CA2704446A1 - Melanocortin receptor binding mimetibodies, compositions, methods and uses - Google Patents
Melanocortin receptor binding mimetibodies, compositions, methods and uses Download PDFInfo
- Publication number
- CA2704446A1 CA2704446A1 CA2704446A CA2704446A CA2704446A1 CA 2704446 A1 CA2704446 A1 CA 2704446A1 CA 2704446 A CA2704446 A CA 2704446A CA 2704446 A CA2704446 A CA 2704446A CA 2704446 A1 CA2704446 A1 CA 2704446A1
- Authority
- CA
- Canada
- Prior art keywords
- seq
- polypeptide
- cell
- melanocortin receptor
- alpha
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Abandoned
Links
- 102000004378 Melanocortin Receptors Human genes 0.000 title claims abstract description 63
- 108090000950 Melanocortin Receptors Proteins 0.000 title claims abstract description 63
- 230000027455 binding Effects 0.000 title claims abstract description 53
- 238000000034 method Methods 0.000 title claims abstract description 27
- 239000000203 mixture Substances 0.000 title description 9
- 108090000765 processed proteins & peptides Proteins 0.000 claims abstract description 118
- 102000004196 processed proteins & peptides Human genes 0.000 claims abstract description 106
- 229920001184 polypeptide Polymers 0.000 claims abstract description 101
- 239000002157 polynucleotide Substances 0.000 claims abstract description 29
- 108091033319 polynucleotide Proteins 0.000 claims abstract description 29
- 102000040430 polynucleotide Human genes 0.000 claims abstract description 29
- 108010021436 Type 4 Melanocortin Receptor Proteins 0.000 claims description 39
- 102000008316 Type 4 Melanocortin Receptor Human genes 0.000 claims description 39
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 20
- 239000013598 vector Substances 0.000 claims description 16
- 208000008589 Obesity Diseases 0.000 claims description 15
- 235000020824 obesity Nutrition 0.000 claims description 15
- 230000001404 mediated effect Effects 0.000 claims description 11
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 claims description 8
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 claims description 8
- 210000000056 organ Anatomy 0.000 claims description 8
- 239000008194 pharmaceutical composition Substances 0.000 claims description 8
- 239000012634 fragment Substances 0.000 claims description 7
- 206010057671 Female sexual dysfunction Diseases 0.000 claims description 6
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 claims description 6
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 claims description 6
- 230000000295 complement effect Effects 0.000 claims description 6
- 208000010228 Erectile Dysfunction Diseases 0.000 claims description 5
- 201000001881 impotence Diseases 0.000 claims description 5
- 108060003951 Immunoglobulin Proteins 0.000 claims description 4
- 239000003085 diluting agent Substances 0.000 claims description 4
- 102000018358 immunoglobulin Human genes 0.000 claims description 4
- 239000000126 substance Substances 0.000 claims description 4
- 201000005569 Gout Diseases 0.000 claims description 3
- 206010018634 Gouty Arthritis Diseases 0.000 claims description 3
- 230000004071 biological effect Effects 0.000 claims description 3
- 206010039073 rheumatoid arthritis Diseases 0.000 claims description 3
- 208000022559 Inflammatory bowel disease Diseases 0.000 claims description 2
- 230000009285 allergic inflammation Effects 0.000 claims description 2
- 230000006931 brain damage Effects 0.000 claims description 2
- 231100000874 brain damage Toxicity 0.000 claims description 2
- 208000029028 brain injury Diseases 0.000 claims description 2
- 238000012258 culturing Methods 0.000 claims description 2
- 239000003937 drug carrier Substances 0.000 claims description 2
- 230000004761 fibrosis Effects 0.000 claims description 2
- 208000006454 hepatitis Diseases 0.000 claims description 2
- 208000028867 ischemia Diseases 0.000 claims description 2
- 230000000302 ischemic effect Effects 0.000 claims description 2
- 208000018191 liver inflammation Diseases 0.000 claims description 2
- 208000027232 peripheral nervous system disease Diseases 0.000 claims description 2
- 208000033808 peripheral neuropathy Diseases 0.000 claims description 2
- 230000004968 inflammatory condition Effects 0.000 claims 2
- 241000699802 Cricetulus griseus Species 0.000 claims 1
- 206010016654 Fibrosis Diseases 0.000 claims 1
- 206010061481 Renal injury Diseases 0.000 claims 1
- 210000001672 ovary Anatomy 0.000 claims 1
- MWUXSHHQAYIFBG-UHFFFAOYSA-N Nitric oxide Chemical compound O=[N] MWUXSHHQAYIFBG-UHFFFAOYSA-N 0.000 description 180
- WHNFPRLDDSXQCL-UAZQEYIDSA-N α-msh Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(N)=O)NC(=O)[C@H](CO)NC(C)=O)C1=CC=C(O)C=C1 WHNFPRLDDSXQCL-UAZQEYIDSA-N 0.000 description 66
- 101800001751 Melanocyte-stimulating hormone alpha Proteins 0.000 description 59
- 102100027467 Pro-opiomelanocortin Human genes 0.000 description 56
- 210000004027 cell Anatomy 0.000 description 45
- 238000006467 substitution reaction Methods 0.000 description 21
- 230000000694 effects Effects 0.000 description 16
- 102400000740 Melanocyte-stimulating hormone alpha Human genes 0.000 description 15
- 101710200814 Melanotropin alpha Proteins 0.000 description 15
- 235000001014 amino acid Nutrition 0.000 description 15
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 15
- 150000001413 amino acids Chemical class 0.000 description 14
- 238000006206 glycosylation reaction Methods 0.000 description 13
- 230000004048 modification Effects 0.000 description 13
- 238000012986 modification Methods 0.000 description 13
- 108090000623 proteins and genes Proteins 0.000 description 12
- 241001465754 Metazoa Species 0.000 description 11
- 102100026120 IgG receptor FcRn large subunit p51 Human genes 0.000 description 10
- 101710177940 IgG receptor FcRn large subunit p51 Proteins 0.000 description 10
- 239000013604 expression vector Substances 0.000 description 10
- 235000018102 proteins Nutrition 0.000 description 10
- 102000004169 proteins and genes Human genes 0.000 description 10
- JDKLPDJLXHXHNV-MFVUMRCOSA-N (3s,6s,9r,12s,15s,23s)-15-[[(2s)-2-acetamidohexanoyl]amino]-9-benzyl-6-[3-(diaminomethylideneamino)propyl]-12-(1h-imidazol-5-ylmethyl)-3-(1h-indol-3-ylmethyl)-2,5,8,11,14,17-hexaoxo-1,4,7,10,13,18-hexazacyclotricosane-23-carboxamide Chemical compound C([C@@H]1C(=O)N[C@@H](CCCN=C(N)N)C(=O)N[C@@H](CC=2C3=CC=CC=C3NC=2)C(=O)N[C@@H](CCCCNC(=O)C[C@@H](C(N[C@@H](CC=2NC=NC=2)C(=O)N1)=O)NC(=O)[C@@H](NC(C)=O)CCCC)C(N)=O)C1=CC=CC=C1 JDKLPDJLXHXHNV-MFVUMRCOSA-N 0.000 description 9
- 230000004540 complement-dependent cytotoxicity Effects 0.000 description 9
- 241000282414 Homo sapiens Species 0.000 description 8
- 108010076504 Protein Sorting Signals Proteins 0.000 description 8
- 230000001965 increasing effect Effects 0.000 description 8
- 102000005962 receptors Human genes 0.000 description 8
- 108020003175 receptors Proteins 0.000 description 8
- 230000003248 secreting effect Effects 0.000 description 8
- 238000011282 treatment Methods 0.000 description 8
- 125000000539 amino acid group Chemical group 0.000 description 7
- 230000037396 body weight Effects 0.000 description 7
- 230000007423 decrease Effects 0.000 description 7
- 230000037406 food intake Effects 0.000 description 7
- 235000012631 food intake Nutrition 0.000 description 7
- 230000000638 stimulation Effects 0.000 description 7
- 108010008364 Melanocortins Proteins 0.000 description 6
- 230000015572 biosynthetic process Effects 0.000 description 6
- 210000004556 brain Anatomy 0.000 description 6
- 230000013595 glycosylation Effects 0.000 description 6
- 238000002347 injection Methods 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 239000002865 melanocortin Substances 0.000 description 6
- 150000007523 nucleic acids Chemical class 0.000 description 6
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 6
- 210000002966 serum Anatomy 0.000 description 6
- 210000001519 tissue Anatomy 0.000 description 6
- 230000004580 weight loss Effects 0.000 description 6
- 108010087819 Fc receptors Proteins 0.000 description 5
- 102000009109 Fc receptors Human genes 0.000 description 5
- 102100023726 Melanocortin receptor 3 Human genes 0.000 description 5
- 241000699670 Mus sp. Species 0.000 description 5
- 230000004988 N-glycosylation Effects 0.000 description 5
- 108010021433 Type 3 Melanocortin Receptor Proteins 0.000 description 5
- 230000004913 activation Effects 0.000 description 5
- 238000003556 assay Methods 0.000 description 5
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 5
- 230000002829 reductive effect Effects 0.000 description 5
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 4
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 4
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 4
- 108010069820 Pro-Opiomelanocortin Proteins 0.000 description 4
- 239000000683 Pro-Opiomelanocortin Substances 0.000 description 4
- 241000700159 Rattus Species 0.000 description 4
- 241000283984 Rodentia Species 0.000 description 4
- 239000000556 agonist Substances 0.000 description 4
- 201000010099 disease Diseases 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 230000014509 gene expression Effects 0.000 description 4
- 238000004519 manufacturing process Methods 0.000 description 4
- 239000013641 positive control Substances 0.000 description 4
- 208000001072 type 2 diabetes mellitus Diseases 0.000 description 4
- CUKWUWBLQQDQAC-VEQWQPCFSA-N (3s)-3-amino-4-[[(2s)-1-[[(2s)-1-[[(2s)-1-[[(2s,3s)-1-[[(2s)-1-[(2s)-2-[[(1s)-1-carboxyethyl]carbamoyl]pyrrolidin-1-yl]-3-(1h-imidazol-5-yl)-1-oxopropan-2-yl]amino]-3-methyl-1-oxopentan-2-yl]amino]-3-(4-hydroxyphenyl)-1-oxopropan-2-yl]amino]-3-methyl-1-ox Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C)C(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@@H](N)CC(O)=O)C(C)C)C1=CC=C(O)C=C1 CUKWUWBLQQDQAC-VEQWQPCFSA-N 0.000 description 3
- 102400000345 Angiotensin-2 Human genes 0.000 description 3
- 101800000733 Angiotensin-2 Proteins 0.000 description 3
- 102000004127 Cytokines Human genes 0.000 description 3
- 108090000695 Cytokines Proteins 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 206010061218 Inflammation Diseases 0.000 description 3
- 108090001090 Lectins Proteins 0.000 description 3
- 102000004856 Lectins Human genes 0.000 description 3
- 102000030612 Melanocortin 5 receptor Human genes 0.000 description 3
- 108010088565 Melanocortin 5 receptor Proteins 0.000 description 3
- 208000001145 Metabolic Syndrome Diseases 0.000 description 3
- 201000000690 abdominal obesity-metabolic syndrome Diseases 0.000 description 3
- 230000001154 acute effect Effects 0.000 description 3
- UAHFGYDRQSXQEB-LEBBXHLNSA-N afamelanotide Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(N)=O)NC(=O)[C@H](CO)NC(C)=O)C1=CC=C(O)C=C1 UAHFGYDRQSXQEB-LEBBXHLNSA-N 0.000 description 3
- 229950006323 angiotensin ii Drugs 0.000 description 3
- 230000003110 anti-inflammatory effect Effects 0.000 description 3
- 239000000427 antigen Substances 0.000 description 3
- 108091007433 antigens Proteins 0.000 description 3
- 102000036639 antigens Human genes 0.000 description 3
- 238000004587 chromatography analysis Methods 0.000 description 3
- 239000002299 complementary DNA Substances 0.000 description 3
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 3
- 230000001461 cytolytic effect Effects 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 238000012217 deletion Methods 0.000 description 3
- 230000037430 deletion Effects 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 3
- 238000010348 incorporation Methods 0.000 description 3
- 230000004054 inflammatory process Effects 0.000 description 3
- 230000005764 inhibitory process Effects 0.000 description 3
- 238000003780 insertion Methods 0.000 description 3
- 230000037431 insertion Effects 0.000 description 3
- 239000002523 lectin Substances 0.000 description 3
- 239000000178 monomer Substances 0.000 description 3
- 210000000440 neutrophil Anatomy 0.000 description 3
- 230000002093 peripheral effect Effects 0.000 description 3
- 239000002953 phosphate buffered saline Substances 0.000 description 3
- 238000000159 protein binding assay Methods 0.000 description 3
- 238000002821 scintillation proximity assay Methods 0.000 description 3
- 230000001568 sexual effect Effects 0.000 description 3
- 238000001356 surgical procedure Methods 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 2
- 108700034262 4-Nle-7-Phe-alpha- MSH Proteins 0.000 description 2
- 201000006474 Brain Ischemia Diseases 0.000 description 2
- 206010007559 Cardiac failure congestive Diseases 0.000 description 2
- 102000019034 Chemokines Human genes 0.000 description 2
- 108010012236 Chemokines Proteins 0.000 description 2
- 239000000055 Corticotropin-Releasing Hormone Substances 0.000 description 2
- -1 D-amino acids Chemical class 0.000 description 2
- 101100257999 Danio rerio stambpa gene Proteins 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 208000021663 Female sexual arousal disease Diseases 0.000 description 2
- 102000003688 G-Protein-Coupled Receptors Human genes 0.000 description 2
- 108090000045 G-Protein-Coupled Receptors Proteins 0.000 description 2
- 239000004471 Glycine Substances 0.000 description 2
- 206010019280 Heart failures Diseases 0.000 description 2
- 206010022489 Insulin Resistance Diseases 0.000 description 2
- 102000003814 Interleukin-10 Human genes 0.000 description 2
- 108090000174 Interleukin-10 Proteins 0.000 description 2
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 2
- 108700026244 Open Reading Frames Proteins 0.000 description 2
- 241000700157 Rattus norvegicus Species 0.000 description 2
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 2
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 2
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 239000012190 activator Substances 0.000 description 2
- 230000002776 aggregation Effects 0.000 description 2
