A variety of in vitro assays are known in the art, including those described below.
[000119] Leptin Binding Assay. Leptin binding can be measured by the potency of a test compound in displacement of recombinant (murine) 125 I-leptin from the surface membrane expressing the chimeric Leptin (Hu) -EPO (Mu) receptor presented by the 32D OBECA cell line ( J Biol Chem 1998; 273 (29): 18365-18373 ). Purified cell membranes can be prepared by homogenizing confluent cell cultures collected from 32D OBECA cells. Membranes can be incubated with 125 I-murine rec-leptin and increasing concentrations of test compound for 3 hours at room temperature in 96-well polystyrene plates. Bound and unbound ligand fractions can then be separated by rapid filtration on 96-well GF/B plates pre-blocked for at least 60' in 0.5% PEI (polyethyleneimine). Fiberglass plates can then be dried, scintillant added and CPM determined by reading on a multiwell scintillation counter capable of reading radiolabeled iodine.
[000120] Leptin Functional Assay. Increased levels of phosphorylated STAT5 (Signal Transducer and Activator of Transcription 5) can be measured after treatment of 32D-Keptin cells ectopically expressing the chimeric Hu-Leptin/Mu-EPO receptor with a test. 32D-Keptin cells (identical to 32D-OBECA cells but maintained in leptin culture) can be leptin starved overnight and then treated with test compounds in 96-well plates for 30 minutes at 37°C , followed by cell extraction. pSTAT5 levels in cell lysates can be determined using Perkin Elmer's AlphaScreen® SureFire® pSTAT5 Assay Kit in a 384-well format (Proxiplate™ 384 Plus). The efficacy of test compounds can be determined with respect to the maximum signal in cell lysates from cells treated with human leptin. pharmaceutical compositions
[000121] In one aspect, pharmaceutical compositions are provided comprising the compounds described herein in combination with a pharmaceutically acceptable excipient (e.g., carrier). The term "pharmaceutically acceptable carrier", as used herein, refers to pharmaceutical excipients, for example, pharmaceutically or physiologically acceptable organic or inorganic carrier substances suitable for enteral or parenteral application that do not adversely react with the active agent. Pharmaceutically acceptable carriers include water, saline solutions (e.g. Ringer's solution and the like), alcohols, oils, gelatins and carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxy methyl cellulose and polyvinylpyrrolidine. Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts to influence the osmotic pressure, buffers, colorings and/or aromatic substances and the like that do not adversely react with the compounds of the invention. methods
[000122] The chimeric polypeptides described herein may be administered alone or may be co-administered to an individual. Co-administration is to be understood as including simultaneous or sequential administration of the compounds individually or in combination (more than one compound). For example, it has been discovered that obesity can be beneficially treated with a combination therapy including a leptin (e.g. metreleptin) and certain other anti-obesity compounds. See, for example, US Published Application No. 2008/0207512. Consequently, a chimeric polypeptide described herein could be useful for the treatment of obesity.
[000123] In some embodiments, the formulations and methods described herein further allow the chimeric polypeptide to be administered with one or more anti-diabetic agents, such as anti-hyperglycemic agents, e.g., insulin, amylins, pramlintide, metformin.
[000124] In some embodiments, the formulations and methods described herein further allow the chimeric polypeptide to be co-administered with one or more cholesterol and/or triglyceride lowering agents. Exemplary agents include HMG CoA reductase inhibitors (e.g., atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin); bile acid scavengers (e.g. colesevelam, cholestyramine, colestipol); fibrates (e.g. fenofibrate, clofibrate, gemfibrozil); ezetimibe, nicotinic acid, probucol, a lovastatin/niacin combination; an atorvastatin/amlodipine combination; and a simvastatin/ezetimibe combination.
[000125] Alternatively, the individual chimeric polypeptides can be co-administered with other anti-obesity agents, such as exenatide or liraglutide.
[000126] The present description provides the composition for use as a medicine, i.e. for use in therapy, since the leptin compound is a therapeutically active compound. Compositions comprising the chimeric polypeptide, either in liquid or dry form, and optionally at least one pharmaceutically acceptable carrier and/or excipient are also specifically considered and are exemplified herein.
[000127] Co-administration can be achieved by separately administering the chimeric polypeptide with the second agent or by administering a single pharmaceutical formulation comprising the chimeric polypeptide and the second agent. Appropriate dosage regimens for the second agents are generally known in the art.
[000128] The preparations may also be co-administered, when desired, with other active substances (eg, to reduce metabolic degradation) as known in the art or other therapeutically active agents.
[000129] Amylins. Amylin is a peptide hormone synthesized by β-cells of the pancreas that is co-secreted with insulin in response to nutrient intake. The amylin sequence is highly conserved across mammalian species, with structural similarities to the calcitonin gene-related peptide (CGRP), calcitonins, intermediins, and adrenomedullin, as known in the art. Glycoregulatory actions of amylin complement those of insulin by regulating the rate of glucose appearance in the circulation via suppression of nutrient-stimulated glucagon secretion and delayed gastric emptying. In patients with diabetes treated with insulin, pramlintide, a synthetic and equipotent analogue of human amylin, reduces postprandial glucose excursions by suppressing inappropriately elevated postprandial glucagon secretion and delaying gastric emptying. Following are the sequences of mouse amylin, human amylin and pramlintide: mouse amylin: KCNTATCATQRLANFLVRSSNNLGPVLPPTNVGSNTY (SEQ ID NO: 86); human amylin: KCNTATCATQRLANFLVHSSNNFGAILSSTNVGSNTY(SEQ ID NO: 87); Pramlintide: KCNTATCATQRLANFLVHSSNNFGPILPPTNVGSNTY (SEQ ID NO: 88).
[000130] Davalintide. Davalintide, also known as "AC-2307", is a potent amylin agonist useful in the treatment of a variety of disease indications. See WO 2006/083254 and WO 2007/114838, each of which is incorporated herein by reference in full and for all intents and purposes. Davalintide is a chimeric peptide that has an N-terminal loop region of amylin or calcitonin and analogues thereof, an alpha-helical region of at least a portion of an alpha-helical region of calcitonin or analogues thereof, or an alpha-helical region of calcitonin or analogues thereof. helical having a portion of an alpha-helical region of amylin and an alpha-helical region of calcitonin or analogues thereof and a C-terminal tail region of amylin or calcitonin. The sequences of human calcitonin, salmon calcitonin, and davalintide follow: human calcitonin: CGNLSTCMLGTYTQDFNKFHTFPQTAIGVGAP (SEQ IDNO: 89); salmon calcitonin: CSNLSTCVLGKLSQELHKLQTYPRTNTGSGTP (SEQ ID NO: 90); davalintide: KCNTATCVLGRLSQELHRLQTYPRTNTGSNTY (SEQ ID NO: 91).
[000131] Without wishing to be bound by any theory, it is believed that amylins and davalintide and fragments and analogues thereof may require C-terminal amidation to trigger a full biological response. It will be understood that amylin compounds, such as those described herein, which include amylins and/or davalintide and fragments and analogs thereof, can be amidated at the C-terminus.
[000132] "Amylin agonist compounds" include native amylin peptides, amylin analog peptides, and other compounds (eg, small molecules) that have amylin agonist activity. "Amylin agonist compounds" may be derived from natural sources, may be synthetic, or may be derived using recombinant DNA techniques. Amylin agonist compounds have amylin agonist receptor binding activity and may comprise amino acids (e.g., natural, unnatural, or a combination thereof), peptide mimetics, chemical moieties, and the like. Those skilled in the art will recognize amylin agonist compounds using amylin receptor binding assays or by measuring amylin agonist activity in soleus muscle assays. In one embodiment, the amylin agonist compounds will have an IC50 of about 200 nM or less, about 100 nM or less, or about 50 nM or less, in an amylin receptor binding assay such as that described herein, in US Patent No. 5,686,411 and US Publication No. 2008/0176804, the disclosures of which are incorporated herein by reference in their entirety and for all intents and purposes. In one embodiment, amylin agonist compounds will have an EC50 of about 20 nM or less, about 15 nM or less, about 10 nM or less, or about 5 nM or less in a soleus muscle assay, as described herein and in US Patent No. 5,686,411. In one embodiment, the amylin agonist compound has at least 90% or 100% sequence identity to human 25,28,29Pro-amylin. In one embodiment, the amylin agonist compound is a peptide chimera of amylin (e.g., human amylin, rat amylin, and the like) and calcitonin (e.g., human calcitonin, salmon calcitonin, and the like). Suitable and exemplary amylin agonist compounds are also described in US Publication No. 2008/0274952, the disclosure of which is incorporated by reference in its entirety and for all intents and purposes.
[000133] By "amylin analogue" as used herein is meant an amylin agonist that has at least 50% sequence identity, preferably at least 70% sequence identity to a naturally occurring form of amylin , whether rat or human, or any other species and is derived therefrom through modifications, including insertions, substitutions, extensions, and/or deletions of the reference amino acid sequence.
[000134] The amylin analog sequence may have at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 25 to 90%, or 95% amino acid sequence identity to the amylin of reference. In one aspect, the analog has 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or even 16 substitutions, insertions, extensions and/or deletions of amino acids relative to the reference compound. In one embodiment, the amylin analog may comprise conservative or non-conservative amino acid substitutions (including unnatural naturally occurring amino acids and the L and D forms). These analogues are preferably peptides, peptide derivatives or peptide mimics. Typical amylin analogues will be peptides, especially of 32-37 amino acids, eg 27-45, especially 28-38 and even 31-36.
[000135] Amylin analogs with identity to rat and human amylin include 25,28,29Pro-h-amylin (pramlintide) (SEQ ID NO: 88); des-1Lys-h-amylin (SEQ ID NO: 111); 25Pro,26Val,28,29Pro-h-amylin (SEQ ID NO: 112); 18Arg,25,28Pro-h-amylin (SEQ ID NO: 113); des-1Lys,18Arg,25,28Pro-h amylin (SEQ ID NO: 114); 18Arg,25,28,29Pro-h-amylin (SEQ ID NO: 115); des-1Lys,18Arg,25,28,29Pro-h-amylin (SEQ ID NO: 116); des-1,Lys25,28,29Pro-h-amylin (SEQ ID NO: 117);25Pro,26Val,28,29Pro-h-amylin (SEQ ID NO: 118); 28Pro-h-amylin, 2,7-Cyclo-[2Asp,7Lys]-h-amylin (SEQ ID NO: 119); 2-37h-amylin (SEQ ID NO: 120); 1Ala-h-amylin (SEQ ID NO: 121); 2Ala-h-amylin (SEQ ID NO: 122); 2,7Ala-h-amylin (SEQ ID NO: 123); 1Ser-h-amylin (SEQ ID NO: 124); 29Pro-h-amylin (SEQ ID NO: 125); 25,28Pro-h-amylin (SEQ ID NO: 126); des-1Lys,25,28Pro-h-amylin (SEQ ID NO: 127); 23Leu,25Pro, 26Val,28,29Pro-h-amylin (SEQ ID NO: 128); 23Leu25Pro26Val28Pro-h-amylin (SEQ ID NO: 129); des-1Lys,23Leu,25Pro,26Val,28Pro-h-amylin (SEQ ID NO: 130); 18Arg,23Leu,25Pro,26Val,28Pro-h-amylin (SEQ ID NO: 131); 18Arg,23Leu, 25,28,29Pro-h-amylin (SEQ ID NO: 132); 18Arg23Leu,25,28Pro-h-amylin (SEQ ID NO: 133); 17Ile,23Leu,25,28,29Pro-h-amylin (SEQ ID NO: 134); 17Ile,25,28,29Pro-h-amylin (SEQ ID NO: 135); des-1Lys,17Ile,23Leu,25,28,29Pro-h-amylin (SEQ ID NO: 136); 17Ile,18Arg,23Leu-h-amylin (SEQ ID NO: 137); 17Ile,18Arg,23Leu, 26Val,29Pro-h-amylin (SEQ ID NO: 138); 17Ile,18Arg,23Leu, 25Pro, 26Val, 28,29Pro-h-amylin (SEQ ID NO: 139); 13Thr,21His,23Leu, 26Ala,28Leu,29Pro, 31Asp-h-amylin (SEQ ID NO: 140); 13Thr,21His,23Leu, 26Ala, 29Pro,31Asp-h-amylin (SEQ ID NO: 141); des-1Lys,13Thr,21His,23Leu,26Ala, 28Pro,31Asp-h-amylin (SEQ ID NO: 142); 13Thr,18Arg,21His,23Leu,26Ala, 29Pro,31Asp-h-amylin (SEQ ID NO: 143); 13Thr,18Arg,21His,23Leu,28,29Pro, 31Asp-h-amylin (SEQ ID NO: 144); and 13Thr,18Arg,21His,23Leu,25Pro,26Ala, 28.29Pro, 31Asp-h-amylin (SEQ ID NO: 145).
[000136] Amylin analogs include amino acid sequences from residues 1-37 of Formula (I) below, wherein up to 25% of the amino acids shown in Formula (I) may be deleted or replaced with a different amino acid: X'-Xaa1 -Cys2-Asn3-Thr4-Ala5-Thr6-Cys7-Ala8-Thr9-Gln10-Arg11-Leu12- Ala13-Asn14-Phe15-Leu16-Val17-His18-Ser19-Ser20- Xaa21-Asn22-Phe23-Xaa24- Xaa25- Xaa26 - Xaa27- Xaa28- Xaa29-Thr30- Xaa31-Val32-Gly33-Ser34-Asn35-Thr36-Tyr37-X (SEQ ID NO: 92) (I).
[000137] In Formula (I), X' represents hydrogen, an N-terminal "capping" group or a linker to a duration-enhancing moiety. Xaa1is Lys or a link, Xaa21is Lys, Cys or Asn, Xaa24is Lys, Cys or Gly, Xaa25is Lys, Cys or Pro, Xaa26is Lys, Cys or Ile, Xaa27is Lys, Cys or Leu, Xaa28is Lys, Cys or Pro, Xaa29is Lys , Cys or Pro and Xaa31 is Lys, Cys or Asn. Still in relation to Formula (I), the variable X represents a C-terminal functionality (for example, a C-terminal "cap"). X is substituted or unsubstituted alkylamino, substituted or unsubstituted amino, substituted or unsubstituted dialkylamino, substituted or unsubstituted cycloalkylamino, substituted or unsubstituted arylamino, substituted or unsubstituted aralkylamino, substituted or unsubstituted aryloxy, substituted or unsubstituted aralkyloxy or hydroxyl. If the C-terminus of the polypeptide component having the sequence of residues 1-37 of Formula (I) is capped with an X functionality, then X is preferably amine, thus forming a C-terminal amide. In some embodiments, more than 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or even 50% of the amino acids from residues 1-37 of Formula (I) are deleted. or substituted by a polypeptide component according to Formula (I). In some embodiments, the amylin analog component has 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or even 16 amino acid substitutions relative to the amino acid sequence shown in Formula (I). In some embodiments, the amylin analog has a sequence that has a defined sequence identity with respect to residues 1-37 of the amino acid sequence according to Formula (I). In some embodiments, the sequence identity between an amylin analog described herein and residues 1-37 of Formula (I) is 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85 %, 90%, 95% or even higher. In some embodiments, more than 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5% or even less of the predicted amino acids at residues 137 of Formula (I) may be deleted or replaced by a different amino acid. In some embodiments, the sequence identity is within the range of 75%-100%. In some embodiments, the sequence identity is within the range of 75%-90%. In some embodiments, the sequence identity is within the range of 80%-90%. In some embodiments, the sequence identity is at least 75%. In some embodiments, the amylin analog has the sequence of residues 1-37 of Formula (I).
[000138] In some embodiments, amylin analogs, including those of formula (I), form the basis of a polypeptide component to which one or more duration-enhancing moieties are attached, optionally via a linker, to form a polypeptide conjugate of amylin. Thus, the polypeptide component functions as a template ("template polypeptide") to which one or more duration-enhancing moieties are linked, preferably through covalent bonding. Attachment of the duration-enhancing moiety to the polypeptide component may be via a linker, as described herein. Alternatively, attachment of the duration-enhancing moiety to the polypeptide component may be via a direct covalent bond. The duration enhancing portion may be a water soluble polymer as described herein. In some embodiments, a plurality of duration-enhancing moieties are linked to the polypeptide component, in which case each linker to each duration-enhancing portion is independently selected from the linkers described herein.
[000139] Amylin analogs useful as polypeptide components described herein include, but are not limited to, the compounds shown at residues 1-37 of Formula (I) shown in Table 3 below. Unless otherwise noted, all peptides described herein, including peptides having an expressly indicated sequence, are considered to be in both free and amidated carboxylate forms.Table 1. Polypeptide Components Useful in Compounds Described herein


[000140] The terms "linker" and the like, in the context of linking duration-enhancing moieties to a polypeptide component in an amylin polypeptide conjugate described herein, means a bivalent (-L-) species covalently linked, in turn, to a polypeptide component having a valence available for binding to a duration-enhancing moiety having a valence available for binding. The available binding site on the polypeptide component is conveniently a side chain residue (e.g. lysine, cysteine, aspartic acid and homologs thereof). In some embodiments, the available binding site on the polypeptide component is the side chain of a lysine or cysteine residue. In some embodiments, the available binding site on the polypeptide component is the N-terminal amine. In some embodiments, the available binding site on the polypeptide component is the C-terminal carboxyl. In some embodiments, the available binding site on the polypeptide component is a backbone atom thereof. As used herein, the term "binding amino acid residue" means an amino acid within residues 1-37 of Formula (I) to which a duration enhancing moiety is attached, optionally via a linker.
[000141] In some embodiments, compounds are provided having a linker that covalently links a polypeptide component with a duration-enhancing moiety. The linker is optional, i.e. any linker can simply be a link. In some embodiments, the linker is attached to a side chain of the polypeptide component. In some embodiments, the linker is attached to an atom of the polypeptide component.
[000142] In another aspect, there is provided an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e., des- Lys1) and an amino acid residue at position 2 to 37 has been replaced by a lysine residue or cysteine residue and wherein said lysine residue or cysteine residue is linked to a polyethylene glycol polymer, optionally via a linker, wherein the amino acid numbering agrees with the amino acid number in SEQ ID NO: 88.
[000143] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. is, des-Lys1) and wherein an amino acid residue at the position of any one of 2, 3,4, 5,6, 7, 8, 9, 10,11, 12, 13, 14,15, 16, 17 , 18,19,20, 21,22,23,24,25,26,27, 5 28, 29, 30, 31, 31, 32, 33, 34, 35, 36 or 37 is replaced by a lysine residue and wherein said lysine residue is linked from a polyethylene glycol polymer, optionally via a linker.
[000144] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. is, des-Lys1) and wherein an amino acid residue at any one of positions 21, 24-29 or 31 is replaced by a lysine residue and wherein said lysine residue is linked to a polyethylene glycol polymer, optionally via of a binder.
[000145] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. i.e. des-Lys1) and wherein an amino acid residue at position 21 is substituted by a lysine residue and wherein said lysine residue is linked to a polyethylene glycol polymer, optionally via a linker.
[000146] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. i.e. des-Lys1) and wherein an amino acid residue at position 24 is substituted by a lysine residue and wherein said lysine residue is linked to a polyethylene glycol polymer, optionally via a linker.
[000147] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. i.e. des-Lys1) and wherein an amino acid residue at position 25 is substituted by a lysine residue and wherein said lysine residue is linked to a polyethylene glycol polymer, optionally via a linker.
[000148] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. i.e. des-Lys1) and wherein an amino acid residue at position 26 is replaced by a lysine residue and wherein said lysine residue is linked to a polyethylene glycol polymer, optionally via a linker.
[000149] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. i.e. des-Lys1) and wherein an amino acid residue at position 27 is replaced by a lysine residue and wherein said lysine residue is linked to a polyethylene glycol polymer, optionally via a linker.
[000150] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. i.e. des-Lys1) and wherein an amino acid residue at position 28 is replaced by a lysine residue and wherein said lysine residue is linked to a polyethylene glycol polymer, optionally via a linker.
[000151] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. i.e. des-Lys1) and wherein an amino acid residue at position 29 is replaced by a lysine residue and wherein said lysine residue is linked to a polyethylene glycol polymer, optionally via a linker.
[000152] In another aspect, the invention relates to an amylin polypeptide conjugate which is a pramlintide derivative of SEQ ID NO: 88 or an analogue thereof, wherein the amino acid residue at position 1 is absent (i.e. i.e. des-Lys1) and wherein an amino acid residue at position 31 is replaced by a lysine residue and wherein said lysine residue is linked to a polyethylene glycol polymer, optionally via a linker.
[000153] In some embodiments, the duration enhancing portion is a water soluble polymer. A "water-soluble polymer" means a polymer that is sufficiently soluble in water under physiological conditions, for example, temperature, ionic concentration, and the like, as known in the art, to be useful in the methods described herein. A water-soluble polymer can increase the solubility of a peptide or other biomolecule to which such a water-soluble polymer is attached. Indeed, such binding has been proposed as a means to improve the circulating life, water solubility and/or antigenicity of administered proteins in vivo. See, for example, U.S. Patent No. 4,179,337; U.S. Published Application No. 2008/0032408. Various water-soluble polymers and fixing chemicals have been used for this purpose, such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxy methyl cellulose, dextran, polyvinyl alcohol, polyvinylpyrrolidone, poly-1,3-dioxolane, poly- 1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyamino acids (homopolymers or random copolymers) and the like.
[000154] In some embodiments, the linked duration enhancing moiety includes a polyethylene glycol. Polyethylene glycol ("PEG") has been used in efforts to obtain therapeutically usable polypeptides. See, for example, Zalipsky, S., 1995, BioconjugateChemistry, 6: 150-165; Mehvar, R., 2000, J. Pharm. Pharmaceut. Sci., 3: 125-136. As appreciated by those skilled in the art, the PEG backbone [(CH2CH2-O-)n, n: number of repeating monomers] is flexible and amphiphilic. Without intending to be bound by any theory or mechanism of action, it is believed that the long chain PEG molecule or moiety is very hydrated and fast moving when in an aqueous medium. This rapid movement is believed to make the PEG cover a large volume and prevent the approach and interference of other molecules. As a result, when linked to another chemical entity (such as a peptide), PEG polymer chains can protect that chemical entity against immune response and other mechanisms of elimination. As a result, pegylation can lead to improved drug safety and efficacy through optimizing pharmacokinetics, increasing bioavailability, and reducing immunogenicity and dosing frequency. "Pegylation" refers, in the usual sense, to the conjugation of a PEG moiety to another compound. For example, PEG binding has been shown to protect proteins against proteolysis. See, for example, Blomhoff, H.K. et al., 1983, Biochim Biophys Acta, 757: 202208 . Unless expressly stated otherwise, the terms "PEG", "polyethylene glycol polymer" and the like refer to polyethylene glycol polymers and derivatives thereof, including methoxy-PEG (mPEG).
[000155] A variety of means have been used to attach polymeric moieties, such as PEG and related polymers, to reactive groups found on the protein. See, for example, U.S. Patent No. 4,179,337; U.S. Patent No. 4,002,531; Abuchowski et al., 1981, in "Enzymes as Drugs", J.S. Holcerberg and J. Roberts, (Eds.), pages 367-383; Zalipsky, S., 1995, Bioconjugate Chemistry, 6: 150-165. The use of PEG and other polymers to modify proteins was discussed. See, for example, Cheng, T.-L. et al., 1999, Bioconjugate Chem., 10: 520-528; Belcheva, N. et al., 1999, Bioconjugate Chem., 10: 932-937; Bettinger, T. et al., 1998, Bioconjugate Chem., 9: 842-846; Huang, S.-Y. et al., 1998, Bioconjugate Chem., 9: 612-617; Xu, B. et al., 1998, Langmuir, 13: 2447-2456; Schwarz, J.B. et al., 1999, J. Amer. Chem. Soc, 121: 2662-2673; Reuter, J.D. et al., 1999, Bioconjugate Chem., 10: 271-278; Chan, T.-H. et al., 1997, J. Org. Chem., 62: 3500-3504. Typical binding sites on proteins include primary amino groups, such as those on lysine residues or on the N-terminus, thiol groups, such as those on cysteine side chains, and carboxyl groups, such as those on glutamate or aspartate residues or on the C-terminus. Common binding sites are to the sugar residues of glycoproteins, cysteines, or to the N-terminus and lysines of the target polypeptide. The terms "pegylated" and the like refer to the covalent linkage of polyethylene glycol to a polypeptide or other biomolecule, optionally through a linker as described herein and/or as known in the art.