- 238000004220 aggregation Methods 0.000 description 2
- 235000004279 alanine Nutrition 0.000 description 2
- 206010003246 arthritis Diseases 0.000 description 2
- 239000011324 bead Substances 0.000 description 2
- FFHBJDQSGDNCIV-MFVUMRCOSA-N bremelanotide Chemical compound C([C@@H]1C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=2C3=CC=CC=C3NC=2)C(=O)N[C@@H](CCCCNC(=O)C[C@@H](C(N[C@@H](CC=2NC=NC=2)C(=O)N1)=O)NC(=O)[C@@H](NC(C)=O)CCCC)C(O)=O)C1=CC=CC=C1 FFHBJDQSGDNCIV-MFVUMRCOSA-N 0.000 description 2
- 238000013262 cAMP assay Methods 0.000 description 2
- 210000003850 cellular structure Anatomy 0.000 description 2
- 208000019425 cirrhosis of liver Diseases 0.000 description 2
- 206010009887 colitis Diseases 0.000 description 2
- 238000012875 competitive assay Methods 0.000 description 2
- 230000024203 complement activation Effects 0.000 description 2
- IDLFZVILOHSSID-OVLDLUHVSA-N corticotropin Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(N)=O)C(=O)NCC(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(O)=O)NC(=O)[C@@H](N)CO)C1=CC=C(O)C=C1 IDLFZVILOHSSID-OVLDLUHVSA-N 0.000 description 2
- 229960000258 corticotropin Drugs 0.000 description 2
- 235000018417 cysteine Nutrition 0.000 description 2
- 230000009089 cytolysis Effects 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 230000001472 cytotoxic effect Effects 0.000 description 2
- 206010012601 diabetes mellitus Diseases 0.000 description 2
- 238000006471 dimerization reaction Methods 0.000 description 2
- 230000006334 disulfide bridging Effects 0.000 description 2
- 239000012636 effector Substances 0.000 description 2
- 230000002708 enhancing effect Effects 0.000 description 2
- 230000001856 erectile effect Effects 0.000 description 2
- 210000003527 eukaryotic cell Anatomy 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 235000013305 food Nutrition 0.000 description 2
- 230000002440 hepatic effect Effects 0.000 description 2
- 230000005847 immunogenicity Effects 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 238000000185 intracerebroventricular administration Methods 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 108091005601 modified peptides Proteins 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 230000000144 pharmacologic effect Effects 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 230000000770 proinflammatory effect Effects 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 101150076714 stambp gene Proteins 0.000 description 2
- 210000002784 stomach Anatomy 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 238000013519 translation Methods 0.000 description 2
- 238000001262 western blot Methods 0.000 description 2
- JARGNLJYKBUKSJ-KGZKBUQUSA-N (2r)-2-amino-5-[[(2r)-1-(carboxymethylamino)-3-hydroxy-1-oxopropan-2-yl]amino]-5-oxopentanoic acid;hydrobromide Chemical compound Br.OC(=O)[C@H](N)CCC(=O)N[C@H](CO)C(=O)NCC(O)=O JARGNLJYKBUKSJ-KGZKBUQUSA-N 0.000 description 1
- AOFUBOWZWQFQJU-SNOJBQEQSA-N (2r,3s,4s,5r)-2,5-bis(hydroxymethyl)oxolane-2,3,4-triol;(2s,3r,4s,5s,6r)-6-(hydroxymethyl)oxane-2,3,4,5-tetrol Chemical compound OC[C@H]1O[C@](O)(CO)[C@@H](O)[C@@H]1O.OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@@H]1O AOFUBOWZWQFQJU-SNOJBQEQSA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- 108010013043 Acetylesterase Proteins 0.000 description 1
- 206010002652 Anorgasmia Diseases 0.000 description 1
- 208000031104 Arterial Occlusive disease Diseases 0.000 description 1
- 101100263837 Bovine ephemeral fever virus (strain BB7721) beta gene Proteins 0.000 description 1
- 125000001433 C-terminal amino-acid group Chemical group 0.000 description 1
- 206010008120 Cerebral ischaemia Diseases 0.000 description 1
- 208000017667 Chronic Disease Diseases 0.000 description 1
- 101100007328 Cocos nucifera COS-1 gene Proteins 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 238000011537 Coomassie blue staining Methods 0.000 description 1
- IVOMOUWHDPKRLL-KQYNXXCUSA-N Cyclic adenosine monophosphate Chemical compound C([C@H]1O2)OP(O)(=O)O[C@H]1[C@@H](O)[C@@H]2N1C(N=CN=C2N)=C2N=C1 IVOMOUWHDPKRLL-KQYNXXCUSA-N 0.000 description 1
- 150000008574 D-amino acids Chemical class 0.000 description 1
- 230000005778 DNA damage Effects 0.000 description 1
- 231100000277 DNA damage Toxicity 0.000 description 1
- 201000004624 Dermatitis Diseases 0.000 description 1
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 1
- 208000032928 Dyslipidaemia Diseases 0.000 description 1
- 208000004483 Dyspareunia Diseases 0.000 description 1
- 238000012286 ELISA Assay Methods 0.000 description 1
- 101100316840 Enterobacteria phage P4 Beta gene Proteins 0.000 description 1
- 108010053070 Glutathione Disulfide Proteins 0.000 description 1
- 101000978418 Homo sapiens Melanocortin receptor 4 Proteins 0.000 description 1
- 206010020710 Hyperphagia Diseases 0.000 description 1
- 206010062767 Hypophysitis Diseases 0.000 description 1
- 108700005091 Immunoglobulin Genes Proteins 0.000 description 1
- 102000004195 Isomerases Human genes 0.000 description 1
- 108090000769 Isomerases Proteins 0.000 description 1
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 206010024419 Libido decreased Diseases 0.000 description 1
- 208000017170 Lipid metabolism disease Diseases 0.000 description 1
- 239000012097 Lipofectamine 2000 Substances 0.000 description 1
- 208000019693 Lung disease Diseases 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 101150004219 MCR1 gene Proteins 0.000 description 1
- 101710201349 Metallothionein B Proteins 0.000 description 1
- 101710094505 Metallothionein-2 Proteins 0.000 description 1
- 102100031347 Metallothionein-2 Human genes 0.000 description 1
- 208000028389 Nerve injury Diseases 0.000 description 1
- 206010033307 Overweight Diseases 0.000 description 1
- 208000002193 Pain Diseases 0.000 description 1
- 229940123333 Phosphodiesterase 5 inhibitor Drugs 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- FELJDCNGZFDUNR-WDSKDSINSA-N Pro-Ala Chemical group OC(=O)[C@H](C)NC(=O)[C@@H]1CCCN1 FELJDCNGZFDUNR-WDSKDSINSA-N 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 208000006262 Psychological Sexual Dysfunctions Diseases 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 206010063897 Renal ischaemia Diseases 0.000 description 1
- 101100206347 Schizosaccharomyces pombe (strain 972 / ATCC 24843) pmh1 gene Proteins 0.000 description 1
- 238000012300 Sequence Analysis Methods 0.000 description 1
- BNRNXUUZRGQAQC-UHFFFAOYSA-N Sildenafil Natural products CCCC1=NN(C)C(C(N2)=O)=C1N=C2C(C(=CC=1)OCC)=CC=1S(=O)(=O)N1CCN(C)CC1 BNRNXUUZRGQAQC-UHFFFAOYSA-N 0.000 description 1
- 208000006011 Stroke Diseases 0.000 description 1
- BMQYVXCPAOLZOK-UHFFFAOYSA-N Trihydroxypropylpterisin Natural products OCC(O)C(O)C1=CN=C2NC(N)=NC(=O)C2=N1 BMQYVXCPAOLZOK-UHFFFAOYSA-N 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- IVOMOUWHDPKRLL-UHFFFAOYSA-N UNPD107823 Natural products O1C2COP(O)(=O)OC2C(O)C1N1C(N=CN=C2N)=C2N=C1 IVOMOUWHDPKRLL-UHFFFAOYSA-N 0.000 description 1
- 208000012931 Urologic disease Diseases 0.000 description 1
- SECKRCOLJRRGGV-UHFFFAOYSA-N Vardenafil Chemical compound CCCC1=NC(C)=C(C(N=2)=O)N1NC=2C(C(=CC=1)OCC)=CC=1S(=O)(=O)N1CCN(CC)CC1 SECKRCOLJRRGGV-UHFFFAOYSA-N 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 108700034688 acetyl-norleucyl(4)-(aspartyl(5)-histidyl(6)-phenylalanyl(7)-arginyl(8)-tryptophyl(9)-lysyl(10))cyclo-alpha-MSH(4-10)amide Proteins 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 208000026816 acute arthritis Diseases 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 238000001042 affinity chromatography Methods 0.000 description 1
- 239000003253 alpha intermedin derivative Substances 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- BFNBIHQBYMNNAN-UHFFFAOYSA-N ammonium sulfate Chemical compound N.N.OS(O)(=O)=O BFNBIHQBYMNNAN-UHFFFAOYSA-N 0.000 description 1
- 229910052921 ammonium sulfate Inorganic materials 0.000 description 1
- 238000012870 ammonium sulfate precipitation Methods 0.000 description 1
- 235000011130 ammonium sulphate Nutrition 0.000 description 1
- 238000005571 anion exchange chromatography Methods 0.000 description 1
- 150000001450 anions Chemical class 0.000 description 1
- 230000001640 apoptogenic effect Effects 0.000 description 1
- PYMYPHUHKUWMLA-WDCZJNDASA-N arabinose Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)C=O PYMYPHUHKUWMLA-WDCZJNDASA-N 0.000 description 1
- PYMYPHUHKUWMLA-UHFFFAOYSA-N arabinose Natural products OCC(O)C(O)C(O)C=O PYMYPHUHKUWMLA-UHFFFAOYSA-N 0.000 description 1
- 208000021328 arterial occlusion Diseases 0.000 description 1
- 230000000923 atherogenic effect Effects 0.000 description 1
- 230000003376 axonal effect Effects 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 230000003542 behavioural effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 102000016966 beta-2 Adrenergic Receptors Human genes 0.000 description 1
- 108010014499 beta-2 Adrenergic Receptors Proteins 0.000 description 1
- SRBFZHDQGSBBOR-UHFFFAOYSA-N beta-D-Pyranose-Lyxose Natural products OC1COC(O)C(O)C1O SRBFZHDQGSBBOR-UHFFFAOYSA-N 0.000 description 1
- 230000002146 bilateral effect Effects 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 230000036772 blood pressure Effects 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 238000006664 bond formation reaction Methods 0.000 description 1
- 210000004958 brain cell Anatomy 0.000 description 1
- 210000005013 brain tissue Anatomy 0.000 description 1
- 108010072543 bremelanotide Proteins 0.000 description 1
- 229950000740 bremelanotide Drugs 0.000 description 1
- 230000003491 cAMP production Effects 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 238000005277 cation exchange chromatography Methods 0.000 description 1
- 230000007910 cell fusion Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 208000015114 central nervous system disease Diseases 0.000 description 1
- 206010008118 cerebral infarction Diseases 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 229940117229 cialis Drugs 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 239000000084 colloidal system Substances 0.000 description 1
- 230000000112 colonic effect Effects 0.000 description 1
- 230000004154 complement system Effects 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 208000010247 contact dermatitis Diseases 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000000875 corresponding effect Effects 0.000 description 1
- 229940095074 cyclic amp Drugs 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 150000001945 cysteines Chemical class 0.000 description 1
- 108010004073 cysteinylcysteine Proteins 0.000 description 1
- 239000000430 cytokine receptor antagonist Substances 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 229960000633 dextran sulfate Drugs 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- XEYBRNLFEZDVAW-ARSRFYASSA-N dinoprostone Chemical compound CCCCC[C@H](O)\C=C\[C@H]1[C@H](O)CC(=O)[C@@H]1C\C=C/CCCC(O)=O XEYBRNLFEZDVAW-ARSRFYASSA-N 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 230000009429 distress Effects 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 210000002889 endothelial cell Anatomy 0.000 description 1
- 238000012869 ethanol precipitation Methods 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 230000002550 fecal effect Effects 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 108010044804 gamma-glutamyl-seryl-glycine Proteins 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- 210000004392 genitalia Anatomy 0.000 description 1
- YPZRWBKMTBYPTK-BJDJZHNGSA-N glutathione disulfide Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@H](C(=O)NCC(O)=O)CSSC[C@@H](C(=O)NCC(O)=O)NC(=O)CC[C@H](N)C(O)=O YPZRWBKMTBYPTK-BJDJZHNGSA-N 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 238000004191 hydrophobic interaction chromatography Methods 0.000 description 1
- 208000017020 hypoactive sexual desire disease Diseases 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 229940076144 interleukin-10 Drugs 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 239000002085 irritant Substances 0.000 description 1
- 231100000021 irritant Toxicity 0.000 description 1
- 238000012004 kinetic exclusion assay Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 239000010410 layer Substances 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 229940097443 levitra Drugs 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 230000004777 loss-of-function mutation Effects 0.000 description 1
- 230000002934 lysing effect Effects 0.000 description 1
- 210000003712 lysosome Anatomy 0.000 description 1
- 230000001868 lysosomic effect Effects 0.000 description 1
- 229910052943 magnesium sulfate Inorganic materials 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 230000004630 mental health Effects 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 238000001823 molecular biology technique Methods 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 238000002887 multiple sequence alignment Methods 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 108010068617 neonatal Fc receptor Proteins 0.000 description 1
- BMQYVXCPAOLZOK-XINAWCOVSA-N neopterin Chemical compound OC[C@@H](O)[C@@H](O)C1=CN=C2NC(N)=NC(=O)C2=N1 BMQYVXCPAOLZOK-XINAWCOVSA-N 0.000 description 1
- 230000008764 nerve damage Effects 0.000 description 1
- 210000000118 neural pathway Anatomy 0.000 description 1
- 230000010004 neural pathway Effects 0.000 description 1
- 210000002241 neurite Anatomy 0.000 description 1
- 230000000324 neuroprotective effect Effects 0.000 description 1
- 125000004433 nitrogen atom Chemical group N* 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 201000008482 osteoarthritis Diseases 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 125000004430 oxygen atom Chemical group O* 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 239000000813 peptide hormone Substances 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 229940080469 phosphocellulose Drugs 0.000 description 1
- 239000002590 phosphodiesterase V inhibitor Substances 0.000 description 1
- 230000008884 pinocytosis Effects 0.000 description 1
- 210000003635 pituitary gland Anatomy 0.000 description 1
- 239000000902 placebo Substances 0.000 description 1
- 229940068196 placebo Drugs 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- OIGNJSKKLXVSLS-VWUMJDOOSA-N prednisolone Chemical compound O=C1C=C[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 OIGNJSKKLXVSLS-VWUMJDOOSA-N 0.000 description 1
- 229960005205 prednisolone Drugs 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000000436 pro-erectile effect Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000009465 prokaryotic expression Effects 0.000 description 1
- 108010017378 prolyl aminopeptidase Proteins 0.000 description 1
- 239000012460 protein solution Substances 0.000 description 1
- 230000002797 proteolythic effect Effects 0.000 description 1
- 238000011552 rat model Methods 0.000 description 1
- 239000000018 receptor agonist Substances 0.000 description 1
- 229940044601 receptor agonist Drugs 0.000 description 1
- 230000000306 recurrent effect Effects 0.000 description 1
- 238000004064 recycling Methods 0.000 description 1
- 230000010410 reperfusion Effects 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 210000001044 sensory neuron Anatomy 0.000 description 1
- 239000004017 serum-free culture medium Substances 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- DEIYFTQMQPDXOT-UHFFFAOYSA-N sildenafil citrate Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O.CCCC1=NN(C)C(C(N2)=O)=C1N=C2C(C(=CC=1)OCC)=CC=1S(=O)(=O)N1CCN(C)CC1 DEIYFTQMQPDXOT-UHFFFAOYSA-N 0.000 description 1
- 229910052709 silver Inorganic materials 0.000 description 1
- 239000004332 silver Substances 0.000 description 1
- 239000002356 single layer Substances 0.000 description 1
- 238000001542 size-exclusion chromatography Methods 0.000 description 1
- 208000017520 skin disease Diseases 0.000 description 1
- 201000002859 sleep apnea Diseases 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 230000000392 somatic effect Effects 0.000 description 1
- 210000000278 spinal cord Anatomy 0.000 description 1
- 210000005250 spinal neuron Anatomy 0.000 description 1
- 238000012453 sprague-dawley rat model Methods 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 230000035882 stress Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- WOXKDUGGOYFFRN-IIBYNOLFSA-N tadalafil Chemical compound C1=C2OCOC2=CC([C@@H]2C3=C(C4=CC=CC=C4N3)C[C@H]3N2C(=O)CN(C3=O)C)=C1 WOXKDUGGOYFFRN-IIBYNOLFSA-N 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 201000010875 transient cerebral ischemia Diseases 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 125000000430 tryptophan group Chemical group [H]N([H])C(C(=O)O*)C([H])([H])C1=C([H])N([H])C2=C([H])C([H])=C([H])C([H])=C12 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 208000014001 urinary system disease Diseases 0.000 description 1
- 229940094720 viagra Drugs 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 238000005303 weighing Methods 0.000 description 1
- 230000004584 weight gain Effects 0.000 description 1
- 235000019786 weight gain Nutrition 0.000 description 1
Landscapes
- Peptides Or Proteins (AREA)
Abstract
Melanocortin receptor binding mimetibody polypeptides are disclosed. Polynucleotides encoding these polypeptides, cells comprising these polynucleotidee or expressing the mimetibodies, and methods of making and using the forgoing are also disclosed.
Description
MELANOCORTIN RECEPTOR BINDING MIMETIBODIES, COMPOSITIONS, METHODS AND USES
Cross-Reference to Related Applications This application is a continuation-in-part of United States Application Serial No. 11/257,851, filed 25 October 2005, which claims priority to United States Provisional Application Serial No.
60/621,960, filed 25 October 2004. This application also claims the benefit of United States Provisional Application Serial No.
60/972,018, filed 13 September 2007. The entire contents of each of the aforementioned applications is incorporated herein by reference in their entirety.
Field of the Invention The present invention relates to melanocortin receptor binding mimetibodies, polynucleotides encoding the mimetibodies, cells comprising the polynucleotides or expressing the mimetibodies, and methods of making and using the foregoing.
Background of the Invention Obesity is a chronic disease manifested by an excess of fat mass in proportion to body size. Today, every third American is considered overweight (Body Mass Index (BMI) >25 kg/m2), thus prompting the United States Centers for Disease Control and Prevention (CDC) to declare that obesity is reaching epidemic proportions (Cummings and Schwartz, Annu. Rev. Med. 54:453-471((2003)). The importance of treating obesity is emphasized by the fact that this disease is either the underlying cause, or a risk factor, for developing diseases such as Type 2 Diabetes, congestive heart failure, osteoarthritis and sleep apnea among others.
Additionally, obesity is linked to "Metabolic Syndrome" which is a medical condition characterized by obesity, atherogenic dyslipidemia, elevated blood pressure and insulin resistance.
Metabolic Syndrome affects an increasing number of people in the United States. Importantly, it has been shown that even a modest decrease in body weight (5-10% of initial body weight) may significantly improve Metabolic Syndrome conditions and decrease the risk factors for developing obesity-associated disease (Wing et al., Arch. Intern. Med. 147:1749-1753 (1987); Tuomilehto et al., New Engl. J. Med. 344:1343-1350 (2001); Knowler et al., New Engl. J Med.
346:393-403 (2002); Franz et al., Diabetes Care 25:148-198 (2002)).
Additionally, treatment of obesity may be important from a mental health perspective due to the social stigma often attached to obese individuals in some cultures.
Melanocortin receptors play a major role in the regulation of overall energy balance and obesity in both humans and rodents.
Alpha-melanocyte stimulating hormone (alpha-MSH) is a 13 amino acid peptide hormone that is an important component of the melanocortin system. Alpha-MSH is produced by the proteolytic processing of proopiomelanocortin (POMC) released by the pituitary gland. Alpha-MSH binds with high affinity to the melanocortin 4 receptor (MC4R), but also binds melanocortin receptor 3 (MC3R) and melanocortin receptor 5 (MC5R) with lower affinity. MC4R is a G-coupled protein receptor found in the brain which, when stimulated by alpha-MSH
binding, causes decreased food intake and increased fat oxidation.
Ultimately, stimulation of melanocortin receptors such as MC4R
results in weight loss.
In humans and rodents, loss of function mutations in the different components of the melanocortin system are closely correlated with obesity and related conditions. In mice, mutations within POMC, or MC4R and MC3R produce obesity, insulin resistance and hyperphagia (Goodfellow and Saunders, Curr. Topics Med. Chem. 3:
855-883 (2003); Huszar et al., Cell 88:131-141 (1997); Yaswen et al., Nat. Med. 5: 1066-1070 (1999)). In man, mutations within POMC
or MC4R lead to the development of obesity associated with increased food intake (Krude et al., Nat. Genet. 19:155-157 (1998); Yeo et al., Nature Genetics 20:111-112 (1998); Branson et al., New Engl. J.
Med. 348: 1096-1103 (2003); Vaisse et al., J. Clin. Invest.
106):253-262 (2000); Ho and MacKenzie, J. Biol. Chem. 275: 35816-35822 (1999)).
Weight loss can result from the pharmacological stimulation of melanocortin system activity. In rodents pharmacological stimulation of melanocortin receptors such as MC4R leads to decreased food intake, increased energy expenditure and weight loss (Pierroz et al., Diabetes 51: 1337-1345 (2002)). In man, the intranasal administration of alpha-MSH to stimulate MC4R in non-obese men results in decreased body weight due to the loss of fat-but not lean body mass (Fehm et al., J. Clin. Endo. Metabol. 86:
1144-1148 (2001)).
Obesity is currently treated, with only limited success, by several different strategies. These strategies primarily involve "life-style" changes (e.g., diet and exercise), small molecule-based pharmaceutical therapies or surgical removal of a portion of the stomach (gastric by-pass surgery). Additionally, weight loss stimulating melanocortin receptor binding peptides such as alpha-MSH
are of limited use as pharmaceuticals due to the extremely short serum half-life of such peptides.
Alpha-MSH also plays a role in enhancing male erectile activity.
Targeting the melanocortin receptor with the synthetic melanocortin receptor activator molecule melanotan II (MTII) produced an unexpected side effect of enhancing erectile dysfunction (20). MTII has also been shown to initiate erections in rodents and humans without sexual stimulation in contrast to selective MC4 receptor agonists. Thus, both MC3 and MC4 receptors are likely necessary for complete proerectile erections. Clinical data showed a statistically significant erectile response in healthy male subjects following intranasal or subcutaneous administration of the MTII derivative, PT-141 (bremelanotide) (22, 23). Male erectile dysfunction (ED) is currently treated primarily with PDE5 inhibitors such as VIAGRA , CIALIS and LEVITRA . However, these agents are required to be taken orally approximately one hour before sexual activity.
Bremelanotide is also being tested for use in treating female sexual dysfunction (FSD). The American Foundation for Urologic Disease defines FSD as: "The persistent or recurrent inability to attain or maintain sufficient sexual excitement, causing personal distress. It may be expressed as a lack of subjective excitement or a lack of genital or other somatic responses." FSD consists of four components, hypoactive sexual desire disorder, female sexual arousal disorder (FSAD), anorgasmia and dyspareunia. Some form of FSD appears to be prevalent in approximately 43 percent of the female population.
Laumann et al., JAMA 281, 537-544 (1999).
Alpha-MSH also functions as a cytokine antagonist that inhibits inflammation caused by some of the most prominent mediators of local inflammation (31). For example, alpha-MSH inhibits production and action of proinflammatory cytokines and chemokines (32, 33). Alpha-MSH also inhibits macrophage production of cytotoxic nitric oxide (NO) and neopterin (34,35), prostaglandin E synthesis (37). Alpha-MSH also activates descending anti-inflammatory neural pathways dependent on peripheral beta 2-adrenergic receptors (38) and increases production of interleukin-10 (39).
In allergic inflammation, the anti-inflammatory effects of alpha-MSH peptides were confirmed in acute skin inflammation induced by nonspecific irritants and cytokines (42-46). In addition to its suppressive effect on induction and elicitation of contact hypersensitivity, alpha-MSH induces hapten-specific tolerance in mice through IL-10 release (47).
In gouty (acute) arthritis, ACTH had an anti-inflammatory effect in a rat model of gouty arthritis. The same authors also showed that targeting MC3R subtype could be useful for clinical management of human gouty arthritis and possibly other acute arthritis (48). In rheumatoid arthritis, treatment of rats, who had a preclinical adjuvant-induced rheumatoid arthritis, with alpha-MSH significantly reduced joint pathology. Effectiveness of alpha-MSH was reported to be similar to that of prednisolone.
In inflammatory bowel disease, alpha-MSH administered to mice with dextran sulfate-induced colitis had reduced fecal blood and less weight loss compared to mice receiving placebo (51). Alpha-MSH
administration reduced colonic macroscopic lesions in both acute and chronic colitis induced by trinitrobenzosulfonic acid in rats (52).
In a mouse model of bilateral renal ischemia, alpha-MSH
significantly reduced ischemia-induced renal damage (32).
In liver inflammation and fibrosis, alpha-MSH gene therapy reversed established liver fibrosis in CC14-treated mice (60) . In another study, alpha-MSH inhibited systemic NO production, hepatic neutrophil infiltration and increased hepatic mRNA abundance for TNF-alpha and neutrophil and monocyte chemokines (33).
In ischemic brain damage (stroke), alpha-MSH treatment abolishes intracerebral proinflammatory cytokine gene expression after transient cerebral ischemia and indicates that systemically administered melanocortins may exert neuroprotective efects in cerebral ischemia.
This study showed that alpha-MSH reduced activation of intracerebral TNF-alpha and IL1 beta gene expression after arterial occlusion and reperfusion (40). In another study, melanocortins provided strong protection, with a broad therapeutic window, against inflammatory, apoptotic (incl DNA damage), and histopathological and behavioral consequences of brain ischemia by activating CNS melanocortin 4 (MC4) receptors (41).
In peripheral neuropathies, alpha-MSH and ACTH demonstrated that both peptides stimulated axonal outgrowth from fetal spinal cord slices in vitro in a dose-dependent manner (53). Also, alpha-MSH
promoted sprouting and neurite elongation from dissociated rat spinal and sensory neurons (55).
A need exists for additional treatments for the conditions discussed above and in particular for melanocortin receptor binding molecules with a potentially fast onset of action that overcome the short serum half-life of melanocortin receptor binding peptides such as alpha-MSH.
Brief Description of the Drawings Fig. 1 shows elements of a melanocortin receptor binding mimetibody polypeptide.
Fig 2 is a cartoon of a melanocortin receptor binding mimetibody.
Fig. 3 shows the amino acid (SEQ ID NO: 62) and cDNA (SEQ ID
NO: 61) sequences of a melanocortin receptor binding alpha-MSH
mimetibody. The amino terminal portions of individual mimetibody elements are underlined.