[000156] In some embodiments, a PEG moiety in an amylin polypeptide conjugate described herein has a nominal molecular weight within a specified range. As is customary in the art, the size of a PEG moiety is indicated by reference to the nominal molecular weight, typically given in kilodaltons (kD). Molecular weight is calculated in a variety of ways known in the art, including number, weight, viscosity and average molecular weight "Z". It will be understood that polymers such as PEG and the like exist as a molecular weight distribution of a nominal average value.
[000157] Exemplary of terminology for the molecular weight of PEGs, the term "mPEG40KD" refers to a polymer of methoxy polyethylene glycol having a nominal molecular weight of 40 kilodaltons. References to PEGs of other molecular weights follow this convention. In some embodiments, the PEG moiety has a nominal molecular weight in the range of 10-100 KD, 20-80 KD, 20-60 KD, or 2040 KD. In some embodiments, the PEG moiety has a nominal molecular weight of 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or even 100 KD. Preferably, the PEG moiety has a molecular weight of 20, 25, 30, 40, 60 or 80 KD.
[000158] PEG molecules useful for derivatizing polypeptides are generally classified into classes of linear, branched, and Warwick PEG (i.e., PolyPEG®), as known in the art. Unless expressly stated otherwise, the PEG moieties described herein are linear PEGs. Furthermore, the terms "with two branched arms", "Y-shaped" and the like refer to branched PEG moieties as known in the art. The term "Warwick", in the context of PEGs, also known as "comb" or "comb-like" PEGs, refers to a variety of PEGs with multiple arms attached to a skeleton, typically poly(methacrylate), as known in the art. . With respect to nomenclature, including conventions employed in the table provided herein, unless otherwise indicated, a PEG moiety is attached to the peptide backbone. For example, Cmpd 19 is the result of mPEG40KD conjugating to the N-terminal nitrogen of Cmpd 1. Similarly, Cmpd 20 is the result of conjugating mPEG40KD to the N-terminal nitrogen of Cmpd 2. Standard single-letter abbreviations for amino acids can be used, as well as standard three-letter abbreviations. For example, Cmpd 24 is an analog of Cmpd 10, wherein the residue at position 26 of Cmpd 19 is replaced with lysine and the pendant amine functionality of lysine 26 (i.e., K26) is conjugated to a PEG40KD moiety. Exemplary compounds are provided in Table 2 below. Table 2. Pegylated Compounds

Formulations
[000159] The pharmaceutical compounds of 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 described in Remington's Pharmaceutical Sciences by E.W. Martin. See also Wang et al. (1988) J. of Parenteral Sci. and Tech., Technical Report No. 10, Sup. 42: 2S.
[000160] In general, chimeric polypeptides can be formulated into a stable, safe pharmaceutical composition for administration to a patient. Pharmaceutical formulations envisaged for use in the methods of the invention may comprise about 0.01 to 1.0% (w/v), in certain cases 0.05 to 1.0% of the chimeric polypeptide, about 0.02 to 0. 5% (wt/v) of an acetate, phosphate, citrate or glutamate buffer, which allows a pH of the final composition to be between about 3.0 and about 7.0%, approximately 1.0 to 10% ( wt/v) of a carbohydrate toning agent or polyhydric alcohol and optionally approximately 0.005 to 1.0% (wt/v) of a preservative selected from the group consisting of m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol. Such a preservative is generally included if the formulated peptide is to be included in a multipurpose product.
[000161] In particular embodiments, a pharmaceutical formulation of the instant chimeric polypeptides may contain a range of concentrations of compound(s), for example, between about 0.01% to about 98% w/w or between about 1 to about 98% weight/weight or preferably between 80% and 90% weight/weight or preferably between about 0.01% to about 50% weight/weight or more preferably between about 10% at about 25% weight/weight in these modalities. A sufficient amount of water for injection can be used to obtain the desired solution concentration.
[000162] Additional toning agents, such as sodium chloride, as well as other known excipients, may also be present, if desired. In some cases, such excipients are useful in maintaining the overall tonicity of the compound. An excipient can be included in the presently described formulations at various concentrations. For example, an excipient can be included in the concentration range of from about 0.02% to about 20% weight/weight, preferably from about 0.02% to 0.5% weight/weight, from about 0 .02% to about 10% weight/weight or about 1% to about 20% weight/weight. Furthermore, similar to the present formulations themselves, an excipient may be included in solid (including powdered), liquid, semi-solid or gel form.
[000163] Pharmaceutical formulations can be composed in various forms, for example solid, liquid, semi-solid or liquid. The term "solid" as used herein is intended to encompass all normal uses of this term including, for example, powders and lyophilized formulations. The presently described formulations may be lyophilized.
[000164] The terms buffer, buffer solution and buffered solution, when used with reference to the concentration of hydrogen ions or pH, refer to the ability of a system, in particular an aqueous solution, to resist a change in pH when addition of acid or alkali or when diluting with a solvent. Characteristic of buffered solutions, which undergo small changes in pH upon addition of acid or base, is the presence of a weak acid and a salt of the weak acid or a weak base and a salt of the weak base. An example of the above system is acetic acid and sodium acetate. The pH change is small, as long as the amount of hydrogen ions or hydroxyl ions added does not exceed the buffer system's ability to neutralize them.
[000165] As described herein, a variety of liquid carriers are suitable for use in chimeric polypeptide formulations, for example, water or an aqueous/organic solvent mixture or suspension.
[000166] The stability of a chimeric polypeptide formulation for use as described herein is enhanced by maintaining the pH of the formulation within a range determined by methods known in the art. In certain embodiments, the pH of the formulation is maintained in the range of about 3.5 to 5.0 or about 3.5 to 6.5, in some embodiments, from about 3.7 to 4.3 or about 3.8 to 4.2. In some embodiments, the pH can be about 4.0, about 5.0, about 6.0, about 7.0, about 8.0, about 9.0, or even greater. In some embodiments, the pH may be in the physiological range, pH 6-8, preferably pH 7-7.6.
[000167] In certain embodiments, the buffer containing the chimeric polypeptide is an acetate buffer (preferably at a final concentration in the formulation of about 1 -5 to about 60 mM), phosphate buffer (preferably at a final concentration in the formulation from about 1-5 to about 30 mM) or glutamate buffer (preferably at a final concentration in the formulation of about 1-5 to about 60 mM). In some embodiments, the buffer is ethyl acetate (preferably at a final concentration in the formulation of about 5 to about 30 mM).
[000168] A stabilizer may be included in the formulations, but is not necessarily required. If included, however, a stabilizer useful in the practice of the present invention is a carbohydrate or a polyhydric alcohol. A suitable stabilizer useful in the practice of the present invention is approximately 1.0 to 10% (wt/v) of a polyhydric alcohol or carbohydrate. Polyhydric alcohols and carbohydrates share the same characteristic in their backbones, that is, -CHOH-CHOH-, which is responsible for protein stabilization. Polyhydric alcohols include compounds such as sorbitol, glycerol, mannitol and polyethylene glycols (PEG). These compounds are straight chain molecules. Carbohydrates such as mannose, ribose, sucrose, fructose, trehalose, maltose, inositol, and lactose, on the other hand, are cyclic molecules that may contain a keto or aldehyde group. These two classes of compounds have been shown to be effective in stabilizing the protein against denaturation caused by elevated temperature and freeze-thaw processes or lyophilization processes. Suitable carbohydrates include: galactose, arabinose, lactose or any other carbohydrate which does not have an adverse effect on a diabetic patient, ie the carbohydrate is not metabolized to form unacceptably large concentrations of blood glucose. Such carbohydrates are well known in the art to be suitable for diabetics. Sucrose and fructose are suitable for use with the compound in non-diabetic applications (eg, obesity treatment).
[000169] In certain embodiments, if a stabilizer is included, the compound is stabilized with a polyhydric alcohol, such as sorbitol, mannitol, glycerol, inositol, xylitol, and polypropylene/ethylene glycol copolymer, as well as polyethylene glycols (PEG) of various molecular weights - 200, 400, 1450, 3350, 4000, 6000, 8000 and even greater). Mannitol is the preferred polyhydric alcohol in some embodiments. Another useful feature of the lyophilized formulations of the present invention is the maintenance of tonicity of the lyophilized formulations described herein with the same formulation component serving to maintain their stability. In some embodiments, mannitol is the preferred polyhydric alcohol used for this purpose.
[000170] The United States Pharmacopeia (USP) states that antimicrobial agents, in bacteriostatic or fungistatic concentrations, should be added to preparations contained in multi-dose containers. They must be present in an adequate concentration at the time of use to prevent the multiplication of microorganisms inadvertently introduced into the preparation while extracting a portion of the contents with a hypodermic needle and syringe or using other invasive means for delivery, such as injection pens. Antimicrobial agents must be evaluated to ensure compatibility with all other components of the formula and their activity must be evaluated in the total formula to ensure that a particular agent that is effective in one formulation is not ineffective in another. It is not uncommon to find that a particular antimicrobial agent will be effective in one formulation but not in another formulation.
[000171] A preservative is, in the common pharmaceutical sense, a substance that inhibits or prevents microbial growth and can be added to pharmaceutical preparations for this purpose to avoid consequent deterioration of the formulation by microorganisms. Although the amount of preservative is not large, it can nevertheless affect the overall stability of the peptide.
[000172] Although the preservative for use in pharmaceutical compositions may vary from 0.005-1.0% (weight/v), in some embodiments, the range for each preservative, alone or in combination with others, is: benzyl alcohol (0. 1-1.0%) or m-cresol (0.1-0.6%) or phenol (0.1-0.8%) or a combination of methyl (0.050.25%) and ethyl or propyl or butyl (0.005%-0.03%) parabens. Parabens are alkyl esters of parahydroxy-benzoic acid. A detailed description of each preservative is given in Remington's Pharmaceutical Sciences (Id.)
[000173] Chimeric polypeptides may not have a tendency to adsorb onto glass in a glass container when in a liquid form, therefore, a surfactant may not be required to further stabilize the pharmaceutical formulation. However, with regard to compounds that have such a tendency when in liquid form, a surface-active agent must be used in their formulation. These formulations can then be lyophilized. Surfactants often cause protein denaturation through hydrophobic cleavage and salt bridge separation. Relatively low concentrations of surfactant can exert potent denaturing activity because of the strong interactions between surfactant moieties and reactive sites on proteins. However, judicious use of this interaction can stabilize proteins against interfacial or surface denaturation. Surfactants which could further stabilize the chimeric polypeptide may optionally be present in the range of about 0.001 to 0.3% (w/v) of the total formulation and include polysorbate 80 (i.e., polyoxyethylene (20) sorbitan monooleate) , CHAPS® (i.e. 3-[(3-cholamidopropyl)dimethylammonium]-1-propanesulfonate), Brij® (e.g. Brij® 35, which is (polyoxyethylene (23) lauryl ether), poloxamer or other nonionic surfactant.
[000174] Also, it may be desirable to add sodium chloride or other salt to adjust the tonicity of the pharmaceutical composition, according to the toning agent selected. However, this is optional and depends on the particular formulation selected. Parenteral formulations may preferably be isotonic or substantially isotonic.
[000175] A preferred vehicle for parenteral products is water. Water of a quality suitable for parenteral administration can be prepared either by distillation or by reverse osmosis. Water for injection is the preferred aqueous vehicle for use in pharmaceutical formulations.
[000176] It is possible that other components may be present in pharmaceutical formulations. Such additional ingredients may include, for example, humectants, emulsifiers, lubricants, antioxidants, bulking agents, tonicity modifiers, chelating agents, metal ions, oleaginous carriers, proteins (e.g., human serum albumin, gelatin or proteins). ) and a zwitterion (e.g., an amino acid, such as betaine, taurine, arginine, glycine, lysine, and histidine). Additionally, polymer solutions or mixtures with polymers provide the opportunity for controlled release of the peptide. Such additional ingredients, of course, should not adversely affect the overall stability of the pharmaceutical formulation of the present invention.
[000177] Containers are also an integral part of the formulation of an injection and can be considered a component, as there is no container that is totally inert or does not affect in any way the liquid it contains, in particular if the liquid is aqueous. Therefore, the selection of a container for a particular injection should be based on a consideration of the composition of the container, as well as the solution and treatment to which it will be subjected. Adsorption of the peptide to the glass surface of the vial can also be minimized, if necessary, by using boron silicate glass, e.g. Wheaton Type 1 #33 boron silicate glass (Wheaton Type 133) or its equivalent (Wheaton Glass Co.). Other suppliers of similar borosilicate glass vials and cartridges acceptable for manufacture include Kimbel Glass Co., West Co., Bunder Glas GMBH and Form a Vitrum. The biological and chemical properties of the compound can be stabilized by formulating and lyophilizing in a vial of Wheaton Type 1-33 borosilicate serum to a final concentration of 0.1 mg/ml and 10 mg/ml of the compound in the presence of 5% mannitol and 0.02% Tween 80.
[000178] For formulations to be delivered by injection, to allow the introduction of a needle from a hypodermic syringe into a multidose vial and to allow re-closing, as soon as the needle is withdrawn, the open end of each one of the vials is preferably sealed with a rubber stopper held in place by an aluminum strip.
[000179] Stoppers for glass vials such as West 4416/50, 4416/50 (Teflon coated) and 4406/40, Abbott 5139 or any equivalent stopper can be used as the seal for pharmaceuticals for injection. For formulations comprising peptide anti-obesity agents, these stoppers are compatible with the peptide as well as the other components of the formulation. The inventors have also found that these stoppers pass the stopper integrity test when tested using patient wear patterns, for example the stopper can withstand at least about 100 injections. Alternatively, the peptide can be lyophilized into vials, syringes or cartridges for subsequent reconstitution. Liquid formulations of the present invention may be filled into one or two septum cartridges or one or two chambered syringes.
[000180] Each of the components of the pharmaceutical formulation described above is known in the art and is described in Pharmaceutical Dosage Forms: Parenteral Medications, vol. 1, 2nd ed., Avis et al. Ed., Mercel Dekker, New York, NY 1992, which is incorporated by reference in its entirety here and for all intents and purposes.
[000181] The manufacturing process of the liquid formulations above involves, in general, steps of composition, sterile filtration and bottling. The compounding procedure involves dissolving the ingredients in a specific order (preservative followed by stabiliser/toning agents, buffers and peptide) or dissolving at the same time.
[000182] Alternative formulations, eg non-parenteral, may not require sterilization. However, if sterilization is desired or necessary, any suitable sterilization process can be used in developing the peptide pharmaceutical formulation of the present invention. Typical filter sterilization processes include steam (moist heat), dry heat, gases (e.g. ethylene oxide, formaldehyde, chlorine dioxide, propylene oxide, beta-propiolactone, ozone, chloropicrin, methyl bromide, peracetic acid and similar), exposure to a radiation source and aseptic handling. Filtration is the preferred method of sterilization for liquid formulations of the present invention. Sterile filtration involves 0.45 μm and 0.22 μm filters (1 or 2) which can be connected in series. After filtration, the solution is poured into ampoules or other suitable containers.
[000183] In certain embodiments, the chimeric polypeptides described herein are administered peripherally to subjects. In some embodiments, the liquid pharmaceutical formulations of the present invention are for parenteral administration. Suitable routes of administration include intramuscular, intravenous, subcutaneous, intradermal, intra-articular, intrathecal administration and the like. In some embodiments, the subcutaneous route of administration is preferred. In certain embodiments, mucosal delivery is also preferred. These routes include, but are not limited to, oral, nasal, sublingual, buccal and pulmonary routes, which may include administration of the peptide in liquid, semi-solid or solid form. For formulations comprising the chimeric polypeptides, administration via these routes may require substantially more compound to obtain the desired biological effects by virtue of decreased bioavailability compared to parenteral delivery. In addition, parenteral delivery with controlled release can be achieved by forming polymeric microcapsules, matrices, solutions, implants and devices and administering them parenterally or through surgical means. Examples of controlled release formulations are described in U.S. Patent Nos. 6,368,630, 6,379,704 and 5,766,627, which are incorporated herein by reference. These dosage forms may have lower bioavailability due to retention of a portion of the peptide in the polymer matrix or device. See, for example, U.S. Patent Nos. 6,379,704, 6,379,703 and 6,296,842, each of which is incorporated herein by reference in full and for all intents and purposes.
[000184] The compounds may be provided in unit dosage form containing an amount of the chimeric polypeptide that will be effective in single or multiple doses.
[000185] As will be appreciated by those skilled in the art, an effective amount of the chimeric polypeptide will vary depending on many factors, including the age and weight of the subject, the physical condition of the subject, the condition being treated, and other factors known in the art. . An effective amount of the chimeric polypeptides will also vary with the particular combination administered. As described herein, administration of the chimeric polypeptides in combination may allow for a decrease in the amount of any of the modified polypeptides to be administered in an effective amount. Effective Dosages
[000186] Pharmaceutical compositions provided in the present invention include compositions wherein the active ingredient is contained in a therapeutically effective amount, i.e., in an amount effective to achieve its intended purpose. The actual effective amount for a particular application will depend, inter alia, on the condition being treated. For example, when administered in methods of treating diabetes, such compositions will contain an amount of active ingredient effective to achieve the desired result (e.g., lowering a subject's fasting blood glucose). When administered in methods of treating obesity, such compositions will contain an amount of active ingredient effective to achieve the desired result (e.g., reduction in body mass).
[000187] The dosage and frequency (single or multiple doses) of the administered compound may vary depending on a variety of factors, including the route of administration, size, age, sex, health, body weight, body mass index and diet of the patient. recipient, the nature and extent of symptoms of the disease being treated (e.g., the disease responds to the compounds described herein), presence of other diseases or other health-related problems; type of concomitant treatment and complications from any disease or treatment regimen. Other therapeutic regimens or agents can be used in conjunction with the methods and compounds of the invention.
[000188] Therapeutically effective amounts for use in humans can be determined from animal models. For example, a dose for humans can be formulated to obtain a concentration found to be effective in animals. Dosage in humans can be adjusted by monitoring one or more physiological parameters including, but not limited to, blood sugar and body mass, and adjusting dosages up or down, as described above and known in the art.
[000189] Dosages can be varied depending on the requirements of the patient and the compound being employed. The dose administered to a patient, in the context of the present invention, should be sufficient to obtain a beneficial therapeutic response in the patient over time. Dose size will also be determined by the existence, nature and extent of any adverse side effects. In general, treatment is started with lower dosages, which are less than the optimal dose of the compound. After which, the dosage is increased in small increments until the optimum effect under the circumstances is reached. In one embodiment of the invention, the dosage range is from 0.001% to 10% w/v. In another embodiment, the dosage range is 0.1% to 5% w/v.
[000190] However, typical doses may contain from a low limit of about 0.1 mg to a high limit of about 200 mg of the pharmaceutical compound per day. Also considered are other dose ranges, such as from 1mg to 100mg of the compound per dose and from 3mg to 70mg per dose. Typically, the dose of long-acting chimeric polypeptides is administered, for example, daily or even once a week. The daily doses may be distributed in separate unit doses, given continuously over a 24 hour period or any portion of these 24 hours.
[000191] Dosage amounts and intervals may be individually adjusted to provide levels of the administered compound effective for the particular clinical indication being treated. This will provide a therapeutic regimen that is commensurate with the severity of the subject's disease state.
[000192] Using the teachings provided herein, an effective prophylactic or therapeutic treatment regimen can be devised which does not cause substantial toxicity and yet is entirely effective in treating the clinical symptoms demonstrated by the particular patient. Such planning should involve careful choice of the active compound by considering factors such as compound potency, relative bioavailability, patient body weight, the presence and severity of adverse side effects, the preferred mode of administration, and the toxicity profile of the selected agent. toxicity
[000193] The relationship between toxicity and therapeutic effect for a particular compound is its therapeutic index and can be expressed as the ratio between the LD50 (the amount of lethal compound in 50% of the population) and the ED50 (the amount of effective compound in 50% of the population). Compounds that exhibit high therapeutic indices are preferred. Therapeutic index data obtained from cell culture assays and/or animal studies can be used in formulating a range of dosages for use in humans. The dosage of such compounds preferably lies within a range of plasma concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range, depending on the dosage form employed and the route of administration used. See, for example, Fingi et al., in: The Pharmacological Basis of Therapeutics, Chapter 1, p. 1, 1975. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the condition of the patient and the method in which the particular compound is used. Examples Example 1: Chimeric polypeptide recovery
[000194] Protein sequences were designed and retranslated using commercial DNA sequence software for cloning into an E. coli expression vector. Sequences were obtained as oligonucleotides and ligated together using standard PCR amplification techniques or digested from existing expression constructs using standard restriction enzymes and then ligated back together. Sequences expressing the protein of interest were placed in pET45 with a T7 promoter for inducible expression. After the constructs were verified through sequencing, the vector DNA was purified and transformed into an expression host, typically BL21 (DE3). A single colony was selected for growth of a starter culture in 4 ml of LB medium for ~6 hours. Glycerol stocks were prepared by adding 100 μl of 80% glycerol to 900 μl of the stock and stored at -80 °C. Optionally, 500 μl of uninduced sample was retained for gel analysis. A 60 ml culture (Magic medium) was inoculated with 60 μl of starter culture in a 125 ml Thompson flask and incubated @ 30 °C overnight. Remove 250 μl of sample for analysis. Centrifuge and freeze the cell pellet for further processing.
[000195] Bacterial cells were collected and subsequently lysed to isolate inclusion bodies. Once the protein was present in the inclusion bodies, they were solubilized and the protein refolded at 4°C. The proteins were then separated using size exclusion chromatography until only a single band remained and endotoxin levels were acceptable for in vivo testing. Analytical HPLC, RP-LC-MS and SDS-PAGE gel were performed as quality control measures on the final protein. Protein was distributed in predetermined aliquots and stored at -80°C. Example 2: Biological and pharmaceutical properties
[000196] As shown in Table 3 below, chimeric polypeptides described herein have comparable and some even superior properties compared to A100 (Compound 37, SEQ ID NO: 24). These properties include biological properties, such as leptin binding activity, functional activity of leptin and food intake in rats, and pharmaceutical properties, such as solubility at neutral pH.
[000197] Exemplary assays for leptin binding activity and leptin functional activity have been previously described.
[000198] The food intake activity in mice was tested with the following assay: female C57BL6 mice and their food were weighed daily 3 hours before lights out. Immediately after weighing, on days 0, 1, 2 and 3, mice were injected SC with the leptin compound or mutant in 1xPBS. Points represent means ± SD of n = 9 cages (three mice/cage). The results, reported under "Food Consumption by Mouse" in Table 3, correspond to the vehicle-corrected % change in body weight measured after Day 4.