Fig. 4 shows alpha-MSH mimetibody binding to MC4R in a competitive binding assay.
Fig. 5 shows alpha-MSH mimetibody activation of MC4R in cells expressing a high level of MC4R.
Fig. 6 shows alpha-MSH mimetibody activation of MC4R in cells expressing a low level of MC4R.
Fig. 7 shows alpha-MSH mimetibody-mediated decrease in animal food intake.
Fig. 8 shows alpha-MSH mimetibody-mediated decrease in animal body weight.
Summary of the Invention One aspect of the invention is a polypeptide according to formula (I):
(Mp-Lk- (V2) y-Hg-CH2-CH3) (t) (I) where Mp is a biologically active melanocortin receptor binding molecule fragment of SEQ ID NO: 87, 89, 91, 93, 95, 97, or 282, Lk is a polypeptide or chemical linkage, V2 is a portion of a C-terminus of an immunoglobulin variable region, Hg is at least a portion of an immunoglobulin variable hinge region, CH2 is an immunoglobulin heavy chain CH2 constant region and CH3 is an immunoglobulin heavy chain CH3 constant region, y is 0 or 1, and t is independently an integer from 1 to 10.
Another aspect of the invention is a polypeptide comprising a polypeptide having the sequence shown in SEQ ID NO: 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268.
Another aspect of the invention is a polypeptide having the sequence shown in SEQ ID NO: 212.
Another aspect of the invention is a polynucleotide comprising a polynucleotide having the sequence shown in SEQ ID NO: 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267 or a polynucleotide having a sequence complementary to the sequence shown in SEQ ID NO: 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267.
Another aspect of the invention is a polynucleotide comprising a polynucleotide encoding the polypeptide having the sequence shown in SEQ ID NO: 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268.
Another aspect of the invention is a pharmaceutical composition comprising a mimetibody composition of the invention.
Another aspect of the invention is a method of modifying the biological activity of a melanocortin receptor in a cell, tissue or organ, comprising contacting a mimetibody composition of the invention with the cell, tissue or organ.
Another aspect of the invention is a method of modulating at least one melanocortin receptor mediated condition comprising administering a mimetibody composition of the invention to a patient in need thereof.
Detailed Description of the Invention All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as though fully set forth.
The present invention provides polypeptides having the properties of binding a melanocortin receptor and mimicking different isotypes of antibody immunoglobulin molecules such as IgA, IgD, IgE, IgG, or IgM, and any subclass thereof, such as IgA1, IgA2, IgG1, IgG2, IgG3 or IgG4, or combinations thereof, herein after generally referred to as "mimetibodies." In some embodiments, the mimetibody polypeptides of the invention contain an alpha melanocyte stimulating hormone peptide (alpha-MSH) sequence and are designated melanocortin receptor binding alpha-MSH mimetibody. Such alpha-MSH
mimetibody polypeptides can bind melanocortin receptor 4 (MC4R) and MCR5 with equal affinity and MC5R with lower affinity. One result of such melanocortin receptor binding can be the stimulation or inhibition of melanocortin receptor activity. Stimulation or inhibition of melanocortin receptor activity can be useful for treatment of melanocortin receptor mediated conditions.
In one embodiment, the polypeptides of the invention have the generic formula (I):
(Mp-Lk- (V2) y-Hg-CH2-CH3) (t) (I) where Mp is a melanocortin receptor binding molecule, Lk is a polypeptide or chemical linkage, V2 is a portion of a C-terminus of an immunoglobulin variable region, Hg is at least a portion of an immunoglobulin variable hinge region, CH2 is an immunoglobulin heavy chain CH2 constant region and CH3 is an immunoglobulin heavy chain CH3 constant region, y is 0 or 1, and t is independently an integer of 1 to 10.
As used herein, "melanocortin receptor binding molecule" means a molecule, which can bind at least one melanocortin receptor such as Homo sapiens MC4R (SEQ ID NO: 77). Examples of other Homo sapiens melanocortin receptors include MCR1 (SEQ ID NO: 71), MCR2 (SEQ ID NO: 73), MCR3 (SEQ ID NO: 75), and MCR5 (SEQ ID NO: 79). A
given peptide chain is a "melanocortin receptor" if it has at least 85% amino acid sequence identity to a known melanocortin receptor sequence or the mature form of a known melanocortin receptor and can function as a G-protein coupled receptor. Percent identity between two peptide chains can be determined by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9Ø0 (Invitrogen Corp., Carslbad, CA). An exemplary melanocortin receptor binding molecule is the 13 amino acid alpha-MSH peptide having the amino acid sequence shown in SEQ ID NO: 2. Other melanocortin receptor binding molecules include biologically active fragments of SEQ ID NO: 2 and other amino acid sequences that can bind a melanocortin receptor. The term "biologically active fragment" as used herein, refers to a portion of an alpha-MSH
peptide that can bind to a melanocortin receptor such as MC4R. The peptide sequence HFRW (SEQ. ID. NO. 81) is an exemplary "biologically active fragment" of the alpha-MSH peptide sequence SYSMEHFRWGKPV (SEQ ID NO: 2). The HFRW fragment has been incorporated into the structure of the synthetic melanocortin receptor activator molecule melanotan II (MTII) (Fan et al., Nature 385: 165-168 (1997)).
Incorporation of melanocortin receptor binding molecules in the mimetibody polypeptides of the invention provides for binding to melanocortin receptors with a wide range of affinities. The mimetibody polypeptides of the invention may bind a melanocortin receptor with a Kd less than or equal to about 10-7, 10-8, 10-9, 10-10, 10-11 or 10-12 M. The range of obtained IC50 values for aMSH peptide, MTII peptide and aMSH mimetibody were 260-400 nM, 5-30 nM and 200-300 nM, respectively. The affinity of a mimetibody polypeptide for a melanocortin receptor can be determined experimentally using any suitable method. Such methods may utilize Biacore or KinExA
instrumentation, ELISA or competitive binding assays. Mimetibody polypeptides binding specific melanocortin receptors with a desired affinity can be selected from libraries of variants or fragments by techniques known to those skilled in the art.
An alpha-MSH peptide having the amino acid sequence shown in SEQ ID NO: 2 may be modified to obtain other melanocortin receptor binding molecules. Such modifications may comprise the incorporation of C-[X]õ-C motifs into the peptide to conformationally constrain the peptide through the formation of disulfide bonds. In a C-[X]õ-C motif, C is a cysteine residue, X is a amino acid residues and n is an integer necessary to acheive the required conformational constraint. In this instance n can be as little as 1 residue and as high as 50. Exemplary C-[X]õ-C modified peptide sequences are shown in SEQ ID NOs: 4, 6, 8, 10, 89, 91, 93, 95, and 97. The C-[X]õ-C modified peptide sequences can be further modified, if necessary, to prevent N-terminal clipping of mature mimetibodies. For example, SEQ ID NO: 4 or SEQ ID NO: 97 can be modified to remove the N-terminal S-Y-S sequence and replace it with G-G as shown in SEQ ID NO: 282 or SEQ ID NO: 271, respectively.
The modification may also comprise the incorporation of a Wa-[X]õ-Wa motif into the peptide to conformationally constrain the peptide through the formation of a tryptophan zipper. In a Wa-[X]õ-Wa motif W is tryptophan residue, X is an amino acid, a is an integer ususlly 2, but can be from 1 to 10, and n is an integer necessary to acheive the required conformational constraint. In this instance n can be as little a 1 residue and as high as 50.
Exemplary Wa-[X]õ-Wa peptides are shown in SEQ ID NOs: 12, 14, 16 and 18. Further, the sequence HFRW (SEQ ID NO: 81) present in the alpha-MSH peptide may also be modified by substituting any residue in this sequence with any one of F, H, W and M; for example, HFRW
(SEQ ID NO: 81) can be substituted to FHWM (SEQ ID NO: 83).
In the polypeptides of the invention, the linker portion (Lk) provides structural flexibility by allowing the mimetibody to have alternative orientations and binding properties. Exemplary linkers include non-peptide chemical linkages or one to 20 amino acids linked by peptide bonds, wherein the amino acids are selected from the 20 naturally occurring amino acids or other amino acids (e.g. D-amino acids, non-naturally occurring amino acids, or rare naturally occuring amino acids). The linker portion can include a majority of amino acids that are sterically unhindered, such as glycine, alanine and serine and can include GS, poly GS (e.g. GSGS (SEQ ID NO: 20)), GGSG (SEQ ID NO: 22), GSGGGS (SEQ ID NO: 24), GSGGGSG (SEQ ID NO:
26), GSSG (SEQ ID NO: 28), GGGS (SEQ ID NO: 85), GGGGS (SEQ ID NO:
99), GGGGSGGGGS (SEQ ID NO: 101), GGGGSGGGGSGGGGS (SEQ ID NO: 103), GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 105) or any combination or polymer thereof. Other exemplary linkers within the scope of the invention may be longer than 20 residues and may include residues other than glycine, alanine and serine.
In the polypeptides of the invention, V2 is a portion of a carboxy terminal domain of an immunoglobulin variable region such as a heavy chain variable region. Exemplary V2 amino acid sequences are GTLVTVSS (SEQ ID NO: 32), TLVAVSS (SEQ ID NO: 34), and TLVTVSS
(SEQ ID NO: 249).
The (Mp-Lk- (V2) y-Hg-CH2-CH3) (t) mimetibody polypeptides of the invention may comprise "y" V2 polypeptides where y is 0 (zero) or 1 (one). The amino acid sequences shown in SEQ ID NOs: 127, 129, 132, 134, 137, 139, 142, 144, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 256, 259, 261, 264, 266, or 269 are exemplary of mimetibody polypeptides comprising one V2 polypeptide. Stated differently, these mimetibody polypeptides are examples of the formula (Mp-Lk-(V2)y-Hg-CH2-CH3) (t) where y is one. The amino acid sequences in SEQ ID NOs: 60, 62, 121, 123, 147, 149, 152, 154, 157, 159, 162, 164, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 232, 234, 237, 239, 242, 244, 251, or 253 are exemplary of mimetibody polypeptides that lack a V2 polypeptide. In other words, these are mimetibody polypeptides of the formula (Mp-Lk-(V2)y-Hg-CH2-CH3) (t) .
In the polypeptides of the invention, Hg is a portion of the hinge domain of an immunoglobulin variable region such as a heavy chain variable region. Exemplary Hg amino acid sequences include EPKSCDKTHTCPPCP (SEQ ID NO: 36), EPKSADKTHTCPPCP (SEQ ID NO: 38), ESKYGPPCPSCP (SEQ ID NO: 40), ESKYGPPCPPCP (SEQ ID NO: 42), CPPCP
(SEQ ID NO: 44) and CPSC (SEQ ID NO: 46).
Hg amino acid sequences can be modified. Such modifications can remove potential sites of 0-linked glycosylation. Such modifications can also remove cysteine residues that may cause aggregates or multimers of the polypeptides of the invention to form.
One way to minimize 0-linked glycosylation in the mimetibodies of the invention is to substitute Ala residues for Thr residues in the Hg portion of the polypeptides of the invention. The Hg amino acid sequence EPKSCDKTHACPPCP (SEQ ID NO: 107) is exemplary of such a Thr to Ala substitution; this particular Hg substitution can also be obtained by a Thr to Ala substitution at position 59 of SEQ ID
NO: 62.
One way to minimize aggregation or multimerization of the mimetibodies of the invention is to substitute Ala residues for Cys residues in the Hg portion of the polypeptides of the invention.
The Hg amino acid sequence EPKSADKTHTCPPCP (SEQ ID NO: 109) is exemplary of such a Cys to Ala substitution; this particular Hg substitution can also be obtained by a Cys to Ala substitution at position 54 of SEQ ID NO: 62.
Modifications to the Hg amino acid sequences of the mimetibody polypeptides of the invention can be made singly or in combination.
The Hg amino acid sequence EPKSADKTHACPPCP (SEQ ID NO: 111) combines both the aforementioned substitutions; and can be obtained by a Cys to Ala substitution at position 54 and a Thr to Ala substitution at position 59 of SEQ ID NO: 62. Those skilled in the art will recognize other amino acid residues that can be used to make substitutions that remove 0-glycosylation sites and aggregation or multimerization associated sites in the mimetibodies of the invention. Such sites can also be deleted by removing amino acid residues.
In the polypeptides of the invention, CH2 is an immunoglobulin heavy chain CH2 constant region. Exemplary CH2 amino acid sequences include:
APELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK (SEQ ID NO: 48), APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK (SEQ ID NO: 50), APEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAK (SEQ ID NO: 52), APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAK (SEQ ID NO: 54), APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKGLSSPIEKTISKAK (SEQ ID NO: 117), and APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK (SEQ ID NO: 246).
In the polypeptides of the invention, CH3 is an immunoglobulin heavy chain CH3 constant region. Exemplary CH3 amino acid sequences include:
GQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS
KLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 56), GQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS
RLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: 58), and GQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS
RLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: 119). It will be recognized by those skilled in the art that the CH3 region of the polypeptides of the invention may have its C-terminal amino acid cleaved off when expressed in certain recombinant systems.
In the mimetibody polypeptides of invention Hg, CH2 or CH3 may be of the IgG1 or IgG4 subclass. A sequence is of the IgG1 or IgG4 subclass if it is formed or developed from a yl or y4 heavy chain respectively. A given peptide chain is a yl or y4 heavy chain if it is at least 80% identical to a known yl or y4 heavy chain sequence of a given species. Percent identity between two peptide chains can be determined by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9Ø0 (Invitrogen Corp., Carlsbad, CA).