[000199] Solubility was measured with the following assay: proteins were concentrated at 4°C, centrifuged to remove precipitates, then allowed to equilibrate at room temperature overnight. They were filtered to remove precipitates and then the concentration was determined by measuring the absorbance at OD280 and using the theoretical molar extinction coefficient.Table 3. Biological and pharmaceutical properties of chimeric polypeptides
*These numbers do not necessarily represent the maximum solubility of each compound. Example 3: Stability of chimeric polypeptides
[000200] As shown in Table 4 below, the chimeric polypeptides described herein have comparable and some even superior physical stability compared to A100 (Compound 37, SEQ ID NO: 24). Compounds were formulated in the following buffer: 10 mM glutamic acid, 2% glycine, 1% sucrose, 0.01% Tween 20, pH 4.25 and stored at 37°C. Samples were collected at T = 0, 2, 5, 7 and 14 days and tested by means of visual analysis, reversed-phase High Performance Liquid Chromatography - HPLC, UV spectrometry and dynamic scattering of light (Dynamic Light Scattering - DLS). As shown in Table 4, the chimeric polypeptides have comparable or greater purity and potency compared to Compound 37.Table 4. Stability of chimeric polypeptides
*relative UV normalized power at T = 0 **relative RP-HPLC normalized purity at T = 0 ***UV power - LC purity (total solubles - soluble grades) Example 4: Change in body weight after daily administration of chimeric polypeptide.
[000201] Method. Female C57BL6 mice and their food were weighed daily 3 hours before lights out. Immediately after weighing, on days 0, 1, 2 and 3, mice were injected SC with leptin compound or mutant in 1xPBS. The points represent the mean ± SD of n = 4 cages (3 mice/cage). Each group (n = 12/group) was assigned to receive one of the following: vehicle; Cmpd 41 at 0.1 mg/kg; Cmpd 41 at 0.3 mg/kg; Cmpd 41 at 1 mg/kg; Cmpd 41 at 3 mg/kg; Cmpd 41 at 5 mg/kg; Cmpd 41 at 10 mg/kg. Food intake and body weight change (% corrected for vehicle) were monitored for 4 days and results recorded as illustrated (Figures 1A and 1B). Points represent the mean ± SD of n = 4 cages (3 mice/cage). Compounds Administered: Vehicle (solid circle); Cmpd 41 at 0.1 mg/kg (tip-down triangle); Cmpd 41 at 0.3 mg/kg (empty diamonds); Cmpd 41 at 1 mg/kg (open circles); Cmpd 41 at 3 mg/kg (upward-pointed triangle); Cmpd 41 at 5 mg/kg (star); Cmpd 41 at 10 mg/kg (solid diamond).
[000202] Results. As illustrated in Figures 1A and 1B, administration of different doses of the chimeric polypeptide resulted in reduced food intake and body weight relative to the vehicle-only group. A dose-response is seen in Figure 1C. Example 5: Change in body weight after daily administration of chimeric polypeptide.
[000203] Method. C57BL6e female mice were weighed daily 3 hours before lights out. Immediately after weighing, on days 0, 1, 2 and 3, mice were injected SC with leptin compound or mutant in 1xPBS. The points represent the mean ± SD of n = 4 cages (3 mice/cage). Each group (n = 12/group) was assigned to receive one of the following: vehicle; Cmpd 42 at 0.1 mg/kg; Cmpd 42 at 0.3 mg/kg; Cmpd 42 at 1 mg/kg; Cmpd 42 at 3 mg/kg; Cmpd 42 at 5 mg/kg; Cmpd 42 at 10 mg/kg. Food intake and body weight change (% corrected for vehicle) were monitored for 4 days and results recorded as illustrated (Figures 2A and 2B). Points represent the mean ± SD of n = 4 cages (3 mice/cage). Compounds Administered: Vehicle (solid circle); Cmpd 42 at 0.1 mg/kg (tip-up triangle); Cmpd 42 at 0.3 mg/kg (tip-down triangle); Cmpd 42 at 1 mg/kg (solid square); Cmpd 42 at 3 mg/kg (bar above and dot below); Cmpd 42 at 5 mg/kg (star); Cmpd 42 at 10 mg/kg (bar above point).
[000204] Results. As depicted in Figures 2A and 2B, administration of different doses of the chimeric polypeptide resulted in reduced food intake and body weight relative to the vehicle-only group. A dose-response is seen in Figure 2C.
[000205] As shown in Table 5 below, the dose responses measured for the chimeric polypeptides of the invention are comparable to the dose responses measured for seal leptin (Cmpd 38) and human leptin (Cmpd 37) from which the polypeptides chimeric are derivatives.Table 5. Dose-responses of chimeric polypeptides
VIII. Modalities
[000206] Additional modalities of chimeric polypeptides, method of use thereof and pharmaceutical compositions described here are as follows:
[000207] Embodiment 1. A chimeric polypeptide comprising a wild-type seal leptin polypeptide wherein at least one contiguous region of 1-30 amino acids of a wild-type seal leptin sequence has been replaced by a contiguous region of 1 -30 amino acids of a mature human leptin sequence.
[000208] Embodiment 2. The chimeric polypeptide according to Embodiment 1, wherein two or more contiguous regions of 1-30 amino acids of a wild-type seal leptin sequence have been replaced in each region by a contiguous region of 1 -30 amino acids of a mature human leptin sequence.
[000209] Embodiment 3. The chimeric polypeptide according to Embodiment 1 or 2, wherein a wild-type seal leptin sequence comprises the amino acid sequence of SEQ ID NO: 28 or SEQ ID NO: 31.
[000210] Embodiment 4. The chimeric polypeptide according to any one of Embodiments 1-3, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51.
[000211] Embodiment 5. The chimeric polypeptide according to any one of Embodiments 1-4, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least one amino acid substitution at a position where divergence is observed at a corresponding position in a leptin from another species.
[000212] Embodiment 6. The chimeric polypeptide according to any one of Embodiments 1-5, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least two amino acid substitutions at positions where divergence is observed at corresponding positions in a leptin from another species.
[000213] Embodiment 7. The chimeric polypeptide according to any one of Embodiments 1-6, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least three amino acid substitutions at positions where divergence is observed at corresponding positions in a leptin from another species.
[000214] Embodiment 8. The chimeric polypeptide according to any one of Embodiments 1-7, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least four amino acid substitutions at positions where divergence is observed at corresponding positions in a leptin from another species.
[000215] Embodiment 9. The chimeric polypeptide according to any one of Embodiments 1-8, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least five amino acid substitutions at positions where divergence is observed at corresponding positions in a leptin from another species.
[000216] Embodiment 10. The chimeric polypeptide according to any one of Embodiments 1-9, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least six amino acid substitutions at positions where divergence is observed at corresponding positions in a leptin from another species.
[000217] Embodiment 11. The chimeric polypeptide according to any one of Embodiments 1-10, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least seven amino acid substitutions at positions where divergence is observed at corresponding positions in a leptin from another species.
[000218] Embodiment 12. The chimeric polypeptide according to any one of Embodiments 1-11, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least eight amino acid substitutions at positions where divergence is observed at corresponding positions in a leptin from another species.
[000219] Embodiment 13. The chimeric polypeptide according to any one of Embodiments 1-12, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least nine amino acid substitutions at positions where divergence is observed at corresponding positions in a leptin from another species.
[000220] Embodiment 14. The chimeric polypeptide according to any one of Embodiments 1-13, wherein a mature human leptin sequence comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50 and SEQ ID NO: 51, wherein the mature human leptin sequence has at least ten amino acid substitutions at positions where divergence is observed at corresponding positions in a leptin from another species.
[000221] Embodiment 15. The chimeric polypeptide according to any one of Embodiments 1-14, wherein the mature human leptin sequence comprises the amino acid sequence of SEQ ID NO: 24.
[000222] Embodiment 16. The chimeric polypeptide according to any one of Embodiments 1-15, wherein the chimeric polypeptide comprises an amino acid sequence having at least 80% identity to an amino acid sequence selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO : 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64 , SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 68, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, SEQ ID NO: 74, SEQ ID NO: 75, SEQ ID NO: 76, SEQ ID NO: 77, SEQ ID NO: 78, SEQ ID NO: 79, SEQ ID NO: 80, ID NO: 81, SEQ ID NO: 82, SEQ ID NO: 83, SEQ ID NO: 84 and SEQ ID NO: 85.
[000223] Embodiment 17. The chimeric polypeptide according to any one of Embodiments 1-16, wherein the chimeric polypeptide comprises an amino acid sequence having at least 80% identity to an amino acid sequence selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33.
[000224] Embodiment 18. The chimeric polypeptide according to any one of Embodiments 1-17, wherein the chimeric polypeptide comprises an amino acid sequence having at least 80% identity to the amino acid sequence of SEQ ID NO: 33.
[000225] Embodiment 19. The chimeric polypeptide according to any one of Embodiments 1-18, wherein the chimeric polypeptide comprises an amino acid sequence having at least 90% identity to the amino acid sequence of SEQ ID NO: 33.
[000226] Embodiment 20. The chimeric polypeptide according to any one of Embodiments 1-19, wherein the chimeric polypeptide comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO : 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66 , SEQ ID NO: 67, SEQ ID NO: 68, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, SEQ ID NO: 74, SEQ ID NO: 75, SEQ ID NO: 76, SEQ ID NO: 77, SEQ ID NO: 78, SEQ ID NO: 79, SEQ ID NO: 80, ID NO:81, SEQ ID NO: 82, SEQ ID NO: 83, SEQ ID NO: 84 and SEQ ID NO: 85.
[000227] Embodiment 21. The chimeric polypeptide according to any one of Embodiments 1-20, wherein the chimeric polypeptide comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33.
[000228] Embodiment 22. The chimeric polypeptide according to any one of Embodiments 1-21, wherein the chimeric polypeptide comprises the amino acid sequence of SEQ ID NO: 29.
[000229] Embodiment 23. The chimeric polypeptide according to any one of Embodiments 1-21, wherein the chimeric polypeptide comprises the amino acid sequence of SEQ ID NO: 30.
[000230] Embodiment 24. The chimeric polypeptide according to any one of Embodiments 1-21, wherein the chimeric polypeptide comprises the amino acid sequence of SEQ ID NO: 32.
[000231] Embodiment 25. The chimeric polypeptide according to any one of Embodiments 1-21, wherein the chimeric polypeptide comprises the amino acid sequence of SEQ ID NO: 33.
[000232] Embodiment 26. A method for treating a disease or disorder in a subject comprising administering a chimeric polypeptide according to any one of Embodiments 1 to 25 to a subject in need thereof in an amount effective to treat said disease or disorder.
[000233] Mode 27. The method according to Mode 26, wherein the disease or disorder is disease or disorder is selected from the group consisting of: lipodystrophy, dyslipidemia, hyperlipidemia, overweight, obesity, hypothalamic amenorrhea, Alzheimer's disease , leptin deficiency, fatty liver disease, diabetes (including type I and type II), non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver disease (NAFLD), metabolic syndrome X, and Huntington's disease.
[000234] Embodiment 28. The method according to Embodiment 26 or Embodiment 27, wherein the disease or disorder is lipodystrophy, dyslipidemia, hyperlipidemia, overweight, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin deficiency, liver disease fat or diabetes.
[000235] Embodiment 29. The method according to any one of Embodiments 26-28, wherein the disease or disorder is type I diabetes or type II diabetes.
[000236] Embodiment 30. The method according to any one of Embodiments 26-28, wherein the disease or disorder is obesity.
[000237] Embodiment 31. The method according to any one of Embodiments 26-28, wherein the disease or disorder is lipodystrophy or leptin deficiency.
[000238] Embodiment 32. A pharmaceutical composition comprising a chimeric polypeptide according to any one of Embodiments 1-25 and a pharmaceutically acceptable excipient.
[000239] Modality 33. The method according to any of Modalities 1 to 32, wherein weight loss is reduced by at least 10%.
[000240] Modality 34. The method according to any of Modalities 1 to 33, wherein weight loss is reduced by at least 12%.
[000241] Modality 35. The method according to any of Modalities 1 to 34, wherein weight loss is reduced by at least 15%. B. Formulations
[000242] The pharmaceutical compounds of 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 described in Remington's Pharmaceutical Sciences by E.W. Martin. See also Wang et al. (1988) J. of Parenteral Sci. and Tech., Technical Report No. 10, Sup. 42: 2S.
[000243] In general, engineered polypeptides can be formulated into a stable, safe pharmaceutical composition for administration to a patient. Pharmaceutical formulations envisaged for use in the methods of the invention may comprise about 0.01 to 1.0% (w/v), in certain cases 0.05 to 1.0% of the manipulated polypeptide, about 0.02 to 0. 5% (wt/v) of an acetate, phosphate, citrate or glutamate buffer, which allows a pH of the final composition to be between about 3.0 and about 7.0%, approximately 1.0 to 10% ( wt/v) of a carbohydrate toning agent or polyhydric alcohol and optionally approximately 0.005 to 1.0% (wt/v) of a preservative selected from the group consisting of m-cresol, benzyl alcohol, methyl, ethyl, propyl and butyl parabens and phenol. Such a preservative is generally included if the formulated peptide is to be included in a multipurpose product.
[000244] In particular embodiments, a pharmaceutical formulation of the present engineered polypeptides may contain a range of concentrations of compound(s), for example, between about 0.01% to about 98% w/w or between about 1 to about 98% weight/weight or preferably between 80% and 90% weight/weight or preferably between about 0.01% to about 50% weight/weight or more preferably between about 10% at about 25% weight/weight in these modalities. A sufficient amount of water for injection can be used to obtain the desired solution concentration.
[000245] Additional toning agents, such as sodium chloride, as well as other known excipients, may also be present, if desired. In some cases, such excipients are useful in maintaining the overall tonicity of the compound. An excipient can be included in the presently described formulations at various concentrations. For example, an excipient can be included in the concentration range of from about 0.02% to about 20% weight/weight, preferably from about 0.02% to 0.5% weight/weight, from about 0 .02% to about 10% weight/weight or about 1% to about 20% weight/weight. Furthermore, similar to the present formulations themselves, an excipient may be included in solid (including powdered), liquid, semi-solid or gel form.
[000246] Pharmaceutical formulations can be composed in various forms, for example solid, liquid, semi-solid or liquid. The term "solid" as used herein is intended to encompass all normal uses of this term including, for example, powders and lyophilized formulations. The presently described formulations may be lyophilized.
[000247] The terms buffer, buffer solution and buffered solution, when used with reference to the concentration of hydrogen ions or pH, refer to the ability of a system, in particular an aqueous solution, to resist a change in pH when addition of acid or alkali or when diluting with a solvent. Characteristic of buffered solutions, which undergo small changes in pH upon addition of acid or base, is the presence of a weak acid and a salt of the weak acid or a weak base and a salt of the weak base. An example of the above system is acetic acid and sodium acetate. The pH change is small, as long as the amount of hydrogen ions or hydroxyl ions added does not exceed the buffer system's ability to neutralize them.
[000248] As described herein, a variety of liquid carriers are suitable for use in manipulated polypeptide formulations, for example, water or an aqueous/organic solvent mixture or suspension.
[000249] The stability of an engineered polypeptide formulation for use as described herein is enhanced by maintaining the pH of the formulation within a range determined by methods known in the art. In certain embodiments, the pH of the formulation is maintained in the range of about 3.5 to 5.0 or about 3.5 to 6.5, in some embodiments, from about 3.7 to 4.3 or about 3.8 to 4.2. In some embodiments, the pH can be about 4.0, about 5.0, about 6.0, about 7.0, about 8.0, about 9.0, or even greater. In some embodiments, the pH may be in the physiological range, pH 6-8, preferably pH 7-7.6.
[000250] In certain embodiments, the buffer with the manipulated polypeptide is an acetate buffer (preferably at a final concentration in the formulation of about 1 -5 to about 60 mM), phosphate buffer (preferably at a final concentration in the formulation from about 1-5 to about 30 mM) or glutamate buffer (preferably at a final concentration in the formulation of about 1-5 to about 60 mM). In some embodiments, the buffer is ethyl acetate (preferably at a final concentration in the formulation of about 5 to about 30 mM).
[000251] A stabilizer may be included in the formulations, but is not necessarily required. If included, however, a stabilizer useful in the practice of the present invention is a carbohydrate or a polyhydric alcohol. A suitable stabilizer useful in the practice of the present invention is approximately 1.0 to 10% (wt/v) of a polyhydric alcohol or carbohydrate. Polyhydric alcohols and carbohydrates share the same characteristic in their backbones, that is, -CHOH-CHOH-, which is responsible for protein stabilization. Polyhydric alcohols include compounds such as sorbitol, glycerol, mannitol and polyethylene glycols (PEG). These compounds are straight chain molecules. Carbohydrates such as mannose, ribose, sucrose, fructose, trehalose, maltose, inositol, and lactose, on the other hand, are cyclic molecules that may contain a keto or aldehyde group. These two classes of compounds have been shown to be effective in stabilizing the protein against denaturation caused by elevated temperature and freeze-thaw processes or lyophilization processes. Suitable carbohydrates include: galactose, arabinose, lactose or any other carbohydrate which does not have an adverse effect on a diabetic patient, ie the carbohydrate is not metabolized to form unacceptably large concentrations of blood glucose. Such carbohydrates are well known in the art to be suitable for diabetics. Sucrose and fructose are suitable for use with the compound in non-diabetic applications (eg, obesity treatment).
[000252] In certain embodiments, if a stabilizer is included, the compound is stabilized with a polyhydric alcohol, such as sorbitol, mannitol, glycerol, inositol, xylitol, and polypropylene/ethylene glycol copolymer, as well as polyethylene glycols (PEG) of various molecular weights - 200, 400, 1450, 3350, 4000, 6000, 8000 and even greater). Mannitol is the preferred polyhydric alcohol in some embodiments. Another useful feature of the lyophilized formulations of the present invention is the maintenance of tonicity of the lyophilized formulations described herein with the same formulation component serving to maintain their stability. In some embodiments, mannitol is the preferred polyhydric alcohol used for this purpose.
[000253] The United States Pharmacopeia (USP) states that antimicrobial agents, in bacteriostatic or fungistatic concentrations, should be added to preparations contained in multi-dose containers. They must be present in an adequate concentration at the time of use to prevent the multiplication of microorganisms inadvertently introduced into the preparation while extracting a portion of the contents with a hypodermic needle and syringe or using other invasive means for delivery, such as injection pens. Antimicrobial agents must be evaluated to ensure compatibility with all other components of the formula and their activity must be evaluated in the total formula to ensure that a particular agent that is effective in one formulation is not ineffective in another. It is not uncommon to find that a particular antimicrobial agent will be effective in one formulation but not in another formulation.
[000254] A preservative is, in the common pharmaceutical sense, a substance that inhibits or prevents microbial growth and can be added to pharmaceutical preparations for this purpose to avoid consequent deterioration of the formulation by microorganisms. Although the amount of preservative is not large, it can nevertheless affect the overall stability of the peptide.
[000255] Although the preservative for use in pharmaceutical compositions may vary from 0.005-1.0% (weight/v), in some embodiments, the range for each preservative, alone or in combination with others, is: benzyl alcohol (0. 1-1.0%) or m-cresol (0.1-0.6%) or phenol (0.1-0.8%) or a combination of methyl (0.050.25%) and ethyl or propyl or butyl (0.005%-0.03%) parabens. Parabens are alkyl esters of parahydroxy-benzoic acid. A detailed description of each preservative is given in Remington's Pharmaceutical Sciences (Id.)
[000256] Engineered polypeptides may not have a tendency to adsorb onto glass in a glass container when in a liquid form, therefore, a surfactant may not be required to further stabilize the pharmaceutical formulation. However, with regard to compounds that have such a tendency when in liquid form, a surface-active agent must be used in their formulation. These formulations can then be lyophilized. Surfactants often cause protein denaturation through hydrophobic cleavage and salt bridge separation. Relatively low concentrations of surfactant can exert potent denaturing activity because of the strong interactions between surfactant moieties and reactive sites on proteins. However, judicious use of this interaction can stabilize proteins against interfacial or surface denaturation. Surfactants which could further stabilize the engineered polypeptide may optionally be present in the range of about 0.001 to 0.3% (w/v) of the total formulation and include polysorbate 80 (i.e., polyoxyethylene (20) sorbitan monooleate) , CHAPS® (i.e., 3-[(3-cholamidopropyl)dimethylammonium]-1-propanesulfonate), Brij® (e.g., Brij® 35, which is (polyoxyethylene (23) lauryl ether), poloxamer or another nonionic surfactant.
[000257] Also, it may be desirable to add sodium chloride or other salt to adjust the tonicity of the pharmaceutical composition, according to the toning agent selected. However, this is optional and depends on the particular formulation selected. Parenteral formulations may preferably be isotonic or substantially isotonic.
[000258] A preferred vehicle for parenteral products is water. Water of a quality suitable for parenteral administration can be prepared either by distillation or by reverse osmosis. Water for injection is the preferred aqueous vehicle for use in pharmaceutical formulations.
[000259] It is possible that other components may be present in pharmaceutical formulations. Such additional ingredients may include, for example, humectants, emulsifiers, lubricants, antioxidants, bulking agents, tonicity modifiers, chelating agents, metal ions, oleaginous carriers, proteins (e.g., human serum albumin, gelatin or proteins). ) and a zwitterion (e.g., an amino acid, such as betaine, taurine, arginine, glycine, lysine, and histidine). Additionally, polymer solutions or mixtures with polymers provide the opportunity for controlled release of the peptide. Such additional ingredients, of course, should not adversely affect the overall stability of the pharmaceutical formulation of the present invention.
[000260] Containers are also an integral part of the formulation of an injection and can be considered a component, as there is no container that is totally inert or does not affect in any way the liquid it contains, in particular if the liquid is aqueous. Therefore, the selection of a container for a particular injection should be based on a consideration of the composition of the container, as well as the solution and treatment to which it will be subjected. Adsorption of the peptide to the glass surface of the vial can also be minimized, if necessary, by using borosilicate glass, e.g. Wheaton Type 1 #33 borosilicate glass (Wheaton Type 133) or its equivalent (Wheaton Glass Co.). Other suppliers of similar borosilicate glass vials and cartridges acceptable for manufacture include Kimbel Glass Co., West Co., Bunder Glas GMBH and Form a Vitrum. The biological and chemical properties of the compound can be stabilized by formulating and lyophilizing in a Wheaton Type 1-33 borosilicate serum vial to a final concentration of 0.1 mg/ml and 10 mg/ml of the compound in the presence of 5% mannitol and 0.02% Tween 80.
[000261] For formulations to be delivered by injection, to allow the introduction of a needle from a hypodermic syringe into a multidose vial and to allow re-closing, as soon as the needle is withdrawn, the open end of each one of the vials is preferably sealed with a rubber stopper held in place by an aluminum strip.
[000262] Glass vial stoppers such as West 4416/50, 4416/50 (Teflon coated) and 4406/40, Abbott 5139 or any equivalent stopper can be used as the seal for pharmaceuticals for injection. For formulations comprising peptide anti-obesity agents, these stoppers are compatible with the peptide as well as the other components of the formulation. The inventors have also found that these stoppers pass the stopper integrity test when tested using patient wear patterns, for example the stopper can withstand at least about 100 injections. Alternatively, the peptide can be lyophilized into vials, syringes or cartridges for subsequent reconstitution. Liquid formulations of the present invention may be filled into one or two septum cartridges or one or two chambered syringes.
[000263] Each of the components of the pharmaceutical formulation described above is known in the art and is described in PHARMACEUTICAL DOSAGE FORMS: PARENTERAL MEDICATIONS, vol. 1, 2nd ed., Avis et al. Ed., Mercel Dekker, New York, NY 1992, which is incorporated by reference in its entirety here and for all intents and purposes.
[000264] the manufacturing process of the above liquid formulations involves, in general, steps of composition, sterile filtration and potting. The compounding procedure involves dissolving the ingredients in a specific order (preservative followed by stabiliser/toning agents, buffers and peptide) or dissolving at the same time.
[000265] Alternative formulations, eg non-parenteral, may not require sterilization. However, if sterilization is desired or necessary, any suitable sterilization process can be used in developing the peptide pharmaceutical formulation of the present invention. Typical filter sterilization processes include steam (moist heat), dry heat, gases (e.g. ethylene oxide, formaldehyde, chlorine dioxide, propylene oxide, beta-propiolactone, ozone, chloropicrin, methyl bromide, peracetic acid and similar), exposure to a radiation source and aseptic handling. Filtration is the preferred method of sterilization for liquid formulations of the present invention. Sterile filtration involves 0.45 μm and 0.22 μm filters (1 or 2) which can be connected in series. After filtration, the solution is poured into ampoules or other suitable containers.
[000266] In certain embodiments, the engineered polypeptides described herein are administered peripherally to subjects. In some embodiments, the liquid pharmaceutical formulations of the present invention are for parenteral administration. Suitable routes of administration include intramuscular, intravenous, subcutaneous, intradermal, intra-articular, intrathecal administration and the like. In some embodiments, the subcutaneous route of administration is preferred. In certain embodiments, mucosal delivery is also preferred. These routes include, but are not limited to, oral, nasal, sublingual, buccal and pulmonary routes, which may include administration of the peptide in liquid, semi-solid or solid form. For formulations comprising the engineered polypeptides, administration via these routes may require substantially more compound to obtain the desired biological effects by virtue of decreased bioavailability compared to parenteral delivery. In addition, parenteral delivery with controlled release can be achieved by forming polymeric microcapsules, matrices, solutions, implants and devices and administering them parenterally or through surgical means. Examples of controlled release formulations are described in U.S. Patent Nos. 6,368,630, 6,379,704 and 5,766,627, which are incorporated herein by reference. These dosage forms may have lower bioavailability due to retention of a portion of the peptide in the polymer matrix or device. See, for example, U.S. Patent Nos. 6,379,704, 6,379,703 and 6,296,842, each of which is incorporated herein by reference in full and for all intents and purposes.
[000267] The compounds may be provided in unit dosage form containing an amount of engineered polypeptide that will be effective in single or multiple doses.
[000268] As will be appreciated by those skilled in the art, an effective amount of the manipulated polypeptide will vary depending on many factors, including the age and weight of the subject, the physical condition of the subject, the condition being treated, and other factors known in the art. . An effective amount of the manipulated polypeptides will also vary with the particular combination administered. As described herein, administration of the engineered polypeptides in combination may allow for a decrease in the amount of any of the modified polypeptides to be administered in an effective amount.
[000269] The long duration of action of the engineered polypeptide can confer the desired prolonged duration of action, such as once-daily or once-weekly administration. The duration of action can be selected, for example, by choosing the ABD and its affinity for albumin. While not wishing to be bound by theory, it is believed that greater affinity for albumin will provide longer circulation times, which confers a longer duration of action. One or both of pharmacodynamics (therapeutic effects) and pharmacokinetics (drug properties) can be measured over time after delivery, such as plasma drug levels, acute or chronic glucose and/or HbA1c decrease, plasma insulin levels, inhibition of food intake or weight loss. C. Effective Dosages
[000270] Pharmaceutical compositions provided in the present invention include compositions wherein the active ingredient is contained in a therapeutically effective amount, i.e., in an amount effective to achieve its intended purpose. The actual effective amount for a particular application will depend, inter alia, on the condition being treated. For example, when administered in methods for treating diabetes, such compositions will contain an amount of active ingredient effective to achieve the desired result (e.g., lowering a subject's fasting blood glucose). When administered in methods of treating obesity, such compositions will contain an amount of active ingredient effective to achieve the desired result (e.g., reduction in body mass).
[000271] The dosage and frequency (single or multiple doses) of the administered compound may vary, depending on a variety of factors, including the route of administration, size, age, sex, health, body weight, body mass index and diet of the patient. recipient, the nature and extent of symptoms of the disease being treated (e.g., the disease responds to the compounds described herein), presence of other diseases or other health-related problems; type of concomitant treatment and complications from any disease or treatment regimen. Other therapeutic regimens or agents can be used in conjunction with the methods and compounds of the invention.
[000272] Therapeutically effective amounts for use in humans can be determined from animal models. For example, a dose for humans can be formulated to obtain a concentration found to be effective in animals. Dosage in humans can be adjusted by monitoring one or more physiological parameters including, but not limited to, blood sugar and body mass, and adjusting dosages up or down, as described above and known in the art.
[000273] Dosages can be varied depending on the requirements of the patient and the compound being employed. The dose administered to a patient, in the context of the present invention, should be sufficient to obtain a beneficial therapeutic response in the patient over time. Dose size will also be determined by the existence, nature and extent of any adverse side effects. In general, treatment is started with lower dosages, which are less than the optimal dose of the compound. After which, the dosage is increased in small increments until the optimum effect under the circumstances is reached. In one embodiment of the invention, the dosage range is from 0.001% to 10% w/v. In another embodiment, the dosage range is 0.1% to 5% w/v.
[000274] However, typical doses may contain from a low limit of about 0.1 mg to a high limit of about 200 mg of the pharmaceutical compound per day. Also considered are other dose ranges, such as from 1mg to 100mg of the compound per dose and from 3mg to 70mg per dose. Typically, the dose of manipulated polypeptides with long duration of action is administered, for example, daily or even once a week. The daily doses may be distributed in separate unit doses, given continuously over a 24 hour period or any portion of these 24 hours.
[000275] Dosage amounts and intervals can be individually adjusted to provide levels of the administered compound effective for the particular clinical indication being treated. This will provide a therapeutic regimen that is commensurate with the severity of the subject's disease state.
[000276] Using the teachings provided herein, an effective prophylactic or therapeutic treatment regimen can be devised which does not cause substantial toxicity and yet is entirely effective in treating the clinical symptoms demonstrated by the particular patient. Such planning should involve careful choice of the active compound by considering factors such as compound potency, relative bioavailability, patient body weight, the presence and severity of adverse side effects, the preferred mode of administration, and the toxicity profile of the selected agent.
[000277] The surprising dose-moderating property of the engineered polypeptides described here, together with their surprising plasma half-life and long duration of pharmacological action, makes for a superior pharmaceutical agent. Superior properties, including dose moderation, allow for lower dosage, thus, fewer or less severe side effects and improved cost of products and/or more cost effective and simpler formulations for once-daily or once-weekly administration not currently obtained by the parent compounds alone. D. Toxicity
[000278] The relationship between toxicity and therapeutic effect for a particular compound is its therapeutic index and can be expressed as the ratio between the LD50 (the amount of lethal compound in 50% of the population) and the ED50 (the amount of effective compound in 50% of the population). Compounds that exhibit high therapeutic indices are preferred. Therapeutic index data obtained from cell culture assays and/or animal studies can be used in formulating a range of dosages for use in humans. The dosage of such compounds preferably lies within a range of plasma concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range, depending on the dosage form employed and the route of administration used. See, for example, Fingi et al., in: THE PHARMACOLOGICAL BASIS OF THERAPEUTICS, Chapter 1, p. 1, 1975. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition and the method in which the particular compound is used.
[000279] Without wishing to be bound by any theory, it is believed that conjugation of an albumin-binding ABD domain with a hormonal domain as described herein may confer diminished immunogenicity, as assessed by a reduction in immune response relative to the hormonal domain without conjugation with ABD. See, for example, WO 2009/016043, incorporated herein by reference in full and for all intents and purposes. VII. Examples Example 1: Recovering manipulated polypeptide
[000280] Protein sequences were designed and retranslated using commercial DNA sequence software for cloning into an E. coli expression vector. The sequences were either obtained as oligonucleotides and ligated together using standard PCR amplification techniques or digested from existing expression constructs using standard restriction enzymes and then ligated back together. Sequences expressing the protein of interest were placed in pET45 with a T7 promoter for inducible expression. After the constructs were verified through sequencing, the vector DNA was purified and transformed into an expression host, typically BL21 (DE3). A single colony was selected for growth of a starter culture in 4 ml of LB medium for ~6 hours. Glycerol stocks were prepared by adding 100 μl of 80% glycerol to 900 μl of the stock and stored at -80 °C. Optionally, 500 μl of uninduced sample was retained for gel analysis. A 60 ml culture (Magic medium) was inoculated with 60 μl of starter culture in a 125 ml Thompson flask and incubated @ 30 °C overnight. Remove 250 μl of sample for analysis. Centrifuge and freeze the cell pellet for further processing.
[000281] Bacterial cells were collected and subsequently lysed to isolate inclusion bodies. Once the protein was present in the inclusion bodies, they were solubilized and the protein refolded at 4°C. The proteins were then separated using size exclusion chromatography until only a single band remained and endotoxin levels were acceptable for in vivo testing. Analytical HPLC, RP-LC-MS and SDS-PAGE gel were performed as quality control measures on the final protein. Protein was distributed in predetermined aliquots and stored at -80°C.
[000282] Typical manipulated polypeptide recoveries for the methods described here are given in Table 5 below. Surprisingly, the recoveries observed for the compounds and production methods described above can be significantly greater than the recoveries observed with previously reported conjugated species, for example, Fc-leptin and the like. Furthermore, the foreign ABD domain did not adversely affect the expression, recovery, refolding, yield, or solubility of recovered engineered polypeptides, particularly for leptin conjugates, despite generally recognized difficulties in leptin recovery and manipulation.Table 5. Recoveries of manipulated polypeptides
Example 2: Functional activity of leptin in vitro
[000283] Method. This assay measures receptor signaling after treatment of cells expressing a modified leptin receptor. Test samples were accepted at 100% purity and re-solved to assay concentration in 10X stimulation buffer. A total of 90 μl of 10X each compound was transferred to a deep well plate and serially diluted (3-fold series) with stimulation buffer using Perkin Elmer's Multiprobe® and the program "MSV_Lep_Func_3-Fold_Dil-Deepwell_96.MPT". The serially diluted plate was composited into the 96-well stimulation plate containing 2.5 x 10-5 cell pellets from 18 hour leptin-deprived Keptin cells, as known in the art, using MultiMek's "MSV_Lep_Func_200μl_Transfer" test program which transfer 200 μl of each of the diluted compounds and mix the cells. At this time, the plate was sealed with an adhesive plate lid and placed at 37°C for 30 minutes to allow for pSTAT5 stimulation. See, for example, Crouse et al., 1998, J. Biol. Chem., 273: 18365-18373. After incubation, the stimulation plate was centrifuged to re-pellet the cells, the supernatant was removed and the remaining cell pellets were frozen at -80°C (>30 minutes). Cell lysates were made by adding 100 µl of 1x lysate to thawed cell pellets (Perkin Elmer pSTAT5 Assay kit) with rotation at RT for 20 minutes. Lysates were clarified at 2500 rpm for 20 minutes and analyzed in the pSTAT5 Assay kit as 4 μl/well in 384-well ProxiplateTM according to the manufacturer's instructions. The pSTAT5 (RFU) signal was determined using a Packard Fusion α-FP HT Plate Reader set to Alpha readout parameters. The assay was completed on 384-well Proxiplate™ plates in a total volume of 11 μl, with values representing the mean of n = 4 assay wells per dose point.
[000284] Referring to Table 6 below, Cmpds C1-C6 are exemplary leptins, leptin analogues and leptin derivatives as described herein. Specifically, Cmpd C1 is SEQ ID NO: 20 as described herein. Cmpd C2 is SEQ ID NO: 30 (i.e. A200). Cmpd C3 is SEQ ID NO: 32 wherein a single 20 kDa polyethylene glycol (PEG) moiety has been linked through the cysteine residue at position 78. Methods for conjugating peptides and proteins to PEG are as known in the art. Cmpd C4 is a PEGylated derivative of SEQ ID NO: 20, wherein a single 20 kDa PEG has been linked via the N-terminus of SEQ ID NO: 20. Cmpd C5 is a PEGylated derivative of SEQ ID NO: 32 doubly PEGylated , wherein a single 20 kDa PEG moiety was linked via the cysteine residue at position 78 and a single 20 kDa PEG moiety was linked via the N-terminus. Cmpd C6 is a PEGylated derivative in which a single 40 kDa PEG has been linked via the N-terminus.
[000285] Results. As shown in Table 6 below, the engineered polypeptides described here (eg, Cmpds 1-4) have comparable and even superior functional activity in the STAT5 Obeca assay compared to a variety of leptin conjugates. Table 6: In vitro functional activity for leptins
Example 3: Change in body weight after a single administration
[000286] Method. Lean Sprague Dawley rats were maintained on a low-fat diet during the study. The average body weight was 319 grams at baseline. The animals were divided into six groups (n = 6/group). Each group was assigned to receive one of the following: vehicle; Cmpd 1 at 2.6 mg/kg in vehicle; Cmpd 2 at 2.7 mg/kg in vehicle; Cmpd 4 at 2.7 mg/kg in vehicle; Cmpd 2 at 10 mg/kg in vehicle. Each test animal received a single subcutaneous injection at time = 0. Food intake and body weight change (% corrected for vehicle) were monitored for 14 days and results recorded as shown (Figures 1A to 1B). Compounds administered: Vehicle (square); Cmpd 1 to 2.6 mg/kg (tip-up triangle); Cmpd 2 at 2.7 mg/kg (tip-down triangle); Cmpd 4 at 2.7 mg/kg (diamond); Cmpd C2 at 10 mg/kg (circle).
[000287] Results. As depicted in Figures 1A to 1B, administration of each manipulated polypeptide resulted in reduced food intake and body weight. All compounds were provided at an equimolar dose to total compound weight; compounds were all fed at 120 nmol/kg (i.e., Cmpd 1 at 2.6 mg/kg; Cmpd 2 and Cmpd 4 at 2.7 mg/kg; Cmpd C2 at 10 mg/kg). It should be noted that Cmpd C2 (e.g. A200) is a dimer of two portions, each portion consisting of the FC region of IgG1 fused to human leptin. Cmpd 1 and Cmpd 2 have similar activity to Cmpd C2 which, by virtue of being a dimer, has two leptins per molecule. Although the efficacy (lower body weight) appears similar, it is evident that the trend favors both manipulated polypeptides over Cmpd C2. When viewed on a per mole of leptin basis, engineered polypeptides are superior for inhibiting food intake and body weight, as Cmpd C2 has 2 moles of leptin for each dimeric Fcleptin complex, while each mole of ABD-leptin portion has only 1 mole of leptin.
[000288] Previous results have shown that approximately 500 µg/kg/day of an A500 compound is required to achieve a 9-10% weight loss at 7 days when given by continuous infusion to a lean rat. This results in 2.5 mg of leptin compound A500 for 5 days and 3.5 mg of compound for 7 days. Since an A500 compound itself is 16067.68 g/mol and the molecular mass of Cmpd 2 is ~22.510 g/mol, one would predict the need for 1.4X more ABD fusion protein during the 5 days. Previously, only 1.08X more compound (2.7mg/2.5mg) was delivered, which indicates a surprising dose-moderating property. Example 4: Change in body weight after a single administration of Cmpd 2.
[000289] Method. Lean Sprague Dawley rats were maintained on a low-fat diet during the study. Mean body weight was 324 grams at baseline. The animals were divided into four groups (n = 6/group). Each group was assigned to receive one of the following: vehicle; Cmpd 2 at 0.3 mg/kg in vehicle; Cmpd 2 at 1.0 mg/kg in vehicle; Cmpd 2 at 3.0 mg/kg in vehicle. Each test animal received a single subcutaneous injection at time = 0. Food intake and body weight change (% corrected for vehicle) were monitored for 14 days and results recorded as shown (Figures 2a and 2b). Compounds administered: Vehicle (square); Cmpd 2 at 0.3 mg/kg (tip-up triangle); Cmpd 2 at 1.0 mg/kg (tip-down triangle); Cmpd 2 at 3.0 mg/kg (diamond).
[000290] Results. As illustrated in Figures 2A to 2B, administration of engineered Cmpd 2 polypeptide at each of the concentrations resulted in reduced food intake and body weight. The dose response is seen in Figure 2B. Example 5: Change in body weight after a single administration of Cmpd C2
[000291] Method. Lean Sprague Dawley rats were maintained on a low-fat diet during the study. Mean body weight was 324 grams at baseline. The animals were divided into four groups (n = 6/group). Each group was assigned to receive one of the following: vehicle; Cmpd C2 at 1.1 mg/kg in vehicle; Cmpd C2 at 3.3 mg/kg in vehicle; Cmpd C2 at 11.1 mg/kg in vehicle. Each test animal received a single subcutaneous injection at time = 0. Food intake and change in body weight (% corrected for vehicle) were monitored for 14 days and results recorded as illustrated (Figures 3A to 3B). Compounds administered: Vehicle (square); Cmpd C2 at 1.1 mg/kg (circle); Cmpd C2 at 3.3 mg/kg (square); Cmpd C2 at 11.1 mg/kg (tip-up triangle).
[000292] Results. As illustrated in Figures 3A through 3B, administration of control Cmpd C2 at each concentration resulted in reduced food intake and body weight. Example 6: Change in body weight after a single administration of Cmpd C6
[000293] Method. Lean Sprague Dawley rats were maintained on a low-fat diet during the study. Mean body weight was 324 grams at baseline. The animals were divided into two groups (n = 6/group). Each group was assigned to receive one of the following: vehicle; Cmpd C6 at 2.2 mg/kg in vehicle. Each test animal received a single subcutaneous injection at time = 0. Food intake and body weight change (% corrected for vehicle) were controlled and results recorded as shown (Figures 4A to 4B)). Compounds administered: Vehicle (square); Cmpd C6 at 2.2 mg/kg (tip-down triangle).
[000294] Results. As depicted in Figures 4A to 4B, administration of control Cmpd C6 at each concentration resulted in reduced food intake and body weight. Example 7: Change in body weight in DIO mice
[000295] Method. Diet-induced obesity (DIO) Sprague Dawley rats weighing on average about 500 grams were injected IP with the test and control compounds on day 0 and day 7 (n = 6 per compound). Test compound was SEQ ID NO: 54 supplied at 1.3 mg/kg/week in vehicle. Body weight and food intake were measured at various points (days 0, 4, 7, 12 and 14 d) during the study period. Compounds administered: Vehicle (square); SEQ ID NO: 54 at 1.3 mg/kg in vehicle (tip-up triangle).
[000296] Results. The results depicted in Figure 5 demonstrate that IP injection at an interval of once a week results in a 3% weight loss after 7 days, as previously observed at this dose. Upon a second injection, mice continued to lose weight, resulting in a vehicle-corrected 7-8% cumulative weight loss at 14 days. In contrast and surprisingly, previous studies with FC-leptin (A200 leptin) resulted in a weight loss of only approximately 4% at 14 days after a dose of 5 mg/kg/week with injections on day 0 and day 7, in a similar DIO model. Example 8: Detection of manipulated polypeptides in plasma
[000297] Method. Diet-Induced Obesity (DIO) Sprague Dawley rats weighing an average of approximately 483 g were divided into five groups, two of which were implanted with osmotic pumps. One of the two osmotic pump groups received a continuous subcutaneous (SC) infusion of vehicle only, the other received a CSI of SEQ ID NO: 33 (i.e., A500) in vehicle at a dose of 250 μg/kg/day . The other three groups were treated as follows: one group received 15 subcutaneous injections of vehicle only once a week on study days 0, 7, 14, and 21; another group received once weekly subcutaneous injections of SEQ ID NO: 54 (an ABD-A500 engineered polypeptide) at a dose of 1.3 mg/kg in vehicle on days 0, 7, 14, and 21 of the study; the remaining group received once weekly subcutaneous injections of SEQ ID NO: 54 at a dose of 3.0 mg/kg in vehicle on days 0, 7, 14, and 21 of the study. Blood samples were taken from each animal on day 27, which was the day of study termination.
[000298] Results. The results depicted in Figure 6 demonstrate that once weekly injection of SEQ ID NO: 54 at 1.3 mg/kg resulted in plasma levels that were slightly lower than those obtained by continuous infusion of SEQ ID NO: 33 and once weekly injection of SEQ ID NO: 54 at 3.0 mg/kg resulted in plasma levels that were significantly higher than those obtained with continuous infusion of SEQ ID NO: 33 (compare left panel with right panel; note the difference on the Y-axis scales of each panel). Example 9: Change in body weight after a single administration of engineered polypeptides
[000299] Method. Lean Sprague Dawley rats were maintained on a low-fat diet during the study. The average body weight was 330 grams at the start of the study. Each test animal (n = 5/group) received a single subcutaneous injection at time = 0. The animals were divided into five groups. Each group was assigned to receive one of the following: vehicle; SEQ ID NO: 54 in vehicle; SEQ ID NO: 56 in vehicle; SEQ ID NO: 58 in vehicle; SEQ ID NO: 59 in vehicle. SEQ ID NOs: 54, 56, 58 and 59 were each administered at a dose of 120 nmol/kg. The percentage change in body weight for each group was monitored over 14 days and the results recorded as shown (Figure 7).
[000300] Results. As illustrated in Figure 7, each group of animals that received a single injection of one of the SEQ ID NOs tested showed significant and constant weight loss over the 14 days of the study relative to the group that received vehicle only. Example 10: Change in body weight after a single administration of engineered polypeptides
[000301] Method. Lean Sprague Dawley rats were maintained on a low-fat diet during the study. The average body weight was 330 grams at the start of the study. The animals were divided into six groups (n = 5/group). Each test animal received a single subcutaneous injection at time = 0. Each group was assigned to receive one of the following: vehicle; SEQ ID NO: 54 in vehicle; SEQ ID NO: 57 in vehicle; SEQ ID NO: 60 in vehicle; SEQ ID NO: 61 in vehicle; SEQ ID NO: 62 in vehicle. SEQ ID NOs: 54, 57, 60, 61 and 62 were each administered at a dose of 120 nmol/kg. Percent change in body weight for each group was monitored over 14 days and the results recorded as shown (Figure 8).
[000302] Results. As illustrated in Figure 8, each group of animals that received a single injection of one of the SEQ ID NOs tested showed a significant and sustained reduction in body weight relative to the group that received vehicle alone. Example 11: Change in body weight and food intake after a single administration of engineered polypeptides
[000303] Method. Lean Sprague Dawley rats were maintained on a low-fat diet during the study. The average body weight was 317 grams at the start of the study. Each test animal (n = 7/group) received a single subcutaneous injection at time = 0. The animals were divided into four groups. Each group was assigned to receive one of the following: vehicle; SEQ ID NO: 54 in vehicle; SEQ ID NO: 63 in vehicle; SEQ ID NO: 64 in vehicle. SEQ ID NOS: 54, 63, and 64 were each administered at a dose of 120 nmol/kg. The percent change in food intake and body weight for each group was monitored over 14 days and the results recorded as shown (Figures 9A and 9B, respectively).
[000304] Results. As depicted in Figures 9A and 9B, each group of animals that received a single injection of one of the SEQ ID NOs tested showed a significant and sustained reduction in food intake (Figure 9A) and body weight (Figure 9B) relative to vehicle alone. . Example 12: Change in body weight after a single administration of engineered polypeptides
[000305] Method. Lean Sprague Dawley rats were maintained on a low-fat diet during the study. Mean body weight was 330 g at baseline. Animals were divided into six groups of 10. Each test animal (n = 5/group) received a single subcutaneous injection at time = 0. Each group was assigned to receive one of the following: vehicle; SEQ ID NO: 54 in vehicle; SEQ ID NO: 67 in vehicle; SEQ ID NO: 68 in vehicle; and SEQ ID NO: 69 in vehicle. SEQ ID NOs: 54, 67, 68 and 69 were each administered at a dose of 120 nmol/kg. The percentage change in body weight for each group was monitored over 10 days and the results recorded as shown (Figure 10).