In the mimetibody polypeptides of the invention Hg, CH2 or CH3 may individually be of the IgG2 or IgG4 subclass. The mimetibodies of the invention may also comprise combinations of Hg, CH2 or CH3 elements from each subclass For example, Hg may be of the IgG4 subclass while CH2 and CH3 are of the IgG1 subclass. Alternatively, Hg, CH2 and CH3 may all of the IgG4 or IgG2 subclass. The polypeptide EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVE
VHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTL
PPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQ
GNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 65) is exemplary of a polypeptide in which Hg (residues 1-15 of SEQ ID NO: 65), CH2 (residues 16-125 of SEQ ID NO: 65), and CH3 (residues 126-232 of SEQ
ID NO: 65) are all of the IgG2 subclass.
The IgG1 and IgG4 subclasses differ in the number of cysteines in the hinge region. Most IgG type antibodies, such as IgG1, are homodimeric molecules made up of two identical heavy (H) chains and two identical light (L) chains, typically abbreviated H2L2. Thus, these molecules are generally bivalent with respect to antigen binding due to the formation of inter-heavy chain disulfide bonds and both antigen binding (Fab) arms of the IgG molecule have identical binding specificity. IgG4 isotype heavy chains, in contrast, contain a CPSC
(SEQ ID NO: 46) motif in their hinge regions capable of forming either inter- or intra-heavy chain disulfide bonds, i.e., the two Cys residues in the CPSC motif may disulfide bond with the corresponding Cys residues in the other H chain (inter) or the two Cys residues within a given CPSC motif may disulfide bond with each other (intra).
Since the HL pairs in those IgG4 molecules with intra-heavy chain bonds in the hinge region are not covalently associated with each other, they may dissociate into HL monomers that then reassociate with HL monomers derived from other IgG4 molecules forming bispecific, heterodimeric IgG4 molecules. In vivo isomerase enzymes may facilitate this process. In a bispecific IgG antibody the two Fab "arms" of the antibody molecule differ in the epitopes that they bind.
Substituting Ser residues in the hinge region of IgG4 with Pro results in "IgG1-like behavior," i.e., the molecules form stable disulfide bonds between heavy chains and therefore, are not susceptible to HL exchange with other IgG4 molecules.
The mimetibody polypeptides of the invention may be made more IgG4-like, or IgG2-like by the modification of sites which are involved in disulfide bond formation and are present in the Hg-CH2-CH3 portion of the mimetibody polypeptides. Such sites may be modified by removal, deletion, insertion or substitution with other amino acids. Typically, the cysteine residues present in disulfide bond associated motifs are removed or substituted. Removal of these sites may avoid covalent disulfide bonding with other cysteine-containing proteins present in the mimetibody producing host cell or intra-heavy chain disulfide bonding in IgG4-based constructs while still allowing for noncovalent dimerization of mimetibody Hg-CH2-CH3 domains. Modification of such sites can permit the formation of bispecific mimetibody polypeptides with two different Mp portions or prevent the formation of such bispecific species.
The IgG1 and IgG4 subclasses also differ in their ability to mediate complement dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). CDC is the lysing of a target cell in the presence of complement. The complement activation pathway is initiated by the binding of the first component of the complement system (Clq) to a molecule complexed with a cognate antigen. IgG1 is a strong inducer of the complement cascade and subsequent CDC activity, while IgG4 has little complement-inducing activity. ADCC is a cell-mediated process in which nonspecific cytotoxic cells that express Fc receptors (FcRs) involved in ADCC (e.g., natural killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. The IgG1 subclass binds with high affinity to Fc receptors involved in ADCC and contributes to ADCC, while IgG4 binds only weakly to such receptors and has little ADCC inducing activity. The relative inability of IgG4 to activate effector functions such as ADCC is desirable since delivery of the mimetibody polypeptide to cells without cell killing is possible.
The CDC and ADCC activity of the mimetibody polypeptides of the invention may be modified by altering sites involved in CDC and ADCC present in the Hg-CH2-CH3 portion of the mimetibody polypeptide.
Such sites may be modified by removal, deletion, insertion or substitution with other amino acids. In the mimetibodies of the invention sites involved in CDC, such as the Clq binding site, are typically removed or otherwise modified to minimize CDC activity.
Additionally, Fc receptor binding sites involved in ADCC can also be similarly modified in the mimetibodies of the invention. In general, such modification will remove Fc receptor binding sites involved in ADCC activity from the mimetibodies of the invention.
The substitution of Leu residues with Ala residues in the CH2 portion of the polypeptides of the invention is one example of a modification which can minimize ADCC activity in the polypeptides of the invention. The CH2 amino acid sequences APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK (SEQ ID NO: 52) and APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKGLSSPIEKTISKAK (SEQ ID NO: 54) are exemplary of such a Leu to Ala substitution at residues 4 and 5 (in these sequences). Further, the V1 domain can be removed such that the N-terminus of the peptide is free following cleavage of the signal peptide, and is accessible to and could be modified by enzymes such as acetylases.
Antibodies of both the IgG4 and IgG1 isotypes contain FcRn salvage receptor binding sites. The FcRn salvage receptor helps maintain IgG antibody levels in the body by recycling or transporting IgG type antibodies across enodothelial cell layers such as those lining the inside of body cavities and blood vessels.
The FcRn salavage receptor does this by binding IgGs that have entered endothelial cells by nonspecific pinocytosis and preventing these IgG antibody molecules from being degraded in the lysosome of the cell. The result of such FcRn receptor activity is that the serum half-life of a molecule with an FcRn binding site is extended relative to an otherwise identical molecule lacking such a site.
Exemplary mature mimetibody polypeptides of the invention including a Ser to Pro substitution in the hinge region and CH2 domain Leu to Ala substitutions on an IgG4 backbone have the nucleic and amino acid sequences shown in SEQ ID NOs: 211-215 and 250-254.
In particular, SEQ ID NOs: 212 and 214 show the amino acid sequence of a mimetibody polypeptide having an Mp sequence as shown in SEQ ID
NO: 4 and an Lk sequence as shown in SEQ ID NO: 101 without and with a signal sequence, respectively. SEQ ID NOs: 211 and 213 show exemplary nucleic acid sequences encoding these polypeptides.
Further, SEQ ID NOs: 251 and 253 show the amino acid sequence of a mimetibody polypeptide having an Mp sequence as shown in SEQ ID NO:
282 and an Lk sequence as shown in SEQ ID NO: 101 without and with a signal sequence, respectively. SEQ ID NOs: 250 and 252 show exemplary nucleic acid sequences encoding these polypeptides.
It is desirable that the Hg-CH2-CH3 portion of the mimetibodies of the invention contain a FcRn binding site at the junction of the CH2 and CH3 regions. It is expected that such FcRn sites will increase the serum half-life of the mimetibodies of the invention as well as improve other pharmacokinetic properties relative to a melanocortin receptor binding molecule, such as alpha-MSH alone. In the mimetibodies of the invention FcRn sites may be modified or added by removal, deletion, insertion or substitution of amino acids. Typically, such modifications are used to improve the binding of a given site to the FcRn. One example of a human FcRn binding sites is the sequence MISRTPTVLHQHNHY (SEQ. ID. NO.: 69) found in both IgG1 and IgG4 antibodies. Other FcRn binding sites are well known by those skilled in the art.
Antibodies with different isotypes, such as IgG4 and IgG1, may contain glycosylation sites. Glycosylation of these sites can alter the properties and activites of antibody molecules. Antibody molecules may be N-glycosylated or 0-glycosylated. N-glycosylation of antibody amino acid residue side chains containing nitrogen atoms (e.g., Asn) can modulate antibody Fc effector functions such as ADCC
by conferring a cytolytic activity to N-glycosylated antibody molecules. This ADCC associated cytolytic activity causes the lysis of cells effected by such N-glycosylated antibodies. Alternatively, an antibody molecule may be 0-glycosylated by modification of amino acid residue side chains containing oxygen atoms (e.g., Ser or Thr).
0-glycosylation can decrease the serum half-life of an antibody molecule through increased lectin mediated clearance of 0-glycosylated antibody molecules from the serum. Additionally, 0-glycosylation can cause undesirable increases in antibody heterogeneity due to differing extents of 0-glycosylation between various antibody molecules. Lastly, both 0-glycosylation and N-glycosylation can alter the structure dependent properties of antibody molecules such as binding affinity and immunogenicity.
Like the antibody molecules they mimic, the mimetibody polypeptides of the invention may also be post-translationally modified by N-glycosylation and 0-glycosylation. In most instances, it is desirable to limit the N-glycosylation of the mimetibodies of the invention to minimize cytolytic activity. N-glycosylation can be limited by the removal or substitution of amino acid residues, such as Asn, which are typically N-glycosylated. It is also desirable to limit mimetibody 0-glycosylation to minimize lectin-mediated clearance, mimetibody heterogeneity and the alteration of structure dependent mimetibody properties such as binding affinity and immunogenicity. One way to minimize 0-linked glycosylation in the mimetibodies of the invention is to substitute Ala residues for Thr residues in the V2 portion of the polypeptides of the invention.
The V2 amino acid sequence TLVAVSS (SEQ ID NO: 34) is exemplary of such a Thr to Ala substitution; this particular V2 substitution can also be obtained by a Thr to Ala substitution at postion 47 of SEQ
ID NO: 62. Those skilled in the art also will recognize other ways to control N-linked and 0-linked glycosylation including modulation of glycosylase enzyme activity.
The monomeric structure Mp-Lk- (V2) y-Hg-CH2-CH3 of the mimetibody polypeptides of the invention can be linked to "t" other monomers where t is an integer from 1 to 10. Such linking can occur through non-covalent interactions or covalent linkages such as a Cys-Cys disulfide bond. In this way multimeric structures such as dimers and higher order multimers of the polypeptides of the invention can be formed. It is expected that dimerization of the polypeptides of the invention will increase the affinity of these polypeptides to melanocortin receptors such as MC4R. The term "multimers" as used herein means molecules that have quaternary structure and are formed by the association of two or more subunits.
The polypeptides of the invention can optionally comprise at the amino terminus, an amino terminal portion of an immunoglobulin variable region, designated V1 as shown in Formula II:
(V1-Mp-Lk- (V2) v-Hg-CH2-CH3) (t) (II) Exemplary V1 amino acid sequences include QIQ, QVQ, QIQGG (SEQ ID
NO: 113), and QIQGGGG (SEQ ID NO: 115).
The polypeptides of the invention may also comprise secretory signals necessary to facilitate protein secretion or other signals necessary for protein trafficking in the cell. An exemplary secretory signal sequence is MAWVWTLLFLMAAAQSIQA (SEQ ID NO: 69).
Those skilled in the art will recognize other secretory signals.
In one embodiment the polypeptides of the invention comprise polypeptides having the sequences shown in SEQ ID NO: 60, 62, 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268. These sequences exemplify melanocortin receptor binding alpha-MSH polypetides either having, or lacking, an amino terminal secretory signal sequence. SEQ ID NO: 62 represents a (V1-Mp-Lk-(V2)y-Hg-CH2-CH3) (t) melanocortin receptor binding alpha-MSH polypetide of generic formula (II) which has the secretory signal MAWVWTLLFLMAAAQSIQA (SEQ ID NO: 69) fused to its amino terminus. SEQ ID NO: 60 represents a (Mp-Lk- (V2) y-Hg-CH2-CH3) (t) melanocortin receptor binding alpha-MSH polypetide of generic formula (I). No secretory signal is present in SEQ ID NO: 60.
Another aspect of the present invention is a polynucleotide comprising, complementary to or having significant identity with, a polynucleotide encoding at least one melanocortin receptor binding mimetibody. Other aspects of the present invention include vectors comprising at least one polynucleotide molecule encoding a melanocortin receptor binding mimetibody. In a different aspect the invention provides a cell comprising a vector of the invention or a cell expressing a mimetibody polypeptide of the invention. The polynucleotides, vectors and cells may be used to produce the mimetibody polypeptides of the invention.
In one embodiment, the polynucleotides of the invention comprise a polynucleotide having the sequence shown in SEQ ID NO:
59, 61, 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267 or a polynucleotide having a sequence complementary to the sequence shown in SEQ ID NO: 59, 61, 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267. SEQ ID NO: 59 is a cDNA encoding a (Mp-Lk- (V2) y-Hg-CH2-CH3) (t) melanocortin receptor binding alpha-MSH
polypetide of generic formula (I) which lacks a signal sequence.
SEQ ID NO: 61 is a cDNA encoding a (V1-Mp-Lk- (V2) Y-Hg-CH2-CH3) (t) melanocortin receptor binding alpha-MSH polypetide of generic formula (II) which has a secretory signal fused to its amino terminus.
In one embodiment, the polynucleotides of the invention comprise a polynucleotide encoding the polypeptide having the sequence shown in SEQ ID NO: 60, 62, 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268.
Exemplary nucleic acid sequences that encode the polypeptide sequences shown in SEQ ID NO 60, 62, 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268 are shown in SEQ ID NO: 59, 61, 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267, respectively.
Also provided are polynucleotides encoding polypeptides that are substantially identical to the polypeptides of the invention.
The term "substantially identical" in the context of polypeptides means that a given polypeptide sequence is identical to a polypeptide sequence of the invention, in particular the V1-Mp-Lk-V2-Hg region, in at least 50% or at least about 60% or at least about 70% or at least about 80% or at least about 90% or at least about 95-98% of the amino acid residues. Percent identity between two polypeptide sequences can be determined by pairwise alignment using the default settings of the AlignX module of Vector NTI
v.9Ø0 (Invitrogen Corp., Carlsbad, CA). Those skilled in the art would recognize polynucleotide sequences which would encode the above-described polypeptides.
Typically, the polynucleotides of the invention are used in expression vectors for the preparation of the mimetibody polypeptides of the invention. Vectors within the scope of the invention provide necessary elements for eukaryotic expression and include viral promoter driven vectors, such as CMV promoter driven vectors, e.g., pcDNA3.1, pCEP4, and their derivatives, Baculovirus expression vectors, Drosophila expression vectors, and expression vectors that are driven by mammalian gene promoters, such as human Ig gene promoters. Other examples include prokaryotic expression vectors, such as T7 promoter driven vectors, e.g. pET41, lactose promoter driven vectors and arabinose gene promoter driven vectors.