[000306] Results. As depicted in Figure 10, each group of animals that received a single injection of one of the SEQ ID NOs tested exhibited a significant and sustained reduction in body weight relative to the group that received vehicle alone. Example 13: Change in body weight after a single administration of engineered polypeptides
[000307] Method. Lean Sprague Dawley rats were maintained on a low-fat diet during the study. The average body weight was 320 grams at the start of the study. The animals were divided into six groups. Each test animal (n = 5/group) received a single subcutaneous injection at time = 0. Each group was assigned to receive one of the following: vehicle; SEQ ID NO: 54 in vehicle; SEQ ID NO: 104 in vehicle; SEQ ID NO: 105 in vehicle; SEQ ID NO: 106 in vehicle; and SEQ ID NO: 107 in vehicle. SEQ ID NOs: 54, 104, 105, 106 and 107 were each administered at a dose of 120 nmol/kg. Change in body weight for each group was monitored for 9 days and results recorded as shown (Figures 11A to 11C).
[000308] Results. As depicted in Figure 11, each group of animals that received a single injection of one of the SEQ ID NOS tested exhibited a significant and sustained reduction in food intake (Figure 11A) and body weight (Figures 11B and 11C) relative to vehicle alone. . Example 14: Determination of affinity for albumin-binding polypeptides
[000309] In this example, Compound 2 and Compound 15 were characterized by affinity in different albumin variants. Material and methods
[000310] All studies were performed on a BioRad ProteOn XPR36 system using a GLC sensor chip at 25 degrees C. For amine coupling, the GLC chip was activated for 5 minutes using a 1:1 mixture of sulfo-NHS/EDC diluted 30 times from the initial stock in water as shown below. Each albumin sample was diluted to 25 μg/ml in 10 mM Na Acetate, pH 5.0 and injected for 5 minutes on separate sensor surfaces. Each surface was then blocked with 1M ethanolamine, pH 8.5. Each albumin was coupled at a density of 20005000 in resonance units.
[000311] Binding of an engineered polypeptide was tested using 5 nM as the highest concentration in a three-fold dilution series. The running buffer contained 10 mM HEPES, pH 7.4, 150 mM NaCl, 3 mM EDTA and 0.005% Tween 20. All samples were tested using a 3-fold dilution series. Each concentration series was tested in duplicate. The dissociation phase for the highest concentration was monitored for 3 hours. Results
[000312] The relative KD measured for the manipulated polypeptides is shown in Table 7 below. The results demonstrate that albumin-binding polypeptides associate with serum albumins (Serum Albumin - SA) with high affinity.Table 7: KD of albumin-binding polypeptides to albumin variants
Example 15: Functional activity of leptin in vitro in the presence of albumin.
[000313] In this assay, the method described in Example 2 was used, except that albumin was added to the stimulation buffer to test the leptin function of Compound 2 in the presence of albumin. Albumins tested included 0.1% or 1% bovine serum albumin (Bovine Serum Albumin - BSA), 1% rat serum albumin (Rat Serum Albumin - RSA) or human serum albumin (Human Serum Albumin - HSA ) to 1%. The control sample was leptin A100 with 0.1% BSA.
[000314] Results. As shown in Figure 12, there was no effect of 1% bovine/mouse/human Albumin on EC50 activity generated by Compound 2 in the Leptin Function Assay. The results are surprising and show that the therapeutic compounds are active even when bound to albumin. Example 16: Prolonged pharmacokinetic profiles conferred by manipulated polypeptides after subcutaneous injections in rats
[000315] This study was performed to evaluate Compound 2 and Compound 15 in rats by comparing their blood concentration versus time profiles, ie pharmacokinetic profiles.
[000316] Mice were placed in treatment groups. Compound 2 was administered subcutaneously at 30 nmol/kg, 60 nmol/kg or 120 nmol/kg. Blood samples were taken at pre, 12, 24, 48, 96 and 144 hours post-administration in the lateral caudal vein. The concentration of Compound 2 in plasma was measured by an enzyme-linked immunosorbent assay method.
[000317] Compound 15 was administered subcutaneously at 120 nmol/kg. Blood samples were taken at pre, 0.5, 1, 2, 4, 6, 24, 48, 72, 96, 120 and 144 hours post-administration in the lateral caudal vein. The concentration of Compound 15 in plasma was measured using an enzyme-linked immunosorbent assay method.
[000318] Compound 2 (Figure 13) and Compound 15 (Figure 14) exhibited prolonged plasma versus time profiles. Example 17: Effect of engineered polypeptides mediated by leptin receptors
[000319] Method. Lean Sprague Dawley rats and ZDF rats were used for this study. ZDF mice have a mutation (fa) which results in a shortened leptin receptor which does not interact effectively with leptin. The average body weight was 225 grams at baseline. The animals were divided into two groups (n = 5/group). Each group was assigned to receive one of the following: vehicle; Cmpd 2 at 2.7 mg/kg in vehicle. Each test animal received a single subcutaneous injection at time = 0. Body weight change (% corrected for vehicle) was monitored and results recorded as illustrated (Figures 15A, 15B)). Administered Compounds: Vehicle (solid circle); Cmpd 2 at 2.7 mg/kg (solid square).
[000320] Results. As depicted in Figures 15A and 15B, Compound 2 is not effective in ZDF mice, indicating that its effects are mediated via leptin receptors. Example 18: Dose moderation with engineered polypeptides
[000321] This study compared doses of A500 (SEQ ID NO: 33) and Compound 2 (SEQ ID NO: 54) needed to achieve a similar amount of weight loss in lean leptin sensitive rats. The results are shown in Figure 16. Compound 2 dosed at 120 nmol/kg/week achieves an 18% weight loss corrected for vehicle. To achieve the same amount of weight loss with A500, a BID dose of 120 nmol/kg/d or 1680 nmol/kg per injection (120 per injection x 2 for BID x 7 days) over a week is required. Without intending to be bound by any theory, this "dose moderation" may be at least partially attributable to Compound 2's improved PK profile relative to the A500. Example 19: Solubility of manipulated polypeptides
[000322] As shown in Table 8 below, the engineered polypeptides described herein have surprisingly high solubility at neutral pH.
[000323] Solubility was measured with the following assay: 6-10 mg of purified proteins were concentrated at 4 °C with centrifuge filter units (Amicon Ultra-15 or Ultra-4, MW cutoff of 3 kDa; Millipore ) for a volume of less than 0.5 ml. They were centrifuged at 14,000 rpm for 10 minutes at 4°C to remove precipitates and the supernatant transferred to a new tube. Proteins were allowed to equilibrate overnight at room temperature in the dark, then filtered with 0.22 micron parasyringe filters (Milex GV; Millipore) to remove precipitates. The absorbance at OD280 was measured with a NanoDrop spectrophotometer and the concentration was calculated using the theoretical molar extinction coefficient of the protein.Table 8. Solubility of manipulated polypeptides
Example 20: Stability of manipulated polypeptides
[000324] As shown in Table 9 below, the engineered polypeptides described herein are chemically stable. Compounds were formulated at 1 mg/mL in buffer solutions of different pH's. As shown in Table 9, chimeric polypeptides have good potency (Table 9A) and purity (Table 9B) after two weeks at 40°C as determined by reversed-phase High Performance Liquid Chromatography (HPLC). .Table 9A. Potency of manipulated polypeptides
aPower = main peak area of the standard areaTable 9B. Purity of manipulated polypeptides
Example 21: Stability of manipulated polypeptides
[000325] As shown in Table 10 below, the engineered polypeptides described herein are chemically stable. Compound 15 was formulated at three different concentrations in the buffer in the following buffer: 10 mM glutamic acid, 2% glycine, 1% sucrose, 0.01% Tween 20, pH 4.25 and stored at 5°C , 15°C or 25°C. As shown in Table 10, Compound 15 is chemically stable at 10, 20 and 30 mg/mL for at least 1 month at 5-25°C as determined by HPLC.Table 10. Stability of engineered polypeptides
Example 22: Stability of manipulated polypeptides
[000326] As shown in Table 11 below, engineered polypeptides described herein are physically stable. Compound 15 was formulated at three different concentrations in the following buffer: 10 mM glutamic acid, 2% glycine, 1% sucrose, 0.01% Tween 20, pH 4.25 and stored at 37°C. As shown in Table 11, Compound 15 is physically stable at 10, 20 and 30 mg/mL for at least 1 month as determined by visual analysis. Table 11. Stability of engineered polypeptides
Example 23: Stability of manipulated polypeptides
[000327] The engineered polypeptides described here are physically stable. Table 12 shows the results of Size Exclusion Chromatography (SEC) performed on A100, ABD1-HuFoca and ABD1-A500. Engineered polypeptides show little or no dimer/oligomer self-association compared to A100.Table 12. Stability of engineered polypeptides
Pk 1 = Monomer Pk 2 = Dimer Pk 3 = Oligomer (Trimer/Tetramer) SEC Method: Column - Tosoh TSK Gel G3000 SW x 1.7.8 mm x 30 cm (# 08541) Mobile phase - 10 mM Na phosphate , pH 7.4 + 238 mM NaCl + 2.7 mM KCl Operating Time - 22 min Flow Rate - 0.8 mL/min Column Temp - 25 °C Sample Temp - 5 °C Sample Load - 40 μg Detection - 214 nm Example 24: Synergy of amylin and leptin is absent in individuals with high BMI
[000328] Previous studies had described synergy between amylin/leptin in rats weighing 500-550 grams. After an inverse relationship between efficacy and BMI was noted, we evaluated the effects of the combination in very obese rats (750 grams) and in very obese rats that had food restricted to the moderately obese range (500-550 g) before starting. drug treatment.
[000329] In this study, a group of very obese rats (750 g) were allowed to eat ad libitum and were treated with amylin, leptin or the combination of amylin + leptin. Although amylin was effective, there was no evident synergy with the addition of leptin. A second group of very obese rats (750 g) had calories restricted to the 500-550 g range, in which synergy was previously demonstrated. These animals were then started on amylin/leptin treatment and were allowed to feed ad libitum. Figure 17 shows the results of the study. Rapid weight regain was evident in rats treated by vehicle and leptin monotherapy. Some weight maintenance was achieved with amylin monotherapy. No additional weight maintenance was achieved with the combination. These results suggest that the lack of synergy in "high" BMI rodents cannot simply be compensated for by a low-calorie diet. Example 25: Synergy of engineered polypeptides with amylin agonists
[000330] This study examined whether a once-weekly administration of rat PEG-amylin (Rat Des-Lys1-[Lys26 (mPEG40K)]-Amylin, Compound 124) would be sufficient for synergy when co-administered with ABD1-A500 (Compound 2). For comparison, ABD1-A500 was also co-administered with infused rat amylin (Fig. 18A). Figure 18B shows that although rat PEG-amylin induced weight loss, it is somewhat slower and of lesser magnitude, the overall amount of weight loss (and synergy) is qualitatively similar to that obtained by rat amylin infused. Amylin was administered at 50 μ/kg/d via a SC mini osmotic pump, rat PEG-amylin was administered at 125 nmol/kg once a week and ABD1-A500 was administered at 120 nmol/kg once a week to male Harlan Sprague Dawley rats with diet-induced obesity (Diet Induced Obese - DIO) of 500 g of average weight. Example 26: Synergy of engineered polypeptides with amylin agonists
[000331] This study shows that once weekly administration of ABD1-HuFoca (Compound 15) is sufficient for synergy when co-administered with infused rat amylin. Figure 19 shows that the combination of the engineered polypeptide and infused amylin resulted in less food consumption (Figure 19A) and greater weight loss (Figure 19B) than the results observed for either agent alone. ABD1-HuFoca was administered at 120 nmol/kg and amylin was administered at 50 µg/kg/d via a SC mini osmotic pump to 500 g average weight male DIO HSD rats. Example 27: Synergy of engineered polypeptides with amylin agonists
[000332] This study shows that once-weekly administration of ABD1-HuFoca (Compound 15) is sufficient for synergy when co-administered with twice-weekly administration of rat PEG-Amylin (Des-Lys1-[Lys26 ( mPEG40K)]-rat amylin, Compound 124). Figure 20 shows that the combination of the engineered polypeptide and rat PEG-amylin resulted in greater weight loss than the results observed for either agent alone. ABD1-HuFoca was administered at 120 nmol/kg and PEG-amylin was administered at 125 nmol/kg to 500 g average weight male DIO HSD rats. Example 28: Synergy of manipulated polypeptides with amylin agonists in a high BMI population
[000333] Figure 21A shows the results of previous studies describing amylin/leptin synergy in rats weighing 500,550 grams. Figure 21B shows that this synergy is not observed in a population of high BMI rats (mean weight 700 g). Figure 21C shows that once weekly administration of ABD1-A500 (Compound 15) is sufficient for synergy when co-administered with twice weekly administration of rat PEG-amylin (Des-Lys1-[Lys26 (mPEG40K) Rat ]-amylin, Compound 124) in high BMI rats. ABD1-A500 was administered at 120 nmol/kg and PEG-amylin was administered at 5 to 125 nmol/kg to 700 g average weight male DIO HSD rats. Example 29: Synergy of engineered polypeptides with amylin agonists in a high BMI population
[000334] This study shows that once weekly administration of ABD1-HuFoca (Compound 15) or ABD1-A500 (Compound 2) is sufficient for synergy when co-administered with rat amylin infused to high BMI rats. ABD1-HuFoca (Figure 22A) or ABD1-A500 (Figure 22B) was administered at 120 nmol/kg and amylin was administered at 50 µg/kg/d via a SC mini osmotic pump to male DIO HSD rats of 700 g of average weight. Example 30: Anti-diabetic effects of engineered polypeptides on non-obese rats with type 1 diabetes
[000335] The aim of this study was to evaluate the in vivo effects of engineered polypeptides on key metabolic and diabetic endpoints in a mouse model of type 1 diabetes mellitus (T1DM) by high-dose STZ. Male C57 BL/6 mice received a single injection of interperitoneal STZ at 200 mg/kg to induce type 1 diabetes. Compounds were administered twice weekly subcutaneously at 120 nmol/kg for two weeks. Measured endpoints included HbA1c levels, glucose levels, body weight and food intake.
[000336] Figure 23 shows that both Compound 15 and Compound 2 normalized blood glucose in rats with STZ-induced diabetes. Both manipulated polypeptides also reduced hemoglobin A1c levels, as shown in Figure 24, and reduced body weight and cumulative food intake, as shown in Figure 25.
[000337] In order to ensure that the glucose lowering effects of the therapy are not due to the effects of insulin, another study was conducted to combine leptin therapy with a low dose of insulin. Compound 15 was administered with or without the addition of a dose of 0.05 U/day of insulin in a mouse model of T1DM by high dose of STZ. Male C57 BL/6 mice received a single injection of interperitoneal STZ at 175 mg/kg to induce type 1 diabetes. Compounds were administered subcutaneously twice weekly at 60 nmol/kg for two weeks. Measured endpoints included HbA1c levels, glucose levels, body weight and food intake.
[000338] Figure 26 shows a glucose lowering effect potentiated with a low dose of insulin in the form of an additive to Compound 15. It also reduces hemoglobin A1c levels as shown in Figure 27 and reduces body weight and intake cumulative amount of food, as shown in Figure 28. VIII. Modalities
[000339] Additional modalities of the manipulated polypeptides, the method of using them and the pharmaceutical compositions described here follow:
[000340] Embodiment 1. An engineered polypeptide comprising: an albumin-binding domain (ABD) peptide and a first hormonal peptide domain (HD1) selected from a leptin, a leptin analogue or an active fragment thereof.
[000341] Embodiment 2. The polypeptide engineered according to Embodiment 1, further comprising a first linker (L1) covalently linked to said HD1.
[000342] Embodiment 3. The engineered polypeptide according to Embodiment 1 or 2, wherein said engineered polypeptide comprises said ABD as an N-terminal portion and said HD1 as a C-terminal portion.
[000343] Embodiment 4. The engineered polypeptide according to Embodiment 1 or 2, wherein said engineered polypeptide comprises said ABD as a C-terminal portion and said HD1 as an N-terminal portion.
[000344] Embodiment 5. The polypeptide manipulated according to Embodiment 3, comprising the structure: ABD-HD1.
[000345] Mode 6. The polypeptide according to engineering Mode 3, comprising the structure: ABD-L1-HD1.
[000346] Embodiment 7. The polypeptide manipulated according to Embodiment 4, 25 comprising the structure: HD1-ABD.
[000347] Embodiment 8. The polypeptide manipulated according to Embodiment 4, comprising the structure: HD1-L1-ABD.
[000348] Embodiment 9. The polypeptide manipulated according to any one of Embodiments 1 to 8, wherein said HD1 is a leptin, a leptin analogue, an active leptin fragment or a leptin derivative.
[000349] Embodiment 10. The polypeptide engineered according to any one of Embodiments 1 to 9, wherein said HD1 has at least 50% identity to an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146.
[000350] Embodiment 11. The polypeptide engineered according to any one of Embodiments 1 to 10, wherein said HD1 has at least 90% identity to an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146.
[000351] Embodiment 12. The polypeptide manipulated according to any one of Embodiments 1 to 11, wherein said HD1 has at least 50% identity to a human leptin.
[000352] Embodiment 13. The polypeptide manipulated according to any one of Embodiments 1 to 12, wherein said HD1 has at least 90% identity to a human leptin.
[000353] Embodiment 14. The polypeptide engineered according to any one of Embodiments 1 to 13, wherein said HD1 is at least 50% with SEQ ID NO: 20.
[000354] Embodiment 15. The polypeptide engineered according to any one of Embodiments 1 to 14, wherein said HD1 is at least 90% with SEQ ID NO: 20.
[000355] Embodiment 16. The polypeptide manipulated according to any one of Embodiments 1 to 15, wherein said HD1 is at least 50% platypus leptin.
[000356] Embodiment 17. The polypeptide manipulated according to any one of Embodiments 1 to 16, wherein said HD1 is at least 50% with a seal leptin.
[000357] Embodiment 18. The polypeptide engineered according to any one of Embodiments 1 to 17, wherein said HD1 has from 1 to 5 amino acid modifications selected independently of any one or a combination of an insertion, deletion, addition or substitution .
[000358] Embodiment 19. The polypeptide engineered according to any one of Embodiments 1 to 18, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO : 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 , SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO : 145 and SEQ ID NO: 146.
[000359] Embodiment 20. The polypeptide engineered according to any one of Embodiments 1 to 19, wherein said HD1 comprises an amino acid sequence which is selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27 , SEQ ID NO: 28, SEQ ID NO: SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO : 144, SEQ ID NO: 145 and SEQ ID NO: 146.
[000360] Embodiment 21. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 1.
[000361] Embodiment 22. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 2.
[000362] Embodiment 23. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 3.
[000363] Embodiment 24. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 4.
[000364] Embodiment 25. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 5.
[000365] Embodiment 26. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 6.
[000366] Embodiment 27. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 7.
[000367] Embodiment 28. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 8.
[000368] Embodiment 29. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 9.
[000369] Embodiment 30. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 10.
[000370] Embodiment 31. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 11.
[000371] Embodiment 32. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 12.
[000372] Embodiment 33. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 13.
[000373] Embodiment 34. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 14.
[000374] Embodiment 35. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 15.
[000375] Embodiment 36. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 16.
[000376] Embodiment 37. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 17.
[000377] Embodiment 38. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 18.
[000378] Embodiment 39. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 19.
[000379] Embodiment 40. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 20.
[000380] Embodiment 41. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 21.
[000381] Embodiment 42. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 22.
[000382] Embodiment 43. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 23.
[000383] Embodiment 44. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 24.
[000384] Embodiment 45. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 25.
[000385] Embodiment 46. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 26.
[000386] Embodiment 47. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 27.
[000387] Embodiment 48. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 28.
[000388] Embodiment 49. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 29.
[000389] Embodiment 50. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 30.
[000390] Embodiment 51. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 31.
[000391] Embodiment 52. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 32.
[000392] Embodiment 53. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 33.
[000393] Embodiment 54. The polypeptide engineered according to any one of Embodiments 1 to 53, wherein said ABD comprises an albumin binding motif (ABM) consisting of the amino acid sequence: GVSD X5 YK X8 X9 I X11 X12 A X14 TVEGV X20 X23 X24 X25 I AL (SEQ ID NO: 34) wherein, independently of one another, X5 is selected from Y and F; X8 is selected from N, P and S; X9 is selected from V, I, L, M, F and Y; X11 is selected from S and D; X12 is selected from R, K and N; X14 is selected from K and R; X20 is selected from D, N, Q, E, H, S, R and K; X23 is selected from K, I and T; X24 is selected from A, S, T, G, H, L and D, and X25 is selected from H and D.
[000394] Embodiment 55. The polypeptide manipulated according to any one of Embodiments 1 to 54 wherein, independently of one another: X5 is Y; X8 is N; X23 is T or I; X24 is S or L; and X25 is E or H.
[000395] Embodiment 56. The polypeptide engineered according to any one of Embodiments 1 to 55, wherein the albumin binding motif comprises an amino acid sequence which is selected from the group consisting of: GVSDYYKNLINKAKTVEGVEALTLHI (SEQ ID NO: 114) and GVSDYYKNLINKAKTVEGVEALISEI (SEQ ID NO: 115).
[000396] Embodiment 57. The polypeptide engineered according to any one of Embodiments 1 to 56, wherein said ABD comprises an albumin-binding motif (ABM) that is not GVSDYYKNLINNAKTVEGVKALIDEI (SEQ ID NO: 35).
[000397] Embodiment 58. The polypeptide manipulated according to any one of Embodiments 1 to 57, wherein said ABD comprises an ABM described in Table 1.
[000398] Embodiment 59. The polypeptide engineered according to any one of Embodiments 1 to 58, wherein said ABD comprises the amino acid sequence: LAEAK Xa Xb A Xc Xd EL Xe KY -[ABM]-LAALP (SEQ ID NO : 36) wherein: [ABM] is an albumin-binding motif, and, independently of each other, Xa is selected from V and E; Xb is selected from L and D; Xc is selected from N, L and I; Xd is selected from R and K; Xe is selected from D and K; leucine at position 45 is present or absent; and the proline at position 46 is present or absent.
[000399] Embodiment 60. The polypeptide manipulated according to any one of Embodiments 1 to 59 wherein, independently of one another: Xa is E; Xb is D; Xc is I; and Xd is K.
[000400] Embodiment 61. The polypeptide manipulated according to any one of Embodiments 1 to 60, wherein the albumin-binding domain (ABD) polypeptide comprises an amino acid sequence that is selected from the group consisting of: LAEABCEDAIKELDKYGVSDYYKNLINKAKTVEGVEALTLH ILAALP ( SEQ ID NO: 50) and LAEAKEDAIKELDKYGVSDYYKNLFNKAKTVEGVEALISEIL AALP (SEQ ID NO: 51).
[000401] Embodiment 62. The polypeptide engineered according to any one of Embodiments 1 to 61, wherein said ABD comprises the amino acid sequence: LAEAK Xa Xb A Xc Xd EL Xe KY-[ABM]-LAALP (SEQ ID NO : 36) wherein: [ABM] is an albumin-binding motif, and, independently of each other, Xa is selected from V and E; Xb is selected from L and D; Xc is selected from N, L and I; Xd is selected from R and K; Xe is selected from D and K; and leucine at position 45 is present or absent; proline at position 46 is present or absent; and wherein ABM is the amino acid sequence: GVSD X5 YK X8 X9 X11 X12 A X14 TVEGV X20 AL X23 X24 X25 (SEQ ID NO: 34) wherein, independently of one another, X5 is selected from Y and F; X8 is selected from N, P and S; X9 is selected from V, I, L, M, F and Y; X11 is selected from, S and D; X12 is selected from R, K and N; X14 is selected from K and R; X20 is selected from D, N, Q, E, H, S, R and K; X23 is selected from K, I and T; X24 is selected from A, S, T, G, H, L and D; and X25 is selected from H and D.
[000402] Embodiment 63. The polypeptide engineered according to any one of Embodiments 1 to 62, wherein said ABD comprises an amino acid sequence having at least 85% identity to an amino acid sequence that is selected from the group consisting of in SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, 10 SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51 and SEQ ID NO: 52.
[000403] Embodiment 64. The polypeptide manipulated according to any one of Embodiments 1 to 63, wherein said ABD comprises any of the peptides selected from the group consisting of: LAEAKVLANRELDKYGVSDFYKSYINRAKTVEGVHTLIGHILAALP (SEQ ID NO: 38), LAEAKVLANRELDKYGVSDFYKRLINKAKTVEGVNALTHHILAALP (SEQ ID NO: 38), LAEAKVLANRELDKYGVSDFYKRLINKAKTVEGVNALTHHILAALP (SEQ ID NO: 38), NO: 39), LAEAKVLANRELDKYGVSDYYKNLINRARTVEGVHALIDHILAALP (SEQ ID NO: 40), LAEAKVLANRELDKYGVSDYYKNIINRAKTVEGVRALKLHILAALP (SEQ ID NO: 41), LAEAKVLANRELDKYGVSDFYKNLINRAKTVEGVSSLKGHILAALP (SEQ ID NO: 42), LAEAKVLANRELDKYGVSDYYKNLINKAKTVEGVEALTLHILAALP (SEQ ID NO: 43), LAEAKVLANRELDKYGVSDFYKNLINRAKTVEGVDALIAHILAALP (SEQ ID NO: 44), LAEAKVLANRELDKYGVSDFYKSLINRAKTVEGVDALTSHILAALP (SEQ ID NO: 45), LAEAKVLANRELDKYGVSDFYKNLINRAKTVEGVNSLTSHILAALP (SEQ ID NO: 46), LAEAKVLANRELDKYGVSDFYKNVINKAKTVEGVEALIADILAALP (SEQ ID NO: 47), LAEAKVLANRELDKYGVSDYYKNLINKAKTVEGVQALIAHILAALP (SEQ ID NO: 48), LAEAKVLANRELDKYGVSDFYKRLINKAKTVEGVEALKLHILAALP (SEQ ID NO: 49), LAEAKEDAIKELDKYGVSDYYKNLINKAKTVEG VEALTLHILAALP (SEQ ID NO: 50), LAEAKEDAIKELDKYGVSDYYKNLINKAKTVEGVEALISEILAALP (SEQ ID NO: 51) and LAEAKEDAIKELDKYGVSDYYKRLISKAKTVEGVKALISEILAALP (SEQ ID NO: 52).
[000404] Embodiment 65. The polypeptide engineered according to any one of Embodiments 1 to 64, wherein said L1 linker is a peptide of 1 to 30 amino acids or less than 30 amino acids.
[000405] Embodiment 66. The polypeptide engineered according to any one of Embodiments 51 to 65, wherein said L1 linker is selected from 20 naturally occurring amino acids.
[000406] Embodiment 67. The polypeptide engineered according to any one of Embodiments 1 to 66, wherein said L1 linker is an unnatural amino acid incorporated by chemical synthesis, post-translational chemical modification, or through in vivo incorporation through expression recombinant in a host cell.
[000407] Embodiment 68. The polypeptide engineered according to any one of Embodiments 1 to 67, wherein the amino acids of said L1 linker are selected from serine, glycine, alanine, asparagine, proline, glutamine, glutamate, aspartate and lysine.
[000408] Embodiment 69. The polypeptide engineered according to any one of Embodiments 1 to 68, wherein said L1 linker comprises a majority of amino acids that are not sterically hindered.
[000409] Embodiment 70. The polypeptide engineered according to any one of Embodiments 1 to 69, wherein said L1 linker comprises one or more of the following: an acidic linker, a basic linker, and a structural motif.
[000410] Embodiment 71. The polypeptide engineered according to any one of Embodiments 1 to 70, wherein said L1 linker comprises polyglycine, polyalanine, poly(Gly-Ala) or poly(Gly-Ser).
[000411] Embodiment 72. The polypeptide engineered according to any one of Embodiments 1 to 71, wherein said L1 linker comprises a polyglycine of (Gly)3, (Gly)4 or (Gly)5.
[000412] Embodiment 73. The polypeptide engineered according to any one of Embodiments 1 to 72, wherein said L1 linker comprises (Gly)3Lys(Gly)4, (Gly)3AsnGlySer(Gly)2, (Gly)3Cys( Gly)4 and GlyProAsnGlyGly.
[000413] Embodiment 74. The polypeptide engineered according to any one of Embodiments 1 to 73, wherein said L1 linker comprises a combination of Gly and Ala.
[000414] Embodiment 75. The polypeptide engineered according to any one of Embodiments 1 to 74, wherein said L1 linker comprises a combination of Gly and Ser.
[000415] Embodiment 76. The polypeptide engineered according to any one of Embodiments 1 to 75, wherein said L1 linker comprises a combination of Gly and Glu.
[000416] Embodiment 77. The polypeptide engineered according to any one of Embodiments 1 to 76, wherein said L1 linker comprises a combination of Gly and Lys.
[000417] Embodiment 78. The polypeptide engineered according to any one of Embodiments 1 to 77, wherein said L1 linker comprises a sequence selected from the group consisting of: [Gly-Ser]n, [Gly-Gly-Ser] n, [Gly-Gly-Gly-Ser]n, and [Gly-Gly-Gly-Gly-Ser]n, where n is 1, 2, 3, 4, 5, 10 6, 7, 8, 9 or 10.
[000418] Embodiment 79. The polypeptide engineered according to any one of Embodiments 1 to 78, wherein said L1 linker comprises a sequence selected from the group consisting of: [Gly-Glu]n, [Gly-Gly-Glu] n, [Gly-Gly-Gly-Glu]n, [Gly-Gly-Gly-Gly-Glu]n, [Gly-Asp]n, [Gly-Gly-Asp]n, [Gly-Gly-Gly-Asp ]n, [Gly-Gly-Gly-Gly-Asp]n, where n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10.
[000419] Embodiment 80. The polypeptide engineered according to any one of Embodiments 1 to 79, wherein said L1 linker comprises a sequence selected from the group consisting of: [Gly-Glu]n, [Gly-Gly-Glu] n, [Gly-Gly-Gly-Glu]n, [Gly-Gly-Gly-Gly-Glu]n, [Gly-Asp]n, [Gly-Gly-Asp]n, [Gly-Gly-Gly-Asp ]n, [Gly-Gly-Gly -Gly-Asp]n, where n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10.
[000420] Embodiment 81. The polypeptide manipulated according to any one of Embodiments 1 to 80, wherein said L1 linker comprises a sequence selected from the group consisting of: [Gly-Lys]n, [Gly-Gly-Lys] n, [Gly-Gly-Gly-Lys]n, [Gly-Gly-Gly-Gly-Lys]n, [Gly-Arg]n, [Gly-Gly-Arg]n, [Gly-Gly-Gly-Arg ]n, [Gly-Gly-Gly-Gly-Arg]n, where n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10.
[000421] Embodiment 82. The polypeptide manipulated according to any one of Embodiments 1 to 81, wherein said L1 linker comprises a sequence selected from the group consisting of: [Glu-Ala-Ala-Ala-Lys]n, where n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10.
[000422] Embodiment 83. The polypeptide engineered according to any one of Embodiments 1 to 81, wherein said L1 linker comprises a sequence selected from the group consisting of: [Gly-Gly-Glu]6[Gly-Gly-Lys ]6, [Glu-Ala-Ala-Ala-Lys]3, [Glu-Ala-Ala-Ala-Lys]4 or [Glu-Ala-Ala-Ala-Lys]5.
[000423] Embodiment 84. The polypeptide engineered according to any one of Embodiments 1 to 83, wherein said L1 linker comprises an N-terminal TG dipeptide.
[000424] Embodiment 85. The polypeptide engineered according to any one of Embodiments 1 to 84, wherein said L1 linker comprises a C-terminal AS dipeptide.
[000425] Embodiment 86. The polypeptide engineered according to any one of Embodiments 1 to 85, wherein said L1 linker comprises an N-terminal TG dipeptide and a C-terminal AS dipeptide.
[000426] Embodiment 87. The polypeptide engineered according to any one of Embodiments 1 to 86, wherein said L1 linker comprises an amino acid sequence that is selected from the group consisting of TG-(GGGS)1, TG-(GGGS )2, TG-(GGGS)3, TG-(GGGS)4, TG-(GGGS)5, (GGGS)1-AS, (GGGS)2-AS, (GGGS)3-AS, (GGGS)4- AS, (GGGS)5-AS, TG-(GGGS)1-AS, TG-(GGGS)2-AS, TG-(GGGS)3-AS, TG-(GGGS)4-AS and TG-(GGGS) 5-AS
[000427] Embodiment 88. The polypeptide engineered according to any one of Embodiments 1 to 87, wherein said TG dipeptide and/or said AS dipeptide are absent or substituted by a pair of amino acids selected from T, A, S and G.
[000428] Embodiment 89. The polypeptide engineered according to any one of Embodiments 1 to 88, wherein said polypeptide further comprises one or more additional linkers.
[000429] Embodiment 90. The engineered polypeptide according to any one of Embodiments 1 to 89, wherein said engineered polypeptide comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 84, SEQ ID NO: 85, SEQ ID NO: 86, SEQ ID NO: 87, SEQ ID NO: 88, SEQ ID NO: 89, SEQ ID NO: 90, SEQ ID NO: 91 , SEQ ID NO: 92, SEQ ID NO: 93, SEQ ID NO: 94, SEQ ID NO: 95, SEQ ID NO: 96, SEQ ID NO: 97, SEQ ID NO: 98, SEQ ID NO: 99, SEQ ID NO: 100, SEQ ID NO: 101, SEQ ID NO: 102, SEQ ID NO: 103, SEQ ID NO: 104, SEQ ID NO: 105, SEQ ID NO: 106 and SEQ ID NO: 107.
[000430] Embodiment 91. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 53.
[000431] Embodiment 92. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 54.
[000432] Embodiment 93. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 55.
[000433] Embodiment 94. The polypeptide engineering according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 56.
[000434] Embodiment 95. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 57.
[000435] Embodiment 96. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 58.
[000436] Embodiment 97. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 59.
[000437] Embodiment 98. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 60.
[000438] Embodiment 99. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 61.
[000439] Embodiment 100. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 62.
[000440] Embodiment 101. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 63.
[000441] Embodiment 102. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 64.
[000442] Embodiment 103. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 65.
[000443] Embodiment 104. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 66.
[000444] Embodiment 105. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 67.
[000445] Embodiment 106. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 68.
[000446] Embodiment 107. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 69.
[000447] Embodiment 108. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 70.
[000448] Embodiment 109. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 71.
[000449] Embodiment 110. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 72.
[000450] Embodiment 111. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 73.
[000451] Embodiment 112. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 74.
[000452] Embodiment 113. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 75.
[000453] Embodiment 114. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 76.
[000454] Embodiment 115. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 77.
[000455] Embodiment 116. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 78.
[000456] Embodiment 117. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 79.
[000457] Embodiment 118. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 80.
[000458] Embodiment 119. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 81.
[000459] Embodiment 120. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 82.
[000460] Embodiment 121. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 83.
[000461] Embodiment 122. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 84.
[000462] Embodiment 123. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 85.
[000463] Embodiment 124. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 86.
[000464] Embodiment 125. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 87.
[000465] Embodiment 126. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 88.
[000466] Embodiment 127. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 89.
[000467] Embodiment 128. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 90.
[000468] Embodiment 129. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 91.
[000469] Embodiment 130. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 92.
[000470] Embodiment 131. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 93.
[000471] Embodiment 132. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 94.
[000472] Embodiment 133. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 95.
[000473] Embodiment 134. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 96.
[000474] Embodiment 135. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 97.
[000475] Embodiment 136. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 98.
[000476] Embodiment 137. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 99.
[000477] Embodiment 138. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 100.
[000478] Embodiment 139. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 102.
[000479] Embodiment 140. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 102.
[000480] Embodiment 141. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 103.
[000481] Embodiment 142. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 104.
[000482] Embodiment 143. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 105.
[000483] Embodiment 144. The engineered polypeptide according to any one of Embodiments 1 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 106.
[000484] Embodiment 145. The second engineered polypeptide with any one of Embodiments 51 to 90, wherein said engineered polypeptide comprises the selected amino acid sequence shown in SEQ ID NO: 107.
[000485] Embodiment 146. The polypeptide engineered according to any one of Embodiments 1 to 145, having affinity for serum albumin with a dissociation constant of less than about 10-6 mol/L.
[000486] Embodiment 147. The polypeptide engineered according to any one of Embodiments 1 to 146, having affinity for serum albumin with a dissociation constant of less than about 10-9 mol/L.
[000487] Embodiment 148. The polypeptide manipulated according to any one of Embodiments 1 to 147, having affinity for serum albumin with a dissociation constant of less than about 10-12 mol/L.
[000488] Embodiment 149. The polypeptide manipulated according to any one of Embodiments 1 to 148, wherein the polypeptide has a duration of action of at least 1 day.
[000489] Embodiment 150. The polypeptide manipulated according to any one of Embodiments 1 to 149, wherein the polypeptide has a duration of action of at least 3 days.
[000490] Embodiment 151. The polypeptide manipulated according to any one of Embodiments 1 to 150, wherein the polypeptide has a duration of action of at least 5 days.
[000491] Embodiment 152. The polypeptide manipulated according to any one of Embodiments 1 to 151, wherein the polypeptide has a duration of action of at least 5 days in a human subject.
[000492] Embodiment 153. A method for treating a disease or disorder in a subject comprising administering a polypeptide engineered according to any one of Embodiments 1 to 152 and 170-192 to a subject in need thereof in an effective amount to treat said disease or disorder.
[000493] Embodiment 154. The method according to Embodiment 153, wherein said disease or disorder may be lipodystrophy, dyslipidemia, hyperlipidemia, obesity, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin deficiency, fatty liver disease, diabetes (including Type I and Type II), non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver disease (NAFLD), and metabolic syndrome X.
[000494] Embodiment 155. The method according to Embodiment 153 or 154, wherein said disease or disorder is lipodystrophy, dyslipidemia, hyperlipidemia, obesity, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin deficiency, fatty liver disease or diabetes.
[000495] Embodiment 156. The method according to any one of Embodiments 153 to 155, wherein said disease or disorder is type I diabetes or type II diabetes.
[000496] Embodiment 157. The method according to any one of Embodiments 153 to 155, wherein said disease or disorder is obesity.
[000497] Embodiment 158. The method according to any one of Embodiments 153 to 155, wherein said disease or disorder is lipodystrophy or leptin deficiency.
[000498] Embodiment 159. A pharmaceutical composition comprising a polypeptide engineered according to any one of Embodiments 1 to 152 and a pharmaceutically acceptable excipient.
[000499] Embodiment 160. The pharmaceutical composition according to Embodiment 159, wherein said pharmaceutical composition is an injectable pharmaceutical composition.
[000500] Embodiment 161. The pharmaceutical composition according to any one of Embodiments 159 to 160, wherein said pharmaceutical composition is a sustained-release or long-term pharmaceutical composition.
[000501] Embodiment 162. The pharmaceutical composition according to any one of Embodiments 159 to 161, wherein said pharmaceutical composition is a once-daily pharmaceutical composition.
[000502] Embodiment 163. The pharmaceutical composition according to any one of Embodiments 159 to 161, wherein said pharmaceutical composition is a once-weekly pharmaceutical composition.
[000503] Embodiment 164. A pharmaceutical composition of any one of embodiments 159 to 163 for treating a disease or disorder in a subject.
[000504] Embodiment 165. The pharmaceutical composition according to any one of Embodiments 159 to 164, wherein the disease or disorder is lipodystrophy, dyslipidemia, hyperlipidemia, obesity, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin deficiency, fatty liver, diabetes (including Type I and Type II), non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver disease (NAFLD) and metabolic syndrome X.
[000505] Embodiment 166. The pharmaceutical composition of Embodiment 164 or 165, wherein said disease or disorder is lipodystrophy, dyslipidemia, hyperlipidemia, obesity, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin deficiency, fatty liver disease, or diabetes .
[000506] Embodiment 167. The method according to any one of Embodiments 164 to 166, wherein said disease or disorder is type I diabetes or type II diabetes.
[000507] Embodiment 168. The method according to any one of Embodiments 164 to 166, wherein said disease or disorder is obesity.
[000508] Embodiment 169. The method according to any one of Embodiments 164 to 166, wherein said disease or disorder is lipodystrophy or leptin deficiency.
[000509] Embodiment 170. The polypeptide engineered according to any one of Embodiments 1 to 18, wherein said HD1 is selected from the group consisting of: (a) the amino acid sequence 1-146 of a leptin selected from the group that consists of: SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID 25 NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146, wherein a different amino acid is substituted at one or more of the following positions and retaining the same numbering (even in the absence of a glutaminyl residue at position 28): 4, 32, 33, 35, 50, 64, 68, 71, 74, 77, 78, 89, 97, 100, 102, 105, 106, 107, 108, 111, 118, 136, 138, 142 and 145; (b) the amino acid sequence of subpart (a), in which the glutaminyl residue at position 28 is absent; (c) the amino acid sequence of subparts (a) or (b), in which a methionyl residue is added to the N-terminus; (d) a leptin consisting of a fragment of the amino acid sequence of (a), (b) or (c) selected from the group consisting of: (i) amino acids 98-146; (ii) amino acids 1-32; (iii) amino acids 40-116, (iv) amino acids 1-99 and 112-146; (v) amino acids 1-99 and 112-146, wherein one or more of amino acids 100-111 are located between amino acids 99 and 112; (vi) the amino acid sequence of subpart (i) wherein one or more of amino acids 100, 102, 105, 106, 107, 108, 111, 118, 136, 138, 142 and 145 are replaced by another amino acid, (vii) the amino acid sequence of subpart (ii) wherein one or more of amino acids 4, 8 and 32 are replaced by another amino acid, (viii) the amino acid sequence of subpart (iii), wherein one or more of amino acids 50, 53, 60, 64, 66, 67, 68, 71, 74, 77, 78, 89, 97, 100, 102, 105, 106, 107, 108, 111 and 112 are replaced by another amino acid , (ix) the amino acid sequence of subpart (iv) wherein one or more of amino acids 4, 8, 32, 33, 35, 48, 50, 53, 60, 64, 66, 67, 68, 71, 74, 77, 78, 89, 97, 112, 30, 118, 136, 138, 142 and 145 are substituted with another amino acid; and (x) the amino acid sequence of subpart (v) wherein one or more of amino acids 4, 32, 33, 35, 50, 64, 68, 71, 74, 77, 78, 89, 97, 100, 102, 105, 106, 107, 108, 111, 118, 136, 138, 142 and 145 are substituted with another amino acid; (xi) the amino acid sequence of any one of sub-parts (i) - (x), wherein a methionine has been added to the N-terminus; (e) the amino acid sequence of any one of subparts (a) to (d) wherein said amino acid sequence is linked to a chemical moiety; (f) the amino acid sequence of subpart (e) wherein said chemical moiety is a water-soluble polymeric moiety; (g) the amino acid sequence of subpart (f) wherein said water-soluble polymeric moiety is selected from the group consisting of: polyethylene glycol, an ethylene glycol/propylene glycol copolymer, a carboxy methyl cellulose, dextran, polyvinyl alcohol , polyvinylpyrrolidone, a poly-1,3-dioxolane, poly-1,3,6-trioxane, an ethylene/maleic anhydride copolymer, a polyamino acid homopolymer, a random polyamino acid copolymer, an albumin, an Fc protein, a poly (n-vinyl pyrrolidone) polyethylene glycol, a propylene glycol homopolymer, a polypropylene oxide/ethylene oxide copolymer, a polyoxyethylated polyol, a polyvinyl alcohol, a polyethylene glycol propionaldehyde, a succinate and a styrene; (h) the amino acid sequence of subpart (g) wherein said water-soluble polymeric moiety is a polyethylene glycol and; (i) the amino acid sequence of subpart (g) wherein said water-soluble polymer is a polyamino acid selected from the group consisting of: an albumin, an antibody, an Fc protein, and a polylysine moiety.
[000510] Embodiment 171. The polypeptide engineered according to any one of Embodiments 1 to 18 and 170, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27 , SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and from SEQ ID NO: 146; wherein one or more amino acid substitutions have been made.
[000511] Embodiment 172. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 10 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which an amino acid substitution has been made.
[000512] Embodiment 173. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 20 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which two amino acid substitutions were made.
[000513] Embodiment 174. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 30 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and from SEQ ID NO: 146; in which three amino acid substitutions were made.
[000514] Embodiment 175. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 10 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which four amino acid substitutions were made.
[000515] Embodiment 176. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 20 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which five amino acid substitutions were made.
[000516] Embodiment 177. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 30 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and from SEQ ID NO: 146; in which six amino acid substitutions were made.
[000517] Embodiment 178. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 10 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which seven amino acid substitutions were made.
[000518] Embodiment 179. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 20 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which eight amino acid substitutions were made.
[000519] Embodiment 180. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 30 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and from SEQ ID NO: 146; in which nine amino acid substitutions were made.
[000520] Embodiment 181. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 10 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which 10 amino acid substitutions were made.
[000521] Embodiment 182. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 20 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which 11 amino acid substitutions were made.
[000522] Embodiment 183. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 30 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and from SEQ ID NO: 146; in which 12 amino acid substitutions were made.
[000523] Embodiment 184. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 10 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which 13 amino acid substitutions were made.
[000524] Embodiment 185. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 20 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which 14 amino acid substitutions were made.
[000525] Embodiment 186. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 30 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and from SEQ ID NO: 146; in which 15 amino acid substitutions were made.
[000526] Embodiment 187. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 10 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which 16 amino acid substitutions were made.
[000527] Embodiment 188. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 20 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which 17 amino acid substitutions were made.
[000528] Embodiment 189. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 30 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and from SEQ ID NO: 146; in which 18 amino acid substitutions were made.
[000529] Embodiment 190. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 10 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which 19 amino acid substitutions were made.
[000530] Embodiment 191. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID 20 NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and SEQ ID NO: 146; in which 20 amino acid substitutions were made.
[000531] Embodiment 192. The polypeptide engineered according to any one of Embodiments 1 to 18 and 171, wherein said HD1 comprises an amino acid sequence selected from the group consisting of: SEQ ID NO: 1, SEQ ID NO: 2 , SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO : 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27 , SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 and from SEQ ID NO: 146; in which 21 amino acid substitutions were made.
[000532] Embodiment 193. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 143.
[000533] Embodiment 194. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 144.
[000534] Embodiment 195. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 145.
[000535] Embodiment 196. The polypeptide engineered according to any one of Embodiments 1 to 20, wherein said HD1 is SEQ ID NO: 146.