The present invention also relates to a cell that expresses a mimetibody of the invention or comprises a vector of the invention.
Open reading frames encoding the mimetibody polypeptides of the invention can be identified by translation of the positive strand reading frame beginning with nucleotide residue 1601 of SEQ ID NOs:
63, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 254, 259, 264, or 269. The various signal peptide, V1, Mp, V2, Hg, CH2, and CH3 portions of the mimetibody polypeptides of the invention, which have been exemplified herein, are also present in these open reading frames and can be identified using standard sequence analysis methods, such as multiple sequence alignment or other methods well know in the art. Such a cell can be prokaryotic or eukaryotic. Exemplary eukaryotic cells are mammalian cells, such as but not limited to, COS-1, COS-7, HEK293, BHK21, CHO, BSC-1, HepG2, 653, SP2/0, NSO, 293, HeLa, myeloma, lymphoma cells or any derivative thereof. Most preferably, the eukaryotic cell is a HEK293, NSO, SP2/0, or CHO cell. E. coli is an exemplary prokaryotic cell. A cell according to the invention may be generated by transfection, cell fusion, immortalization, or other procedures that are well known in the art. Polynucleotides transfected into a cell may be extrachromasomal or stably integrated into the chromosome of the cell.
The mimetibodies of the invention can be made more compatible with a given host cell by modification of the Hg-CH2-CH3 portion of the polypeptide. For example, when a mimetibody of the invention is expressed recombinantly in a bacterial cell such as E. coli, the Pro-Ala sequence in the Hg element may be removed to prevent digestion by the E. coli enzyme proline iminopeptidase. Similarly, a portion of the Hg element can be deleted or substituted with other amino acids in the mimetibodies of the invention to prevent heterogeneity in the products expressed in a selected host cell.
The present invention further provides a method to produce a mimetibody polypeptide comprising the steps of culturing a cell of the invention and purifying an expressed mimetibody polypeptide of the invention. Cell components, such as those necessary for in vitro transcription and translation, may also be used to express the polypeptides of the invention. The present invention encompasses mimetibodies produced by both methods. Expressed mimetibody polypeptides can be recovered and purified from cells or cell component based systems by methods well known in the art including, but not limited to, protein A purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylatpatite chromatography and lectin chromatography. High performance liquid chroatography (HPLC) can also be employed for purification. Typically purfication will require a combination of several different methods.
Another aspect of the present invention is a pharmaceutical composition comprising an effective amount of at least one mimetibody polypeptide and a pharmaceutically acceptable carrier or diluent. The term "effective amount" generally refers to the quantity of mimetibody necessary for effective therapy, i.e., the partial or complete alleviation of the symptom or disorder for which treatment was sought. The composition can optionally comprise at least one further compound, protein or composition useful for treating obesity and the other conditions described below. The parmaceutically acceptable carrier or diluent in the compositions can be a solution, suspension, emulsion, colloid or powder. Those skilled in the art will recognize other parmaceutically acceptable carriers and diluents.
Another aspect of the present invention is a method of modifying the biological activity of a melanocortin receptor in a cell, tissue or organ comprising contacting the pharmaceutical compositions of the invention with the cell, tissue or organ. The method may be used to modify melanocortin receptor activity in the brain, brain tissue, or brain cells. Alternatively, the method of the invention may be used to modify melanocortin receptor activity in other peripheral cells or tissues such as muscle, or other organs such as the stomach. Those skilled in the art will recognize other cells, tissues or organs, which may be used.
Another aspect of the invention is a method of modulating at least one melanocortin receptor-mediated condition comprising administering a pharmaceutical composition of the invention to a patient in need thereof. The pharmaceutical compositions of the invention can be administered by any suitable route. Such routes may be intrathecal, intranasal, peripheral (e.g., subcutaneous, intramuscular, intradermal, intravenous) or by any other means known in the art. As described previously, abnormal melanocortin receptor activity has been implicated in a number of pathological conditions, such as obesity and Type 2 diabetes. Stimulation of MC4R can cause weight loss while inhibition may cause weight gain. The mimetibody polypeptides of the invention may be also be used to modulate other melanocortin receptor mediated conditions such as male and female erectile dysfunction, inflammation, congestive heart failure, central nervous system disorders, nerve damage, infectious disease, pulmonary disease, skin disease, fever and pain.
The present invention is further described with reference to the following examples. These examples are merely to illustrate aspects of the present invention and are not intended as limitations of this invention.
Example 1 Alpha-MSH Mimetibody and Expression Vector Construction An alpha-MSH mimetibody protein comprising a secretory signal sequence, an alpha-MSH peptide sequence, a linker sequence, VH
sequence, a hinge sequence, a human IgG1 CH2 sequence and a human IgG1 CH3 sequence was designed (Fig. 3 and SEQ ID NO. 62) Analytical data, e.g., mass spectroscopy, has confirmed that a mature polypeptide is generated (61,344.6 for G1/G1 form). Nucleic acid sequences encoding this alpha-MSH mimetibody protein (Fig. 3; SEQ ID
NO: 61) were generated using standard molecular biology techniques.
Nucleic acid sequences encoding the alpha-MSH mimetibody sequence were subcloned into the p2389 expression vector to generate an alpha-MSH mimetibody expression vector (SEQ ID NO: 63).
Example 2 Alpha-MSH Mimetibody Expression The alpha-MSH mimetibody was transiently expressed in HEK293E
cells. Cells were cultured using standard conditions and transiently transfected with the alpha-MSH mimetibody expression vector using Lipofectamine 2000 (Invitrogen, Carlsbad, CA) as directed by the manufacturer. 24 h after transfection cells were transferred to a serum free media formulation and cultured for 5 days. The culture media was then removed and centrifuged to remove debris. Clarified media was incubated with Protein A-SepharoseTM
(HiTrap rProtein A FF, Amersham Biosciencies, Piscataway, NJ) and proteins were eluted from the Protein A-SepharoseTM conjugate as directed by the manufacturer. The eluted protein solution was then further purified via SuperoseTM 12 size exclusion chromatography (Superose 12 10/300 GL, Amersham Biosciencies, Piscataway, NJ) using standard methods. Column eluant was then subjected to SDS-PAGE and visualized by silver and Coomassie blue staining. Western blots were then prepared and the blots were probed with either an Fc specific primary antibody or an alpha-MSH specific primary antibody.
Together, the Western Blot and SDS-PAGE staining results indicated that a purified alpha-MSH mimetibody, composed of two polypeptide chains, had been obtained from the transiently transfected HEK293 cells.
Example 3 Alpha-MSH Mimetibody Binds MC4R
The alpha-MSH mimetibody binds to MC4R and can compete with radiolabeled [Nle(4), D-Phe(7)]-alpha-MSH (NDP-alpha-MSH) agonist molecules for MC4R binding (Fig. 4). MC4R is a receptor for alpha-MSH. alpha-MSH binding to recombinantly expressed MC4R in HEK293 cell membranes (Perkin Elmer Life and Analytical Sciences, Boston, MA) was examined by competive binding assays in which increasing amounts of unlabeled MC4R agonists (positive controls) and the Fc domain of a human antibody (negative control) were added to assay cocktails containing [125I]-NDP-alpha-MSH as indicated in Fig. 4.
The unlabeled MC4R agonists were melanotan II (MTII; an alpha MSH
analog), alpha-MSH, and NDP-alpha-MSH. Alpha-MSH mimetibody binding to MC4R was stable after two weeks of storage at 4 C, -20 C, and -80 C in PBS (phosphate buffered saline) as assessed by competive binding assays.
Competivive binding assays were performed using Scintillation Proximity Assays (Amersham Biosciences Corp, Piscataway, NJ) as directed by the assay manufacturer. Assay cocktails contained [1251]-NDP-alpha-MSH at EC80, i.e., -0.5 nM, 0.1 pg of MC4R
membranes, 1 mM MgS04, 1.5 mM CaC12, 25 mM Hepes, 0.2% BSA, 1 mM
1,10-phenthroline, an assay manufacturer recommended quantity of protease inhibitor cocktail (Roche Diagnostics Corp., Indianapolis, IN) and SPA beads. Light emission from Scintillation Proximity Assay beads was measured with a Packard Top Count NXT Instrument (Perkin Elmer Life and Analytical Sciences, Boston, MA) for 5 minutes.
Example 4 Alpha-MSH Mimetibody Activates MC4R
The alpha-MSH mimetibody can activate MC4R signalling to increase cAMP production in CHOK1 cells expressing MC4R (Fig. 5 and Fig. 6). MC4R is a seven transmembrane (7TM) G-protein coupled receptor. Activation of MC4R by ligand or agonist results in an increase in cyclic AMP levels (cAMP).
MC4R receptor activation assays were performed using two different clonal CHOK1 cell lines stably transfected with a MC4R
expression vector and expressing MC4R. Clone 1 (Fig. 5) expressed MC4R at high levels relative to Clone 2 (Fig. 6). Clone 1 and Clone 2 cells were grown as a monolayer using standard culture conditions to a density of approximately 100,000 cells/well and then incubated with increasing amounts (0-100 pM) of alpha-MSH, MTII, or alpha-MSH
mimetibody for 15 minutes as indicated in Fig. 5 and Fig. 6. Cells were then lysed and cAMP assays were performed using the cAMP-Screen Direct'' Chemiluminescent Immunoassay System (Applied Biosystems, Foster City, CA) as directed by the manufacturer. EC50 values from cAMP assays using Clone 1 (Fig. 5) and Clone 2 (Fig. 6) are listed in Table 1 below Table 1 Clone 1 Clone 2 alpha-MSH peptide EC50 = 3.29 nM EC50 = 9.46 nM
(Positive control) MT II EC50 = 0.52 nM EC50 = 0.52 nM
(Positive control) alpha-MSH mimetibody EC50 = 14.36 nM EC50 = 52.4 nM
Example 5 Alpha-MSH Mimetibody Administration Decreases Animal Food Intake and Body Weight Alpha-MSH mimetibody administration to Rattus norvegicus brain ventricules decreases animal food intake (Fig. 7) and body weight (Fig. 8). Alpha-MSH mimetibody was supplied to brain ventricules by intracerebroventricular injections (ICV) via a cannula surgically inserted into the left lateral brain ventricle.
Cannulae were surgically inserted into male Sprague-Dawley or Wistar rats weighing 250 g to 350 g. Cannula placement coordinates were as follows: -0.8 mm from bregma, -4.5 mm ventral and -1.5 posterior-anterior. Animals recovered for 7 to 10 days after surgery. Animals were acclimatized to the experimental procedures by both daily handling and mock injection, in order to minimize stress. In addition animals were submitted to the reversal of dark-light cycle.
Proper cannula placement was confirmed by an angiotensin II
test. The test confirmed proper cannula placement if the ICV
administration of 10 ng of angiotensin II via the cannula caused the rats to drink 5-10 ml of water in 30 minutes. Only animals that passed this angiotensin II test were used in food intake experiments.
Animals were fasted for 18-24 hours and alpha-MSH mimetibody, alpha-MSH (positive control), or PBS (negative control) were then administered to the brain ventricles via the cannula at an injection rate of 9 pl/min. Each treatment group had a minimum of 7 animals.
Treatments and dosages were as indicated in Fig. 7 and Fig. 8.
Food and water was given to the animals after injection. The amount of food and water consumed was measured at 0 h, 4 h, 24 h, 48 h and 72 h (Fig. 7) after injection. Body weight at 72 hours post injection was measured as ahown in Fig. 8.
The present invention now being fully described, it will be apparent to one of ordinary skill in the art that many changes and modifications can be made thereto without departing from the spirit or scope of the appended claims.
Cross-Reference to Related Applications This application is a continuation-in-part of United States Application Serial No. 11/257,851, filed 25 October 2005, which claims priority to United States Provisional Application Serial No.
60/621,960, filed 25 October 2004. This application also claims the benefit of United States Provisional Application Serial No.
60/972,018, filed 13 September 2007. The entire contents of each of the aforementioned applications is incorporated herein by reference in their entirety.
Field of the Invention The present invention relates to melanocortin receptor binding mimetibodies, polynucleotides encoding the mimetibodies, cells comprising the polynucleotides or expressing the mimetibodies, and methods of making and using the foregoing.
Background of the Invention Obesity is a chronic disease manifested by an excess of fat mass in proportion to body size. Today, every third American is considered overweight (Body Mass Index (BMI) >25 kg/m2), thus prompting the United States Centers for Disease Control and Prevention (CDC) to declare that obesity is reaching epidemic proportions (Cummings and Schwartz, Annu. Rev. Med. 54:453-471((2003)). The importance of treating obesity is emphasized by the fact that this disease is either the underlying cause, or a risk factor, for developing diseases such as Type 2 Diabetes, congestive heart failure, osteoarthritis and sleep apnea among others.
Additionally, obesity is linked to "Metabolic Syndrome" which is a medical condition characterized by obesity, atherogenic dyslipidemia, elevated blood pressure and insulin resistance.
Metabolic Syndrome affects an increasing number of people in the United States. Importantly, it has been shown that even a modest decrease in body weight (5-10% of initial body weight) may significantly improve Metabolic Syndrome conditions and decrease the risk factors for developing obesity-associated disease (Wing et al., Arch. Intern. Med. 147:1749-1753 (1987); Tuomilehto et al., New Engl. J. Med. 344:1343-1350 (2001); Knowler et al., New Engl. J Med.
346:393-403 (2002); Franz et al., Diabetes Care 25:148-198 (2002)).
Additionally, treatment of obesity may be important from a mental health perspective due to the social stigma often attached to obese individuals in some cultures.
Melanocortin receptors play a major role in the regulation of overall energy balance and obesity in both humans and rodents.
Alpha-melanocyte stimulating hormone (alpha-MSH) is a 13 amino acid peptide hormone that is an important component of the melanocortin system. Alpha-MSH is produced by the proteolytic processing of proopiomelanocortin (POMC) released by the pituitary gland. Alpha-MSH binds with high affinity to the melanocortin 4 receptor (MC4R), but also binds melanocortin receptor 3 (MC3R) and melanocortin receptor 5 (MC5R) with lower affinity. MC4R is a G-coupled protein receptor found in the brain which, when stimulated by alpha-MSH
binding, causes decreased food intake and increased fat oxidation.
Ultimately, stimulation of melanocortin receptors such as MC4R
results in weight loss.
In humans and rodents, loss of function mutations in the different components of the melanocortin system are closely correlated with obesity and related conditions. In mice, mutations within POMC, or MC4R and MC3R produce obesity, insulin resistance and hyperphagia (Goodfellow and Saunders, Curr. Topics Med. Chem. 3:
855-883 (2003); Huszar et al., Cell 88:131-141 (1997); Yaswen et al., Nat. Med. 5: 1066-1070 (1999)). In man, mutations within POMC
or MC4R lead to the development of obesity associated with increased food intake (Krude et al., Nat. Genet. 19:155-157 (1998); Yeo et al., Nature Genetics 20:111-112 (1998); Branson et al., New Engl. J.
Med. 348: 1096-1103 (2003); Vaisse et al., J. Clin. Invest.
106):253-262 (2000); Ho and MacKenzie, J. Biol. Chem. 275: 35816-35822 (1999)).
Weight loss can result from the pharmacological stimulation of melanocortin system activity. In rodents pharmacological stimulation of melanocortin receptors such as MC4R leads to decreased food intake, increased energy expenditure and weight loss (Pierroz et al., Diabetes 51: 1337-1345 (2002)). In man, the intranasal administration of alpha-MSH to stimulate MC4R in non-obese men results in decreased body weight due to the loss of fat-but not lean body mass (Fehm et al., J. Clin. Endo. Metabol. 86:
1144-1148 (2001)).
Obesity is currently treated, with only limited success, by several different strategies. These strategies primarily involve "life-style" changes (e.g., diet and exercise), small molecule-based pharmaceutical therapies or surgical removal of a portion of the stomach (gastric by-pass surgery). Additionally, weight loss stimulating melanocortin receptor binding peptides such as alpha-MSH
are of limited use as pharmaceuticals due to the extremely short serum half-life of such peptides.
Alpha-MSH also plays a role in enhancing male erectile activity.
Targeting the melanocortin receptor with the synthetic melanocortin receptor activator molecule melanotan II (MTII) produced an unexpected side effect of enhancing erectile dysfunction (20). MTII has also been shown to initiate erections in rodents and humans without sexual stimulation in contrast to selective MC4 receptor agonists. Thus, both MC3 and MC4 receptors are likely necessary for complete proerectile erections. Clinical data showed a statistically significant erectile response in healthy male subjects following intranasal or subcutaneous administration of the MTII derivative, PT-141 (bremelanotide) (22, 23). Male erectile dysfunction (ED) is currently treated primarily with PDE5 inhibitors such as VIAGRA , CIALIS and LEVITRA . However, these agents are required to be taken orally approximately one hour before sexual activity.
Bremelanotide is also being tested for use in treating female sexual dysfunction (FSD). The American Foundation for Urologic Disease defines FSD as: "The persistent or recurrent inability to attain or maintain sufficient sexual excitement, causing personal distress. It may be expressed as a lack of subjective excitement or a lack of genital or other somatic responses." FSD consists of four components, hypoactive sexual desire disorder, female sexual arousal disorder (FSAD), anorgasmia and dyspareunia. Some form of FSD appears to be prevalent in approximately 43 percent of the female population.
Laumann et al., JAMA 281, 537-544 (1999).
Alpha-MSH also functions as a cytokine antagonist that inhibits inflammation caused by some of the most prominent mediators of local inflammation (31). For example, alpha-MSH inhibits production and action of proinflammatory cytokines and chemokines (32, 33). Alpha-MSH also inhibits macrophage production of cytotoxic nitric oxide (NO) and neopterin (34,35), prostaglandin E synthesis (37). Alpha-MSH also activates descending anti-inflammatory neural pathways dependent on peripheral beta 2-adrenergic receptors (38) and increases production of interleukin-10 (39).
In allergic inflammation, the anti-inflammatory effects of alpha-MSH peptides were confirmed in acute skin inflammation induced by nonspecific irritants and cytokines (42-46). In addition to its suppressive effect on induction and elicitation of contact hypersensitivity, alpha-MSH induces hapten-specific tolerance in mice through IL-10 release (47).
In gouty (acute) arthritis, ACTH had an anti-inflammatory effect in a rat model of gouty arthritis. The same authors also showed that targeting MC3R subtype could be useful for clinical management of human gouty arthritis and possibly other acute arthritis (48). In rheumatoid arthritis, treatment of rats, who had a preclinical adjuvant-induced rheumatoid arthritis, with alpha-MSH significantly reduced joint pathology. Effectiveness of alpha-MSH was reported to be similar to that of prednisolone.
In inflammatory bowel disease, alpha-MSH administered to mice with dextran sulfate-induced colitis had reduced fecal blood and less weight loss compared to mice receiving placebo (51). Alpha-MSH
administration reduced colonic macroscopic lesions in both acute and chronic colitis induced by trinitrobenzosulfonic acid in rats (52).
In a mouse model of bilateral renal ischemia, alpha-MSH
significantly reduced ischemia-induced renal damage (32).
In liver inflammation and fibrosis, alpha-MSH gene therapy reversed established liver fibrosis in CC14-treated mice (60) . In another study, alpha-MSH inhibited systemic NO production, hepatic neutrophil infiltration and increased hepatic mRNA abundance for TNF-alpha and neutrophil and monocyte chemokines (33).
In ischemic brain damage (stroke), alpha-MSH treatment abolishes intracerebral proinflammatory cytokine gene expression after transient cerebral ischemia and indicates that systemically administered melanocortins may exert neuroprotective efects in cerebral ischemia.
This study showed that alpha-MSH reduced activation of intracerebral TNF-alpha and IL1 beta gene expression after arterial occlusion and reperfusion (40). In another study, melanocortins provided strong protection, with a broad therapeutic window, against inflammatory, apoptotic (incl DNA damage), and histopathological and behavioral consequences of brain ischemia by activating CNS melanocortin 4 (MC4) receptors (41).
In peripheral neuropathies, alpha-MSH and ACTH demonstrated that both peptides stimulated axonal outgrowth from fetal spinal cord slices in vitro in a dose-dependent manner (53). Also, alpha-MSH
promoted sprouting and neurite elongation from dissociated rat spinal and sensory neurons (55).
A need exists for additional treatments for the conditions discussed above and in particular for melanocortin receptor binding molecules with a potentially fast onset of action that overcome the short serum half-life of melanocortin receptor binding peptides such as alpha-MSH.
Brief Description of the Drawings Fig. 1 shows elements of a melanocortin receptor binding mimetibody polypeptide.
Fig 2 is a cartoon of a melanocortin receptor binding mimetibody.
Fig. 3 shows the amino acid (SEQ ID NO: 62) and cDNA (SEQ ID
NO: 61) sequences of a melanocortin receptor binding alpha-MSH
mimetibody. The amino terminal portions of individual mimetibody elements are underlined.
Fig. 4 shows alpha-MSH mimetibody binding to MC4R in a competitive binding assay.
Fig. 5 shows alpha-MSH mimetibody activation of MC4R in cells expressing a high level of MC4R.
Fig. 6 shows alpha-MSH mimetibody activation of MC4R in cells expressing a low level of MC4R.
Fig. 7 shows alpha-MSH mimetibody-mediated decrease in animal food intake.
Fig. 8 shows alpha-MSH mimetibody-mediated decrease in animal body weight.
Summary of the Invention One aspect of the invention is a polypeptide according to formula (I):
(Mp-Lk- (V2) y-Hg-CH2-CH3) (t) (I) where Mp is a biologically active melanocortin receptor binding molecule fragment of SEQ ID NO: 87, 89, 91, 93, 95, 97, or 282, Lk is a polypeptide or chemical linkage, V2 is a portion of a C-terminus of an immunoglobulin variable region, Hg is at least a portion of an immunoglobulin variable hinge region, CH2 is an immunoglobulin heavy chain CH2 constant region and CH3 is an immunoglobulin heavy chain CH3 constant region, y is 0 or 1, and t is independently an integer from 1 to 10.
Another aspect of the invention is a polypeptide comprising a polypeptide having the sequence shown in SEQ ID NO: 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268.
Another aspect of the invention is a polypeptide having the sequence shown in SEQ ID NO: 212.
Another aspect of the invention is a polynucleotide comprising a polynucleotide having the sequence shown in SEQ ID NO: 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267 or a polynucleotide having a sequence complementary to the sequence shown in SEQ ID NO: 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267.
Another aspect of the invention is a polynucleotide comprising a polynucleotide encoding the polypeptide having the sequence shown in SEQ ID NO: 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268.
Another aspect of the invention is a pharmaceutical composition comprising a mimetibody composition of the invention.
Another aspect of the invention is a method of modifying the biological activity of a melanocortin receptor in a cell, tissue or organ, comprising contacting a mimetibody composition of the invention with the cell, tissue or organ.
Another aspect of the invention is a method of modulating at least one melanocortin receptor mediated condition comprising administering a mimetibody composition of the invention to a patient in need thereof.
Detailed Description of the Invention All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as though fully set forth.
The present invention provides polypeptides having the properties of binding a melanocortin receptor and mimicking different isotypes of antibody immunoglobulin molecules such as IgA, IgD, IgE, IgG, or IgM, and any subclass thereof, such as IgA1, IgA2, IgG1, IgG2, IgG3 or IgG4, or combinations thereof, herein after generally referred to as "mimetibodies." In some embodiments, the mimetibody polypeptides of the invention contain an alpha melanocyte stimulating hormone peptide (alpha-MSH) sequence and are designated melanocortin receptor binding alpha-MSH mimetibody. Such alpha-MSH
mimetibody polypeptides can bind melanocortin receptor 4 (MC4R) and MCR5 with equal affinity and MC5R with lower affinity. One result of such melanocortin receptor binding can be the stimulation or inhibition of melanocortin receptor activity. Stimulation or inhibition of melanocortin receptor activity can be useful for treatment of melanocortin receptor mediated conditions.
In one embodiment, the polypeptides of the invention have the generic formula (I):
(Mp-Lk- (V2) y-Hg-CH2-CH3) (t) (I) where Mp is a melanocortin receptor binding molecule, Lk is a polypeptide or chemical linkage, V2 is a portion of a C-terminus of an immunoglobulin variable region, Hg is at least a portion of an immunoglobulin variable hinge region, CH2 is an immunoglobulin heavy chain CH2 constant region and CH3 is an immunoglobulin heavy chain CH3 constant region, y is 0 or 1, and t is independently an integer of 1 to 10.
As used herein, "melanocortin receptor binding molecule" means a molecule, which can bind at least one melanocortin receptor such as Homo sapiens MC4R (SEQ ID NO: 77). Examples of other Homo sapiens melanocortin receptors include MCR1 (SEQ ID NO: 71), MCR2 (SEQ ID NO: 73), MCR3 (SEQ ID NO: 75), and MCR5 (SEQ ID NO: 79). A
given peptide chain is a "melanocortin receptor" if it has at least 85% amino acid sequence identity to a known melanocortin receptor sequence or the mature form of a known melanocortin receptor and can function as a G-protein coupled receptor. Percent identity between two peptide chains can be determined by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9Ø0 (Invitrogen Corp., Carslbad, CA). An exemplary melanocortin receptor binding molecule is the 13 amino acid alpha-MSH peptide having the amino acid sequence shown in SEQ ID NO: 2. Other melanocortin receptor binding molecules include biologically active fragments of SEQ ID NO: 2 and other amino acid sequences that can bind a melanocortin receptor. The term "biologically active fragment" as used herein, refers to a portion of an alpha-MSH
peptide that can bind to a melanocortin receptor such as MC4R. The peptide sequence HFRW (SEQ. ID. NO. 81) is an exemplary "biologically active fragment" of the alpha-MSH peptide sequence SYSMEHFRWGKPV (SEQ ID NO: 2). The HFRW fragment has been incorporated into the structure of the synthetic melanocortin receptor activator molecule melanotan II (MTII) (Fan et al., Nature 385: 165-168 (1997)).
Incorporation of melanocortin receptor binding molecules in the mimetibody polypeptides of the invention provides for binding to melanocortin receptors with a wide range of affinities. The mimetibody polypeptides of the invention may bind a melanocortin receptor with a Kd less than or equal to about 10-7, 10-8, 10-9, 10-10, 10-11 or 10-12 M. The range of obtained IC50 values for aMSH peptide, MTII peptide and aMSH mimetibody were 260-400 nM, 5-30 nM and 200-300 nM, respectively. The affinity of a mimetibody polypeptide for a melanocortin receptor can be determined experimentally using any suitable method. Such methods may utilize Biacore or KinExA
instrumentation, ELISA or competitive binding assays. Mimetibody polypeptides binding specific melanocortin receptors with a desired affinity can be selected from libraries of variants or fragments by techniques known to those skilled in the art.