[000536] While the foregoing description describes the present invention, with examples provided for purposes of illustration, it will be understood that the practice of the present invention embraces all customary variations, adaptations or modifications as being within the scope of the claimed invention. Therefore, the descriptions and examples should not be construed as limiting the scope of the invention, which is outlined by the appended claims. Informal Sequence Listing
[000537] The following is an informal listing of sequences described here: VPIQKVQDDTKTLIKTIVTRINDISHT-Xaa- SVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQILTSMPSRNVIQIS NDLENLRDLLHVLAFSKSCHLPQASGLETLESLGGVLEASGYSTEVV ALSRLQGSLQDMLQQLDLSPGC, where Xaa at position 28 is absent Q or ID 28 is. VPIQKVQDDTKTLIKTIVTRINDISHTQSVSAKQRVTGLDFIPGLHPILSL SKMDQTLAVYQQVLTSLPSQNVLQIANDLENLRDLLHLLAFSKSCSLP QTSGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDVSPEC (SEQ ID NO: 2). VPIQKVQDDTKTLIKTIVTRINDISHTSVSAKQRVTGLDFIPGLHPILSLS KMDQTLAVYQQVLTSLPSQNVLQIANDLENLRDLLHLLAFSKSCSLPQ TSGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDVSPEC (SEQ ID NO: 3). MVPIQKVQDDTKTLIKTIVTRINDISHT-Xaa- SVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQILTSMPSRNVIQIS NDLENLRDLLHVLAFSKSCHLPQASGLETLESLGGVLEASGYSTEVV ALSRLQGSLQDMLQQLDLSPGC, where Xaa at position 29 is either Q or is absent (SEQ ID NO: 4). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSAKQRVTGLDFIPGLHPIL SLSKMDQTLAVYQQVLTSLPSQNVLQIANDLENLRDLLHLLAFSKSCS LPQTSGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDVSP EC (SEQ ID NO: 5). MVPIQKVQDDTKTLIKTIVTRINDISHTSVSAKQRVTGLDFIPGLHPILSL SKMDQTLAVYQQVLTSLPSQNVLQIANDLENLRDLLHLLAFSKSCSLP QTSGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDVSPEC (SEQ ID NO: 6). VPIWRVQDDTKTLIKTIVTRISDISHMQSVSSKQRVTGLDFIPGLHPVL SLSKMDQTLAIYQQILTSLPSRNVIQISNDLENLRDLLHLLASSKSCPLP QARALETLESLGGVLEASLYSTEVVALSRLQGALQDMLRQLDLSPGC (SEQ ID NO: 7). MVPIWRVQDDTKTLIKTIVTRISDISHMQSVSSKQRVTGLDFIPGLHPV LSLSKMDQTLAIYQQILTSLPSRNVIQISNDLENLRDLLHLLASSKSCPL PQARALETLESLGGVLEASLYSTEVVALSRLQGALQDMLRQLDLSPG C (SEQ ID NO: 8). VPICKVQDDTKTLIKTIVTRINDISHT-Xaa- SVSSKQRVTGLDFIPGLHPLLSKMDQTLAIYQQILTSLPSRNVVQIS NDLENLRDLLHLLAASKSCPLPQVRALESLESLGVVLEASLYSTEVVA LSRLQGSLQDMLRQLDLSPGC, where Xaa at position 28 is either Q or is absent (SEQ ID NO: 9). MVPICKVQDDTKTLIKTIVTRINDISHT-Xaa- SVSSKQRVTGLDFIPGLHPLLSKMDQTLAIYQQILTSLPSRNVVQIS NDLENLRDLLHLLAASKSCPLPQVRALESLESLGVVLEASLYSTEVVA LSRLQGSLQDMLRQLDLSPGC, where Xaa at position 29 is either Q or is absent (SEQ ID NO: 10). MHWGTLCGFLWLWPYLFYVQAVPIQKVQDDTKTLIKTIVTRINDISHT QSVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQILTSMPSRNVIQI SNDLENLRDLLHVLAFSKSCHLPWASGLETLDSLGGVLEASGYSTEV VALSRLQGSLQD MLWQLDLSPGC (SEQ ID NO: 11) Xaa-Xaa-VPIQKVQDDTKTLIKTIVTRINDISH SVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQILTSMPSRNVIQIS NDLENLRDLLHVLAFSKSCHLPWASGLETLDSLGGVLEASGYSTEVV ALSRLQGSLQDMLWQLDLSPGC wherein: Xaa at position 27 is T or A; and Xaa at position 28 is Q or is absent (SEQ ID NO: 12). MVPIQKVQDDTKTLIKTIVTRINDISH-Xaa-Xaa- SVSSKQKVTGLDFIPGLHPI LTLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSC HLPWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDL SPGC, where: Xaa at position 28 is T or A ; and Xaa at position 29 is Q or is absent (SEQ ID NO: 13). VPIQKVQSDTKTLIKTIVTRINDISHTQSVSSKQRVTGLDFIPGLHPVLT LSQMDQTLAIYQQILINLPSRNVIQISNDLENLRDLLHLLAFSKSCHLPL ASGLETLESLGDVLEASLYSTEVVALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO: 14). MVPIQKVQSDTKTLIKTIVTRINDISHTQSVSSKQRVTGLDFIPGLHPVL TLSQMDQTLAIYQILINLPSRNVIQISNDLENLRDLLHLLAFSKSCHLP LASGLETLESLGDVLEASLYSTEVVALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO: 15). VPIHKVQDDTKTLIKTIVTRINDISHTQSVSARQRVTGLDFIPGLHPILSL SKMDQTLAVYQQILTSLPSQNVLQIAHDLENLRDLLHLLAFSKSCSLP QTRGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDLSPEC (SEQ ID NO: 16). MVPIHKVQDDTKTLIKTIVTRINDISHTQSVSARQRVTGLDFIPGLHPIL SLSKMDQTLAVYQQILTSLPSQNVLQIAHDLENLRDLLHLLAFSKSCSL PQTRGLQKPESLDGVLEASLYSTEVVALSRLQGSLQDILQQLDLSPE C (SEQ ID NO: 17). ISIEKIQADTKTLTKTIITRIIQLSTQNGVSTDQRVSGLDFIPGNQQFQNL ADMDQTLAVYQQILSSLPMPDRTQISNDLENLRSLFALLATLKNCPFT RSDGLDTMEIWGGIVEESLYSTEVVTLDRLRKSLKNIEKQLDHIQG (SEQ ID NO: 18). MRCILLYGFLCVWQHLYYSHPISIEKIQADTKTLTKTIITRIIQLSTQNGV STDQRVSGLDFIPGNQQFQNLADMDQTLAVYQQILSSLPMPDRTQIS NDLENLRSLFALLATLKNCPFTRSDGLDTMEIWGGIVEESLYSTEVVT LDRLRKSLKNIEKQLDHIQG (SEQ ID NO: 19). VPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPG C (SEQ ID NO: 20). VPIQKVQDDTKTLIKTIVTRINDISHAQSVSSKQKVTGLDFIPGLHPILTL SKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPG C (SEQ ID NO: 21). VPIQKVQDDTKTLIKTIVTRINDISHTSVSSKQKVTGLDFIPGLHPILTLS KMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLPW ASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO: 22). VPIQKVQDDTKTLIKTIVTRINDISHASVSSKQKVTGLDFIPGLHPILTLS KMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLPW ASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO: 23). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLS PGC (SEQ ID NO: 24). MVPIQKVQDDTKTLIKTIVTRINDISHAQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLS PGC (SEQ ID NO: 25). MVPIQKVQDDTKTLIKTIVTRINDISHTSVSSKQKVTGLDFIPGLHPILTL SKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPG C (SEQ ID NO: 26). MVPIQKVQDDTKTLIKTIVTRINDISHASVSSKQKVTGLDFIPGLHPILTL SKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPG C (SEQ ID NO: 27). PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 28). PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 29). PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 30). MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 31). MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 32). MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 33). MDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQD WLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELT KNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFL YSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKVPIQK VQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPILTLSKMD QTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHLPWASG LETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPGC (SEQ ID NO: 34) MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLQQLDLSP GC (SEQ ID NO: 35). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQICNDLENLRDLLHVLAFSKSCH LPWASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDMLWQLDLS PGC (SEQ ID NO: 36). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLEFIPGLHPILT LSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCHL PQASGLETLESLGGVLEASGYSTEVVALSRLQGSLQDMLQQLDLSP GC (SEQ ID NO: 37). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLEFIPGLHPILT LSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQASGLETLESLGEVLEASGYSTEVVALSRLQGSLQDILQQLDLSPE C (SEQ ID NO: 38). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDILQQLDLSPE C (SEQ ID NO: 39). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDILQQLDLSPE C (SEQ ID NO: 40). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDILQQLDLSPE C (SEQ ID NO: 41). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDILQQLDLSPE C (SEQ ID NO: 42). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDILQQLDVSP EC (SEQ ID NO: 43). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQTSGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDILQQLDLSPE C (SEQ ID NO: 44). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQASGLETLESLGEVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPE C (SEQ ID NO: 45). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDILQQLDLSP EC (SEQ ID NO: 46). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDILQQLDVSP EC (SEQ ID NO: 47). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQTSGLETLDSLGGVLEASGYSTEVVALSRLQGSLQDILQQLDVSPE C (SEQ ID NO: 48). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCSL PQTSGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDMLWQLDLSPE C (SEQ ID NO: 49). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDMLQQLDLSP GC (SEQ ID NO: 50). MVPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPIL TLSKMDQTLAVYQQILTSMPSRNVIQISNDLENLRDLLHVLAFSKSCH LPQASGLETLDSLGEVLEASGYSTEVVALSRLQGSLQDMLQQLDLSP EC (SEQ ID NO: 51). PIQKVQDDTKTLIKTIVTRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 52). MPIQKVQDDTKTLIKTIVTRINDISPPQGVCSRPRVAGLDFIPRVQSVR TLSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 53). PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SKMDQTLAVYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 54). MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSKMDQTLAVYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 55). PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 56). MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 57). PIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 58). MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 59). PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 60). MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 61). PIQKVQDDTKTLIKTIVTRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 62) MPIQKVQDDTKTLIKTIVTRINDISPPQGVCSRPRVAGLDFIPRVQSVR TLSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 63) PIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPRA RGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 64) MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 65) PIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 66) MPIQRVQDDTKTLIKTIITRINDISPPQGVCSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 67) PIQRVQDDTK TLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPGC (SEQ ID NO: 68) MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 69) PIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLPW ASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 70) MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 71) PIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTLS GMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLPW ASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 72) MPIQRVQDDTKTLIKTIITRINDISHTQSVSSKQKVTGLDFIPGLHPILTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 73). PIQKVQDDTKTLIKTIVTRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 74). MPIQKVQDDTKTLIKTIVTRINDISPPQGVSSRPRVAGLDFIPRVQSVR TLSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 75). PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SKMDQTLAVYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 76). MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSKMDQTLAVYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 77). PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 78). MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCPVP RARGSDTIKGLGNVLRASVHSTEVVALSRLQGSLQDMLWQLDLNPG C (SEQ ID NO: 79). PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 80). MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRSVVQIANDLANLRALLRLLASAKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 81). PIQKVQDDTKTLIKTIVTRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPVPR ARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPGC (SEQ ID NO: 82) MPIQKVQDDTKTLIKTIVTRINDISPPQGVSSRPRVAGLDFIPRVQSVR TLSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCPV PRARGSDTIKGLGNVLRASVHSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 83) PIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRTL SGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 84) MPIQRVQDDTKTLIKTIITRINDISPPQGVSSRPRVAGLDFIPRVQSVRT LSGMDQILATYQQILTSLQSRNVIQISNDLENLRDLLHVLAFSKSCHLP WASGLETLDSLGGVLEASGYSTEVVALSRLKAALQDMLRQLDRNPG C (SEQ ID NO: 85) KCNTATCATQRLANFLVRSSNNLGPVLPPTNVGSNTY (SEQ ID NO: 86 ); KCNTATCATQRLANFLVHSSNNFGAILSSTNVGSNTY (SEQ ID NO: 87); KCNTATCATQRLANFLVHSSNNFGPILPPTNVGSNTY (SEQ ID NO: 88). CGNLSTCMLGTYTQDFNKFHTFPQTAIGVGAP (SEQ ID NO: 89); CSNLSTCVLGKLSQELHKLQTYPRTNTGSGTP (SEQ ID NO: 90); KCNTATCVLGRLSQELHRLQTYPRTNTGSNTY (SEQ ID NO: 91). X'-Xaa1-Cys2-Asn3-Thr4-Ala5-Thr6-Cys7-Ala8-Thr9-Gln10-Arg11-Leu12-Ala13-Asn14-Phe15-Leu16-Val17-His18-Ser19-Ser20-Xaa21-Asn22-Phe23- Xaa24 - Xaa25- Xaa26- Xaa27- Xaa28- Xaa29-Thr30- Xaa31- Val32-Gly33-Ser34-Asn35-Thr36-Tyr37-X (SEQ ID NO: 92) CNTATCATQRLANFLVRSSNNLGPVLPPTNVGSNTY-NH2 (SEQ ID NO: 93) KCNTATNVSKQRLPV2 ID NO: 94) CNTATCATQRLANFLVRSSKNLGPVLPPTNVGSNTY-NH2 (SEQ ID NO: 95) KCNTATCATQRLANFLVRSSNNLGPKLPPTNVGSNTY-NH2 (SEQ ID NO: 96) CNTATCATQRLANFLVRSSNNLGPKLPPTNVGSNTY-NH2 (SEQ ID NO: 97) KCNTATCATQRLANFLVRSSNNLGPVLPPTKVGSNTY-NH2 (SEQ ID NO: 98) CNTATCATQRLANFLVRSSNNLGPVLPPTKVGSNTY-NH2 (SEQ ID NO: 99) KCNTATCATQRLANFLVHSSNNFGPILPPTNVGSNTY-NH2 (SEQ ID NO: 100) CNTATCATQRLANFLVHSSNNFGPILPPTNVGSNTY-NH2 (SEQ ID NO: 101) CNTATCATQRLANFLVHSSKNFGPILPPTNVGSNTY-NH2 (SEQ ID NO: 102) CNTATCATQRLANFLVHSSNNFGPKLPPTNVGSNTY-NH2 (SEQ ID NO: 103) CNTATCATQRLANFLVHSSNNFGPILPPTKVGSNTY-NH2 (SEQ ID NO: 104) CNTATCATQRLANFLVHSSNNFKPILPPTNVGSNTY-NH2 (SEQ ID NO: 105) CNTAT CATQRLANFLVHSSNNFGKILPPTNVGSNTY-NH2 (SEQ ID NO: 106) CNTATCATQRLANFLVHSSNNFGPIKPPTNVGSNTY-NH2 (SEQ ID NO: 107) CNTATCATQRLANFLVHSSNNFGPILKPTNVGSNTY-NH2 (SEQ ID NO: 108) CNTATCATQRLANFLVHSSNNFGPILPKTNVGSNQ1 ID-NH2
权利要求:
Claims (34)
[0001]
1. A constructed polypeptide, characterized in that it comprises: an albumin binding domain (ABD) polypeptide comprising an albumin binding motif (ABM) comprising the amino acid sequence: GVSD X5 YK X8 X9 I X11 X12 A X14 TVEGV X20 AL X23 X24 X25 I (SEQ ID NO: 34), wherein, independently of one another, X5 is selected from Y and F; X8 is selected from N, R and S; X9 is selected from V, I, L, M, F and Y; X11 is selected from N, S, E and D; X12 is selected from R, K and N; X14 is selected from K and R; X20 is selected from D, N, Q, E, H, S, R and K; X23 is selected from K, I and T; X24 is selected from A, S, T, G, H, L and D, and X25 is selected from H, E and D; and a first peptide hormone domain (HD1) having the amino acid sequence selected from: (a) SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO: 5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO: 30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, amino acids 2-147 of SEQ ID NO:31, and amino acids 2-147 of SEQ ID NO:33; or (b) the amino acid sequence 1-146 of a leptin selected from: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO :6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14 , SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22 SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO: 31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:143, SEQ ID NO:144, SEQ ID NO:145, and SEQ ID NO:146; wherein a different amino acid is substituted at one or more of the following positions and retaining the same numbering (even in the absence of a glutaminyl residue at position 28): 4, 32, 33, 35, 50, 64, 68, 71, 74, 77, 78, 89, 97, 100, 102, 105, 106, 107, 108, 111, 118, 136, 138, 142, and 145; (c) the amino acid sequence of subpart (b) in which the glutaminyl residue at position 28 is absent; (d) the amino acid sequence of subparts (b) or (c) wherein a methionyl residue is added to the N-terminus; (e) a leptin consisting of a fragment of the amino acid sequence of (b), (c) or (d) selected from: (i) amino acids 98-146; (ii) amino acids 1-32; (iii) amino acids 40-116; (iv) amino acids 1-99 and 112-146; (v) amino acids 1-99 and 112-146 wherein one or more of amino acids 100-111 is placed between amino acids 99 and 112; (vi) the amino acid sequence of subpart (i) wherein one or more of amino acids 100, 102, 105, 106, 107, 108, 111, 118, 136, 138, 142 and 145 are replaced by another amino acid; (vii) the amino acid sequence of subpart (ii) wherein one or more of amino acids 4, 8 and 32 are replaced by another amino acid; (viii) the amino acid sequence of subpart (iii) wherein one or more of amino acids 50, 53, 60, 64, 66, 67, 68, 71, 74, 77, 78, 89, 97, 100, 102, 105 , 106, 107, 108, 111 and 112 are substituted with another amino acid; (ix) the amino acid sequence of subpart (iv) wherein one or more of amino acids 4, 8, 32, 33, 35, 48, 50, 53, 60, 64, 66, 67, 68, 71, 74, 77 , 78, 89, 97, 112, 118, 136, 138, 142 and 145 is replaced by another amino acid; and (x) the amino acid sequence of subpart (v) wherein one or more of amino acids 4, 32, 33, 35, 50, 64, 68, 71, 74, 77, 78, 89, 97, 100, 102, 105, 106, 107, 108, 111, 118, 136, 138, 142 and 145 is substituted with another amino acid; (xi) the amino acid sequence of any one of subparts (i) (x) in which a methionine has been added to the N-terminus; (f) the amino acid sequence of any one of subparts (b) to (e) wherein said amino acid sequence is linked to a chemical moiety; (g) the amino acid sequence of subpart (f) wherein said chemical moiety is a water-soluble polymer moiety; (h) the amino acid sequence of subpart (g) wherein said water-soluble polymer fraction is selected from: polyethylene glycol, an ethylene glycol/propylene glycol copolymer, a carboxymethyl cellulose, a dextran, a polyvinyl alcohol, a polyvinylpyrolidone, a poly-1,3-dioxolane, a poly-1,3,6-trioxane, an ethylene/maleic anhydride copolymer, a polyamino acid homopolymer, a random polyamino acid copolymer, an albumin, an Fc protein, a poly (n-vinylpyrolidone), a propylene glycol homopolymer, a polypropylene oxide/ethylene oxide copolymer, a polyoxyethylated polyol, a polyvinyl alcohol, a polyethylene glycol propionaldehyde, a succinate and a styrene; (i) the amino acid sequence of subpart (h) wherein said water-soluble polymer moiety is a polyethylene glycol; and (j) the amino acid sequence of subpart (h) wherein said water-soluble polymer is a polyamino acid selected from: an albumin, an antibody, an Fc protein and a polylysine moiety; and a first linker (L1) covalently linked to said HD1, said linker comprising an amino acid sequence selected from Gly-Gly-Gly, [Gly-Ser]n, [Gly-Gly-Ser]n, [Gly- Gly-Gly-Ser]ne [Gly-Gly-Gly-Gly-Ser]n, where n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
[0002]
2. Constructed polypeptide according to claim 1, characterized in that HD1 has a sequence selected from the sequences of subpart (a).
[0003]
3. Polypeptide constructed, according to claim 2, characterized in that it comprises said ABD as an N-terminal fraction and said HD1 as a C-terminal fraction.
[0004]
4. Polypeptide constructed, according to claim 2, characterized in that it comprises said ABD as a C-terminal fraction and said HD1 as an N-terminal fraction.
[0005]
5. A constructed polypeptide according to any one of claims 1 to 4, characterized in that said HD1 has an amino acid sequence selected from: SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO: 9, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, amino acids 2-147 of SEQ ID NO:31, and amino acids 2-147 of SEQ ID NO:33.
[0006]
6. A constructed polypeptide according to any one of claims 1 to 5, characterized in that said HD1 has an amino acid sequence selected from: SEQ ID NO: 26, SEQ ID NO: 29, SEQ ID NO: 33 and amino acids 2-147 of SEQ ID NO: 33.
[0007]
7. Constructed polypeptide according to any one of claims 1 to 6, characterized in that, independently of one another, X5 is Y; X8 is N; X23 is T or I; X24 is S or L; and X25 is E or H.
[0008]
8. A constructed polypeptide according to any one of claims 1 to 7, characterized in that the albumin binding motif comprises an amino acid sequence that is selected from: GVSDYYKNLINKAKTVEGVEALTLHI (SEQ ID NO:114) and GVSDYYKNLINKAKTVEGVEALISEI ( SEQ ID NO:115).
[0009]
9. A constructed polypeptide according to any one of claims 1 to 8, characterized in that said ABD comprises an albumin binding motif (ABM) other than GVSDYYKNLINNAKTVEGVKALIDEI (SEQ ID NO: 35).
[0010]
10. A constructed polypeptide according to any one of claims 1 to 9, characterized in that the albumin-binding domain (ABD) polypeptide comprises an amino acid sequence that is selected from: LAEAKEDAIKELDKYGVSDYYKNLINKAKTVEGVEALTLHIL AALP (SEQ ID NO :50); eLAEAKEDAIKELDKYGVSDYYKNLINKAKTVEGVEALISEILAALP (SEQ ID NO:51).
[0011]
11. Polypeptide constructed according to claim 1 or 2, characterized in that said ABD comprises the amino acid sequence: LAEAK Xa Xb A Xc Xd EL Xe KY -[ABM]- LAALP (SEQ ID NO:36) wherein [ABM] is the albumin-binding motif, and, independently of one another, Xa is selected from V and E; Xb is selected from L, E and D; Xc is selected from N, L and I; Xd is selected from R and K; Xe is selected from D and K; leucine at position 45 is present or absent; and the proline at position 46 is either present or absent.
[0012]
12. A constructed polypeptide according to claim 1, characterized in that said ABD comprises an amino acid sequence with at least 85% identity with an amino acid sequence that is selected from: SEQ ID NO: 37, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:50, SEQ ID NO:51 and SEQ ID NO:52.
[0013]
13. Polypeptide constructed, according to claim 1 or 2, characterized in that said ABD comprises any of the peptides selected from: LAEAKVLANRELDKYGVSDFYKRLINKAKTVEGVNALTHHI LAALP (SEQ ID NO:39), LAEAKVLANRELDKYGVSDYYKNLINRARTVEGVHALIDHI LAALP (SEQ ID NO:40 ) LAEAKVLANRELDKYGVSDYYKNIINRAKTVEGVRALKLHI LAALP (SEQ ID NO: 41), LAEAKVLANRELDKYGVSDYYKNLINKAKTVEGVEALTLHI LAALP (SEQ ID NO: 43), LAEAKVLANRELDKYGVSDFYKNLINRAKTVEGVDALIAHI LAALP (SEQ ID NO: 44), LAEAKVLANRELDKYGVSDFYKSLINRAKTVEGVDALTSHI LAALP (SEQ ID NO: 45), LAEAKVLANRELDKYGVSDFYKNVINKAKTVEGVEALIADI LAALP (SEQ ID NO: 47 ) LAEAKVLANRELDKYGVSDYYKNLINKAKTVEGVQALIAHI LAALP (SEQ ID NO: 48), LAEAKVLANRELDKYGVSDFYKRLINKAKTVEGVEALKLHI LAALP (SEQ ID NO: 49), LAEAKEDAIKELDKYGVSDYYKNLINKAKTVEGVEALTLHIL AALP (SEQ ID NO: 50), LAEAKEDAIKELDKYGVSDYYKNLINKAKTVEGVEALiseILA ALP (SEQ ID NO: 51) and LAEAKEDAIKELDKYGVSDYYKRLISKAKTVEGVKALISEIL AALP (SEQ ID NO: 52).
[0014]
14. Constructed polypeptide according to claim 1 or 2, characterized in that said ABD comprises the amino acid sequence of SEQ ID NO: 49.
[0015]
15. Polypeptide constructed according to any one of claims 1 to 14, characterized in that said L1 ligand is a peptide of 1 to 30 amino acids or less than 30 amino acids.
[0016]
16. Constructed polypeptide according to any one of claims 1 to 15, characterized in that said L1 ligand comprises an N-terminal TG dipeptide or a C-terminal AS dipeptide, or both.
[0017]
17. A constructed polypeptide according to any one of claims 1 to 16, characterized in that said L1 ligand comprises an amino acid sequence that is selected from: TG-(GGGS)1 (SEQ ID NO: 215) , TG-(GGGS)2 (SEQ ID NO: 216), TG(GGGS)3 (SEQ ID NO: 217), TG-(GGGS)4 (SEQ ID NO: 218), TG-(GGGS)5 (SEQ ID NO: 219), (GGGS)1-AS (SEQ ID NO: 220), (GGGS)2-AS (SEQ ID NO: 221), (GGGS)3-AS (SEQ ID NO: 222), (GGGS )4-AS (SEQ ID NO: 223), (GGGS)5AS (SEQ ID NO: 224), TG-(GGGS)1-AS (SEQ ID NO: 225), TG-(GGGS)2-AS (SEQ ID NO: 226), TG-(GGGS)3-AS (SEQ ID NO: 227), TG (GGGS)4-AS (SEQ ID NO: 228), and TG-(GGGS)5-AS (SEQ ID NO : 229).
[0018]
18. A constructed polypeptide according to any one of claims 1 to 17, characterized in that it comprises an amino acid sequence selected from: SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:62, SEQ ID NO:63, SEQ ID NO :64, SEQ ID NO:65, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:70, SEQ ID NO:71, SEQ ID NO:72 , SEQ ID NO:73, SEQ ID NO:74, SEQ ID NO:75, SEQ ID NO:76, SEQ ID NO:77, SEQ ID NO:78, SEQ ID NO:79, SEQ ID NO:80, SEQ ID NO:81, SEQ ID NO:82, SEQ ID NO:83, SEQ ID NO:84, SEQ ID NO:85, SEQ ID NO:86, SEQ ID NO:87, SEQ ID NO:88, SEQ ID NO: 89, SEQ ID NO:90, SEQ ID NO:91, SEQ ID NO:92, SEQ ID NO:93, SEQ ID NO:94, SEQ ID NO:95, SEQ ID NO:96, SEQ ID NO:97, SEQ ID NO:98, SEQ ID NO:99, SEQ ID NO:100, SEQ ID NO:101, SEQ ID NO:102, SEQ ID NO:103, SEQ ID NO:104, SEQ ID NO:105, SEQ ID NO:106, and SEQ ID NO:107.
[0019]
19. A constructed polypeptide according to claim 18, characterized in that it comprises an amino acid sequence selected from: SEQ ID NO: 54 and SEQ ID NO: 61.
[0020]
20. Constructed polypeptide according to claim 1, characterized in that said HD1 has a sequence selected from the sequences in subparts (b) - (j).
[0021]
21. A constructed polypeptide according to claim 20, characterized in that said HD1 comprises an amino acid sequence selected from: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO: 12, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:143, SEQ ID NO:144, SEQ ID NO:145, and SEQ ID NO :146; wherein one or more amino acid substitutions have been made.
[0022]
22. The constructed polypeptide of claim 19, wherein said HD1 is SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145 or SEQ ID NO: 146.
[0023]
23. The constructed polypeptide of claim 20, wherein said HD1 is SEQ ID NO: 26, SEQ ID NO: 29 or SEQ ID NO: 33, wherein one or more amino acid substitutions have been made.
[0024]
24. Constructed polypeptide according to any one of claims 20 to 23, characterized in that said ABD comprises the amino acid sequence of SEQ ID NO: 49.
[0025]
25. A constructed polypeptide according to any one of claims 1 to 24, characterized in that it is for use in treating a disease or disorder in an individual.
[0026]
26. A constructed polypeptide according to any one of claims 1 to 24, characterized in that it is for use in the treatment of a disease or disorder selected from lipodystrophy, dyslipidemia, hyperlipidemia, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin, fatty liver disease, diabetes (including type I and type II), non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver disease (NAFLD), metabolic syndrome X and Huntington's disease.
[0027]
27. Polypeptide constructed according to claim 26, characterized in that said disease or disorder is lipodystrophy, dyslipidemia, hyperlipidemia, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin deficiency, fatty liver disease or diabetes.
[0028]
28. Pharmaceutical composition, characterized in that it comprises a polypeptide constructed, as defined in any one of claims 1 to 24, and a pharmaceutically acceptable excipient.
[0029]
29. Pharmaceutical composition, according to claim 28, characterized in that it is an injectable pharmaceutical composition.
[0030]
30. Pharmaceutical composition, according to claim 28 or 29, characterized in that it is a sustained-release or long-lasting pharmaceutical composition.
[0031]
31. Pharmaceutical composition according to any one of claims 28 to 30, characterized in that it is for use in the treatment of lipodystrophy, dyslipidemia, hyperlipidemia, obesity, hypothalamic amenorrhea, Alzheimer's disease, leptin deficiency, fatty liver disease, diabetes (including type I and type II), non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver disease (NAFLD), or metabolic syndrome X.
[0032]
32. Use of a constructed polypeptide, as defined in any one of claims 1 to 24, for the manufacture of a pharmaceutical composition for the treatment of a disease or disorder in an individual.
[0033]
33. Use of at least two different anti-obesity agents, wherein at least one anti-obesity agent is a polyethylene glycol (PEG) conjugated amylin and at least one anti-obesity agent is a constructed polypeptide as defined in any one of claims 1 to 24, said use characterized in that it is for the manufacture of a pharmaceutical composition for the treatment of obesity in an individual.
[0034]
34. Use of at least two different anti-obesity agents, wherein at least one anti-obesity agent is a polyethylene glycol (PEG) conjugated amylin and at least one anti-obesity agent is a constructed polypeptide as defined in any one of claims 1 to 24, said use characterized by the fact that it is in the manufacture of a pharmaceutical composition to reduce the body weight of an individual.
类似技术:
公开号 | 公开日 | 专利标题
BR112013007388B1|2022-01-04|POLYPEPTIDE HAVING ALBUMIN AND LEPTIN BINDING DOMAIN, ITS USE IN THE TREATMENT OF DISEASES, AS WELL AS A PHARMACEUTICAL COMPOSITION THAT COMPRISES IT
US20180305430A1|2018-10-25|Engineered Polypeptides Having Enhanced Duration of Action and Reduced Immunogenicity
JP5743371B2|2015-07-01|Pancreatic polypeptide family motifs, polypeptides and methods comprising these
JP2013006841A|2013-01-10|Pancreatic polypeptide family motif, polypeptide and method comprising the same
US8748375B2|2014-06-10|Methods for affecting body composition using amylin agonists
同族专利:
公开号 | 公开日
EP2621515B1|2017-03-29|
ES2630031T3|2017-08-17|
US20130274182A1|2013-10-17|
CA2813038A1|2012-04-19|
DK2621515T3|2017-07-17|
CN103547590B|2017-11-28|
ES2873253T3|2021-11-03|
EP3241558A3|2018-05-16|
JP2014502252A|2014-01-30|
MX351128B|2017-10-03|
EA032917B1|2019-08-30|
CA2813038C|2021-12-28|
US20160083446A1|2016-03-24|
CN103403019A|2013-11-20|
PT2621519T|2017-10-04|
EP3305315A1|2018-04-11|
CA2813087C|2020-07-21|
US20200362006A1|2020-11-19|
EP2621519A4|2014-07-09|
WO2012050930A2|2012-04-19|
EA201390474A1|2013-07-30|
MX2013003472A|2013-10-30|
CY1119023T1|2018-01-10|
CN103547590A|2014-01-29|
EP2621515A4|2014-07-16|
WO2012050925A3|2012-07-05|
JP2020033366A|2020-03-05|
EP3241558A2|2017-11-08|
MX349054B|2017-07-07|
US10087228B2|2018-10-02|
CA2813087A1|2012-04-19|
WO2012050930A3|2012-06-14|
JP6174489B2|2017-08-02|
EP3241558B1|2021-03-03|
JP2017141232A|2017-08-17|
CA3138758A1|2012-04-19|
PL3241558T3|2021-08-30|
JP6412183B2|2018-10-24|
EA201390497A1|2013-09-30|
EA024507B1|2016-09-30|
CY1119498T1|2018-03-07|
EP2621515A2|2013-08-07|
US20130203661A1|2013-08-08|
DK2621519T3|2017-10-16|
BR112013007388A2|2020-10-06|
CN103403019B|2016-10-12|
PT2621515T|2017-07-12|
JP6608799B2|2019-11-20|
JP2013543497A|2013-12-05|
US20160137709A1|2016-05-19|
ES2641869T3|2017-11-14|
MX2013003482A|2013-10-25|
JP2017081939A|2017-05-18|
EP2621519A2|2013-08-07|
EP2621519B1|2017-06-28|
WO2012050925A2|2012-04-19|
DK3241558T3|2021-04-26|
BR112013007385A2|2021-04-20|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

US4179337A|1973-07-20|1979-12-18|Davis Frank F|Non-immunogenic polypeptides|
US4002531A|1976-01-22|1977-01-11|Pierce Chemical Company|Modifying enzymes with polyethylene glycol and product produced thereby|
US6936694B1|1982-05-06|2005-08-30|Intermune, Inc.|Manufacture and expression of large structural genes|
US4572208A|1983-06-29|1986-02-25|Utah Medical Products, Inc.|Metabolic gas monitoring apparatus and method|
US6319685B1|1984-09-27|2001-11-20|Unigene Laboratories, Inc.|Alpha-amidating enzyme compositions and processes for their production and use|
FI78231C|1984-11-21|1989-07-10|Instrumentarium Oy|Measuring device for metabolic quantities connectable to a respirator|
CA2006596C|1988-12-22|2000-09-05|Rika Ishikawa|Chemically-modified g-csf|
HU222249B1|1991-03-08|2003-05-28|Amylin Pharmaceuticals Inc.|Process for producing amyline antagonistic peptide derivatives and pharmaceutical preparatives containing them|
US5912015A|1992-03-12|1999-06-15|Alkermes Controlled Therapeutics, Inc.|Modulated release from biocompatible polymers|
US5766627A|1993-11-16|1998-06-16|Depotech|Multivescular liposomes with controlled release of encapsulated biologically active substances|
US6309853B1|1994-08-17|2001-10-30|The Rockfeller University|Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof|
US5824784A|1994-10-12|1998-10-20|Amgen Inc.|N-terminally chemically modified protein compositions and methods|
EP0725079A1|1995-01-31|1996-08-07|Eli Lilly And Company|Anti-obesity proteins|
US5605886A|1995-01-31|1997-02-25|Eli Lilly And Company|Anti-obesity proteins|
US5691309A|1995-01-31|1997-11-25|Eli Lilly And Company|Anti-obesity proteins|
US5552523A|1995-01-31|1996-09-03|Eli Lilly And Company|Anti-obesity proteins|
US5559208A|1995-01-31|1996-09-24|Eli Lilly And Company|Anti-obesity proteins|
US5554727A|1995-01-31|1996-09-10|Eli Lilly And Company|Anti-obesity proteins|
US5580954A|1995-01-31|1996-12-03|Eli Lilly And Company|Anti-obesity proteins|
US5594101A|1995-03-03|1997-01-14|Eli Lilly And Company|Anti-obesity proteins|
US5840517A|1995-04-26|1998-11-24|Eli Lilly And Company|Process for preparing obesity protein analogs|
EP0836479A2|1995-06-30|1998-04-22|Eli Lilly And Company|Methods for treating diabetes|
US6936439B2|1995-11-22|2005-08-30|Amgen Inc.|OB fusion protein compositions and methods|
GB9526733D0|1995-12-30|1996-02-28|Delta Biotechnology Ltd|Fusion proteins|
US6013009A|1996-03-12|2000-01-11|Karkanen; Kip Michael|Walking/running heart rate monitoring system|
KR20010029537A|1996-09-20|2001-04-06|훽스트 악티엔게젤샤프트|Use of leptin antagonists for treating insulin resistance in type II diabetes|
CA2275183A1|1996-12-20|1998-07-02|Amgen Inc.|Ob fusion protein compositions and methods|
US6309360B1|1997-03-17|2001-10-30|James R. Mault|Respiratory calorimeter|
AU6569998A|1997-03-20|1998-10-12|Eli Lilly And Company|Obesity protein formulations|
HU0003428A3|1997-06-06|2001-12-28|Smithkline Beecham Plc|Use of leptin antagonists for the treatment of diabetes|
ES2235336T3|1997-06-13|2005-07-01|Gryphon Therapeutics, Inc.|NATIVE CHEMICAL BINDING IN SOLID PHASE OF PEPTIDES DESPROTEGED OR PROTECTED IN THE N-TERMINAL CISTEINE IN A WATER SOLUTION.|
EP1091440B1|1998-05-29|2010-06-02|JGC Catalysts and Chemicals Ltd.|Method of manufacturing photoelectric cell|
US6660843B1|1998-10-23|2003-12-09|Amgen Inc.|Modified peptides as therapeutic agents|
US6194006B1|1998-12-30|2001-02-27|Alkermes Controlled Therapeutics Inc. Ii|Preparation of microparticles having a selected release profile|
KR20020001719A|1999-01-14|2002-01-09|마크 엠. 폴렛타|Method for Glucagon Suppression|
US6475984B2|1999-04-29|2002-11-05|The Nemours Foundation|Administration of leptin|
TW514510B|1999-06-11|2002-12-21|Tanita Seisakusho Kk|Method and apparatus for measuring distribution of body fat|
US6468222B1|1999-08-02|2002-10-22|Healthetech, Inc.|Metabolic calorimeter employing respiratory gas analysis|
WO2001021647A2|1999-09-22|2001-03-29|Genset|Methods of screening for compounds that modulate the lsr-leptin interaction and their use in the prevention and treatment of obesity-related diseases|
US6530886B1|1999-10-08|2003-03-11|Tanita Corporation|Method and apparatus for measuring subcutaneous fat using ultrasonic wave|
US7057015B1|1999-10-20|2006-06-06|The Salk Institute For Biological Studies|Hormone receptor functional dimers and methods of their use|
US7157564B1|2000-04-06|2007-01-02|Affymetrix, Inc.|Tag nucleic acids and probe arrays|
JP2003530846A|2000-04-12|2003-10-21|ヒューマンゲノムサイエンシズインコーポレイテッド|Albumin fusion protein|
US6264987B1|2000-05-19|2001-07-24|Alkermes Controlled Therapeutics Inc. Ii|Method for preparing microparticles having a selected polymer molecular weight|
TW515705B|2000-05-31|2003-01-01|Yamato Scale Co Ltd|Visceral fat meter|
US6492117B1|2000-07-12|2002-12-10|Gendaq Limited|Zinc finger polypeptides capable of binding DNA quadruplexes|
US6296842B1|2000-08-10|2001-10-02|Alkermes Controlled Therapeutics, Inc.|Process for the preparation of polymer-based sustained release compositions|
US6475158B1|2000-10-24|2002-11-05|Korr Medical Technologies, Inc.|Calorimetry systems and methods|
EP2219031B1|2001-10-22|2013-04-24|Amgen, Inc.|Use of leptin for treating human lipoatrophy and method of determining predisposition to said treatment|
US20050287153A1|2002-06-28|2005-12-29|Genentech, Inc.|Serum albumin binding peptides for tumor targeting|
AU2003290563A1|2002-10-31|2004-05-25|Clf Medical Technology Acceleration Program, Inc.|Leptin-related peptides|
CN1684990A|2002-11-01|2005-10-19|株式会社德山|Polymerizable composition, process for producing cured object thereof, and optical article|
JP2007537981A|2003-09-19|2007-12-27|ノボ ノルディスク アクティーゼルスカブ|Novel plasma protein affinity tag|
KR101241862B1|2003-09-19|2013-03-13|노보 노르디스크 에이/에스|Novel glp-1 derivatives|
JP2008500281A|2004-02-11|2008-01-10|アミリン・ファーマシューティカルズ,インコーポレイテッド|Amylin family peptides and methods for making and using them|
US7399744B2|2004-03-04|2008-07-15|Amylin Pharmaceuticals, Inc.|Methods for affecting body composition|
US8394765B2|2004-11-01|2013-03-12|Amylin Pharmaceuticals Llc|Methods of treating obesity with two different anti-obesity agents|
CA2647568A1|2006-03-31|2007-10-11|Amylin Pharmaceuticals, Inc.|Amylin and amylin agonists for treating psychiatric diseases and disorders|
US7898623B2|2005-07-04|2011-03-01|Semiconductor Energy Laboratory Co., Ltd.|Display device, electronic device and method of driving display device|
GB0524788D0|2005-12-05|2006-01-11|Affibody Ab|Polypeptides|
WO2007082264A2|2006-01-11|2007-07-19|Bristol-Myers Squibb Company|Human glucagon-like-peptide-1 modulators and their use in the treatment of diabetes and related conditions|
WO2007104789A2|2006-03-15|2007-09-20|Novo Nordisk A/S|Amylin derivatives|
EP2636680A3|2006-05-26|2013-12-11|Amylin Pharmaceuticals, LLC|Composition and methods for treatment of congestive heart failure|
US20100022457A1|2006-05-26|2010-01-28|Bristol-Myers Squibb Company|Sustained release glp-1 receptor modulators|
JP2008169195A|2007-01-05|2008-07-24|Hanmi Pharmaceutical Co Ltd|Insulinotopic peptide drug combo using carrier material|
CN101113175A|2007-04-28|2008-01-30|中国科学院西北高原生物研究所|Rat-rabbit family thin element protein and its cDNA sequence|
JP2009019027A|2007-07-16|2009-01-29|Hanmi Pharmaceutical Co Ltd|Insulin secretion peptide derivative in which amino acid of amino terminal is varied|
JP5718638B2|2007-07-31|2015-05-13|アフィボディ・アーベー|Novel compositions, methods and uses|
EP2036923A1|2007-09-11|2009-03-18|Novo Nordisk A/S|Improved derivates of amylin|
EP2195034A2|2007-09-27|2010-06-16|Amylin Pharmaceuticals, Inc.|Peptide-peptidase inhibitor conjugates and methods of making and using same|
JP2011503180A|2007-11-14|2011-01-27|アミリン・ファーマシューティカルズ,インコーポレイテッド|Methods for treating obesity and obesity-related diseases and disorders|
NZ620606A|2008-02-08|2015-08-28|Ambrx Inc|Modified leptin polypeptides and their uses|
US8501686B2|2008-06-05|2013-08-06|University Of Michigan|Method of treating fatty liver diseases and conditions in non-lipodystrophic subjects|
CN102197049B|2008-10-21|2015-03-25|诺沃-诺迪斯克有限公司|Amylin derivatives|
WO2010054699A1|2008-11-17|2010-05-20|Affibody Ab|Conjugates of albumin binding domain|
WO2010085700A2|2009-01-22|2010-07-29|Unigene Laboratories Inc.|Treatment for obesity|
DK2621515T3|2010-09-28|2017-07-17|Aegerion Pharmaceuticals Inc|Chimeric seal-human leptin polypeptide with increased solubility|
DK2729160T3|2011-07-08|2019-07-01|Aegerion Pharmaceuticals Inc|MANIPULATED POLYPEPTIDES WHICH HAVE IMPROVED EFFECT TIME AND REDUCED IMMUNOGENICITY|US6765372B2|2001-12-14|2004-07-20|Intersil Americas Inc.|Programmable current-sensing circuit providing continuous temperature compensation for DC-DC Converter|
EP2933262B1|2010-07-09|2018-05-02|Affibody AB|Polypeptides|
DK2621515T3|2010-09-28|2017-07-17|Aegerion Pharmaceuticals Inc|Chimeric seal-human leptin polypeptide with increased solubility|
DK2729160T3|2011-07-08|2019-07-01|Aegerion Pharmaceuticals Inc|MANIPULATED POLYPEPTIDES WHICH HAVE IMPROVED EFFECT TIME AND REDUCED IMMUNOGENICITY|
EP2844269A4|2012-03-28|2016-01-06|Amylin Pharmaceuticals Llc|Transmucosal delivery of engineered polypeptides|
KR102123457B1|2012-09-25|2020-06-16|애피바디 에이비|Albumin binding polypeptide|
US9943568B2|2013-04-18|2018-04-17|Armo Biosciences, Inc.|Methods of using pegylated interleukin-10 for treating cancer|
EP3434277A1|2013-06-17|2019-01-30|Armo Biosciences, Inc.|Method for assessing protein identity and stability|
AU2014311432A1|2013-08-30|2016-03-03|Armo Biosciences, Inc.|Methods of using interleukin-10 for treating diseases and disorders|
EP3083675B1|2013-12-20|2018-03-07|Affibody AB|Engineered albumin binding polypeptide|
CN106573072A|2014-06-02|2017-04-19|阿尔莫生物科技股份有限公司|Methods of lowering serum cholesterol|
ES2887370T3|2014-09-04|2021-12-22|Novo Nordisk As|New amylin and calcitonin receptor agonist|
EP3206713A4|2014-10-14|2018-06-27|Armo Biosciences, Inc.|Interleukin-15 compositions and uses thereof|
CA2963995A1|2014-10-22|2016-04-28|Armo Biosciences, Inc.|Methods of using interleukin-10 for treating diseases and disorders|
WO2016126615A1|2015-02-03|2016-08-11|Armo Biosciences, Inc.|Methods of using interleukin-10 for treating diseases and disorders|
CN107847583A|2015-05-28|2018-03-27|阿尔莫生物科技股份有限公司|PEGylated Interleukin 10 for treating cancer|
EP3333177B1|2015-08-05|2021-07-14|Shaanxi Micot Technology Limited Company|Multi-target compound with anticoagulation and antiplatelet activity, preparation method therefor, and use thereof|
KR20180045001A|2015-09-22|2018-05-03|더 리젠츠 오브 더 유니버시티 오브 캘리포니아|Modified cytotoxins and their therapeutic uses|
US10286079B2|2015-09-22|2019-05-14|The Regents Of The University Of California|Modified cytotoxins and their therapeutic use|
CA3000275A1|2015-10-02|2017-04-06|University Of Copenhagen|Small molecules blocking histone reader domains|
CN110139663A|2016-09-12|2019-08-16|艾珠雷昂医药有限公司|The method for detecting anti-leptin neutralizing antibody|
法律状态:
2020-10-20| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]|
2021-03-30| B07D| Technical examination (opinion) related to article 229 of industrial property law [chapter 7.4 patent gazette]|Free format text: DE ACORDO COM O ARTIGO 229-C DA LEI NO 10196/2001, QUE MODIFICOU A LEI NO 9279/96, A CONCESSAO DA PATENTE ESTA CONDICIONADA A ANUENCIA PREVIA DA ANVISA. CONSIDERANDO A APROVACAO DOS TERMOS DO PARECER NO 337/PGF/EA/2010, BEM COMO A PORTARIA INTERMINISTERIAL NO 1065 DE 24/05/2012, ENCAMINHA-SE O PRESENTE PEDIDO PARA AS PROVIDENCIAS CABIVEIS. |
2021-04-20| B07E| Notification of approval relating to section 229 industrial property law [chapter 7.5 patent gazette]|
2021-05-11| B06U| Preliminary requirement: requests with searches performed by other patent offices: procedure suspended [chapter 6.21 patent gazette]|
2021-11-16| B09A| Decision: intention to grant [chapter 9.1 patent gazette]|
2021-11-23| B350| Update of information on the portal [chapter 15.35 patent gazette]|
2022-01-04| B16A| Patent or certificate of addition of invention granted [chapter 16.1 patent gazette]|Free format text: PRAZO DE VALIDADE: 20 (VINTE) ANOS CONTADOS A PARTIR DE 28/09/2011, OBSERVADAS AS CONDICOES LEGAIS. PATENTE CONCEDIDA CONFORME ADI 5.529/DF, QUE DETERMINA A ALTERACAO DO PRAZO DE CONCESSAO. |
优先权:
申请号 | 申请日 | 专利标题
US38740210P| true| 2010-09-28|2010-09-28|
US61/387,402|2010-09-28|
US42209110P| true| 2010-12-10|2010-12-10|
US61/422,091|2010-12-10|
PCT/US2011/053786|WO2012050930A2|2010-09-28|2011-09-28|Engineered polypeptides having enhanced duration of action|
[返回顶部]