An alpha-MSH peptide having the amino acid sequence shown in SEQ ID NO: 2 may be modified to obtain other melanocortin receptor binding molecules. Such modifications may comprise the incorporation of C-[X]õ-C motifs into the peptide to conformationally constrain the peptide through the formation of disulfide bonds. In a C-[X]õ-C motif, C is a cysteine residue, X is a amino acid residues and n is an integer necessary to acheive the required conformational constraint. In this instance n can be as little as 1 residue and as high as 50. Exemplary C-[X]õ-C modified peptide sequences are shown in SEQ ID NOs: 4, 6, 8, 10, 89, 91, 93, 95, and 97. The C-[X]õ-C modified peptide sequences can be further modified, if necessary, to prevent N-terminal clipping of mature mimetibodies. For example, SEQ ID NO: 4 or SEQ ID NO: 97 can be modified to remove the N-terminal S-Y-S sequence and replace it with G-G as shown in SEQ ID NO: 282 or SEQ ID NO: 271, respectively.
The modification may also comprise the incorporation of a Wa-[X]õ-Wa motif into the peptide to conformationally constrain the peptide through the formation of a tryptophan zipper. In a Wa-[X]õ-Wa motif W is tryptophan residue, X is an amino acid, a is an integer ususlly 2, but can be from 1 to 10, and n is an integer necessary to acheive the required conformational constraint. In this instance n can be as little a 1 residue and as high as 50.
Exemplary Wa-[X]õ-Wa peptides are shown in SEQ ID NOs: 12, 14, 16 and 18. Further, the sequence HFRW (SEQ ID NO: 81) present in the alpha-MSH peptide may also be modified by substituting any residue in this sequence with any one of F, H, W and M; for example, HFRW
(SEQ ID NO: 81) can be substituted to FHWM (SEQ ID NO: 83).
In the polypeptides of the invention, the linker portion (Lk) provides structural flexibility by allowing the mimetibody to have alternative orientations and binding properties. Exemplary linkers include non-peptide chemical linkages or one to 20 amino acids linked by peptide bonds, wherein the amino acids are selected from the 20 naturally occurring amino acids or other amino acids (e.g. D-amino acids, non-naturally occurring amino acids, or rare naturally occuring amino acids). The linker portion can include a majority of amino acids that are sterically unhindered, such as glycine, alanine and serine and can include GS, poly GS (e.g. GSGS (SEQ ID NO: 20)), GGSG (SEQ ID NO: 22), GSGGGS (SEQ ID NO: 24), GSGGGSG (SEQ ID NO:
26), GSSG (SEQ ID NO: 28), GGGS (SEQ ID NO: 85), GGGGS (SEQ ID NO:
99), GGGGSGGGGS (SEQ ID NO: 101), GGGGSGGGGSGGGGS (SEQ ID NO: 103), GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 105) or any combination or polymer thereof. Other exemplary linkers within the scope of the invention may be longer than 20 residues and may include residues other than glycine, alanine and serine.
In the polypeptides of the invention, V2 is a portion of a carboxy terminal domain of an immunoglobulin variable region such as a heavy chain variable region. Exemplary V2 amino acid sequences are GTLVTVSS (SEQ ID NO: 32), TLVAVSS (SEQ ID NO: 34), and TLVTVSS
(SEQ ID NO: 249).
The (Mp-Lk- (V2) y-Hg-CH2-CH3) (t) mimetibody polypeptides of the invention may comprise "y" V2 polypeptides where y is 0 (zero) or 1 (one). The amino acid sequences shown in SEQ ID NOs: 127, 129, 132, 134, 137, 139, 142, 144, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 256, 259, 261, 264, 266, or 269 are exemplary of mimetibody polypeptides comprising one V2 polypeptide. Stated differently, these mimetibody polypeptides are examples of the formula (Mp-Lk-(V2)y-Hg-CH2-CH3) (t) where y is one. The amino acid sequences in SEQ ID NOs: 60, 62, 121, 123, 147, 149, 152, 154, 157, 159, 162, 164, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 232, 234, 237, 239, 242, 244, 251, or 253 are exemplary of mimetibody polypeptides that lack a V2 polypeptide. In other words, these are mimetibody polypeptides of the formula (Mp-Lk-(V2)y-Hg-CH2-CH3) (t) .
In the polypeptides of the invention, Hg is a portion of the hinge domain of an immunoglobulin variable region such as a heavy chain variable region. Exemplary Hg amino acid sequences include EPKSCDKTHTCPPCP (SEQ ID NO: 36), EPKSADKTHTCPPCP (SEQ ID NO: 38), ESKYGPPCPSCP (SEQ ID NO: 40), ESKYGPPCPPCP (SEQ ID NO: 42), CPPCP
(SEQ ID NO: 44) and CPSC (SEQ ID NO: 46).
Hg amino acid sequences can be modified. Such modifications can remove potential sites of 0-linked glycosylation. Such modifications can also remove cysteine residues that may cause aggregates or multimers of the polypeptides of the invention to form.
One way to minimize 0-linked glycosylation in the mimetibodies of the invention is to substitute Ala residues for Thr residues in the Hg portion of the polypeptides of the invention. The Hg amino acid sequence EPKSCDKTHACPPCP (SEQ ID NO: 107) is exemplary of such a Thr to Ala substitution; this particular Hg substitution can also be obtained by a Thr to Ala substitution at position 59 of SEQ ID
NO: 62.
One way to minimize aggregation or multimerization of the mimetibodies of the invention is to substitute Ala residues for Cys residues in the Hg portion of the polypeptides of the invention.
The Hg amino acid sequence EPKSADKTHTCPPCP (SEQ ID NO: 109) is exemplary of such a Cys to Ala substitution; this particular Hg substitution can also be obtained by a Cys to Ala substitution at position 54 of SEQ ID NO: 62.
Modifications to the Hg amino acid sequences of the mimetibody polypeptides of the invention can be made singly or in combination.
The Hg amino acid sequence EPKSADKTHACPPCP (SEQ ID NO: 111) combines both the aforementioned substitutions; and can be obtained by a Cys to Ala substitution at position 54 and a Thr to Ala substitution at position 59 of SEQ ID NO: 62. Those skilled in the art will recognize other amino acid residues that can be used to make substitutions that remove 0-glycosylation sites and aggregation or multimerization associated sites in the mimetibodies of the invention. Such sites can also be deleted by removing amino acid residues.
In the polypeptides of the invention, CH2 is an immunoglobulin heavy chain CH2 constant region. Exemplary CH2 amino acid sequences include:
APELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK (SEQ ID NO: 48), APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK (SEQ ID NO: 50), APEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAK (SEQ ID NO: 52), APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAK (SEQ ID NO: 54), APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKGLSSPIEKTISKAK (SEQ ID NO: 117), and APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK (SEQ ID NO: 246).
In the polypeptides of the invention, CH3 is an immunoglobulin heavy chain CH3 constant region. Exemplary CH3 amino acid sequences include:
GQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS
KLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 56), GQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS
RLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: 58), and GQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS
RLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: 119). It will be recognized by those skilled in the art that the CH3 region of the polypeptides of the invention may have its C-terminal amino acid cleaved off when expressed in certain recombinant systems.
In the mimetibody polypeptides of invention Hg, CH2 or CH3 may be of the IgG1 or IgG4 subclass. A sequence is of the IgG1 or IgG4 subclass if it is formed or developed from a yl or y4 heavy chain respectively. A given peptide chain is a yl or y4 heavy chain if it is at least 80% identical to a known yl or y4 heavy chain sequence of a given species. Percent identity between two peptide chains can be determined by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9Ø0 (Invitrogen Corp., Carlsbad, CA).
In the mimetibody polypeptides of the invention Hg, CH2 or CH3 may individually be of the IgG2 or IgG4 subclass. The mimetibodies of the invention may also comprise combinations of Hg, CH2 or CH3 elements from each subclass For example, Hg may be of the IgG4 subclass while CH2 and CH3 are of the IgG1 subclass. Alternatively, Hg, CH2 and CH3 may all of the IgG4 or IgG2 subclass. The polypeptide EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVE
VHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTL
PPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQ
GNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 65) is exemplary of a polypeptide in which Hg (residues 1-15 of SEQ ID NO: 65), CH2 (residues 16-125 of SEQ ID NO: 65), and CH3 (residues 126-232 of SEQ
ID NO: 65) are all of the IgG2 subclass.
The IgG1 and IgG4 subclasses differ in the number of cysteines in the hinge region. Most IgG type antibodies, such as IgG1, are homodimeric molecules made up of two identical heavy (H) chains and two identical light (L) chains, typically abbreviated H2L2. Thus, these molecules are generally bivalent with respect to antigen binding due to the formation of inter-heavy chain disulfide bonds and both antigen binding (Fab) arms of the IgG molecule have identical binding specificity. IgG4 isotype heavy chains, in contrast, contain a CPSC
(SEQ ID NO: 46) motif in their hinge regions capable of forming either inter- or intra-heavy chain disulfide bonds, i.e., the two Cys residues in the CPSC motif may disulfide bond with the corresponding Cys residues in the other H chain (inter) or the two Cys residues within a given CPSC motif may disulfide bond with each other (intra).
Since the HL pairs in those IgG4 molecules with intra-heavy chain bonds in the hinge region are not covalently associated with each other, they may dissociate into HL monomers that then reassociate with HL monomers derived from other IgG4 molecules forming bispecific, heterodimeric IgG4 molecules. In vivo isomerase enzymes may facilitate this process. In a bispecific IgG antibody the two Fab "arms" of the antibody molecule differ in the epitopes that they bind.
Substituting Ser residues in the hinge region of IgG4 with Pro results in "IgG1-like behavior," i.e., the molecules form stable disulfide bonds between heavy chains and therefore, are not susceptible to HL exchange with other IgG4 molecules.
The mimetibody polypeptides of the invention may be made more IgG4-like, or IgG2-like by the modification of sites which are involved in disulfide bond formation and are present in the Hg-CH2-CH3 portion of the mimetibody polypeptides. Such sites may be modified by removal, deletion, insertion or substitution with other amino acids. Typically, the cysteine residues present in disulfide bond associated motifs are removed or substituted. Removal of these sites may avoid covalent disulfide bonding with other cysteine-containing proteins present in the mimetibody producing host cell or intra-heavy chain disulfide bonding in IgG4-based constructs while still allowing for noncovalent dimerization of mimetibody Hg-CH2-CH3 domains. Modification of such sites can permit the formation of bispecific mimetibody polypeptides with two different Mp portions or prevent the formation of such bispecific species.
The IgG1 and IgG4 subclasses also differ in their ability to mediate complement dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). CDC is the lysing of a target cell in the presence of complement. The complement activation pathway is initiated by the binding of the first component of the complement system (Clq) to a molecule complexed with a cognate antigen. IgG1 is a strong inducer of the complement cascade and subsequent CDC activity, while IgG4 has little complement-inducing activity. ADCC is a cell-mediated process in which nonspecific cytotoxic cells that express Fc receptors (FcRs) involved in ADCC (e.g., natural killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. The IgG1 subclass binds with high affinity to Fc receptors involved in ADCC and contributes to ADCC, while IgG4 binds only weakly to such receptors and has little ADCC inducing activity. The relative inability of IgG4 to activate effector functions such as ADCC is desirable since delivery of the mimetibody polypeptide to cells without cell killing is possible.
The CDC and ADCC activity of the mimetibody polypeptides of the invention may be modified by altering sites involved in CDC and ADCC present in the Hg-CH2-CH3 portion of the mimetibody polypeptide.
Such sites may be modified by removal, deletion, insertion or substitution with other amino acids. In the mimetibodies of the invention sites involved in CDC, such as the Clq binding site, are typically removed or otherwise modified to minimize CDC activity.
Additionally, Fc receptor binding sites involved in ADCC can also be similarly modified in the mimetibodies of the invention. In general, such modification will remove Fc receptor binding sites involved in ADCC activity from the mimetibodies of the invention.
The substitution of Leu residues with Ala residues in the CH2 portion of the polypeptides of the invention is one example of a modification which can minimize ADCC activity in the polypeptides of the invention. The CH2 amino acid sequences APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK (SEQ ID NO: 52) and APEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNS
TYRVVSVLTVLHQDWLNGKEYKCKVSNKGLSSPIEKTISKAK (SEQ ID NO: 54) are exemplary of such a Leu to Ala substitution at residues 4 and 5 (in these sequences). Further, the V1 domain can be removed such that the N-terminus of the peptide is free following cleavage of the signal peptide, and is accessible to and could be modified by enzymes such as acetylases.
Antibodies of both the IgG4 and IgG1 isotypes contain FcRn salvage receptor binding sites. The FcRn salvage receptor helps maintain IgG antibody levels in the body by recycling or transporting IgG type antibodies across enodothelial cell layers such as those lining the inside of body cavities and blood vessels.
The FcRn salavage receptor does this by binding IgGs that have entered endothelial cells by nonspecific pinocytosis and preventing these IgG antibody molecules from being degraded in the lysosome of the cell. The result of such FcRn receptor activity is that the serum half-life of a molecule with an FcRn binding site is extended relative to an otherwise identical molecule lacking such a site.
Exemplary mature mimetibody polypeptides of the invention including a Ser to Pro substitution in the hinge region and CH2 domain Leu to Ala substitutions on an IgG4 backbone have the nucleic and amino acid sequences shown in SEQ ID NOs: 211-215 and 250-254.
In particular, SEQ ID NOs: 212 and 214 show the amino acid sequence of a mimetibody polypeptide having an Mp sequence as shown in SEQ ID
NO: 4 and an Lk sequence as shown in SEQ ID NO: 101 without and with a signal sequence, respectively. SEQ ID NOs: 211 and 213 show exemplary nucleic acid sequences encoding these polypeptides.
Further, SEQ ID NOs: 251 and 253 show the amino acid sequence of a mimetibody polypeptide having an Mp sequence as shown in SEQ ID NO:
282 and an Lk sequence as shown in SEQ ID NO: 101 without and with a signal sequence, respectively. SEQ ID NOs: 250 and 252 show exemplary nucleic acid sequences encoding these polypeptides.
It is desirable that the Hg-CH2-CH3 portion of the mimetibodies of the invention contain a FcRn binding site at the junction of the CH2 and CH3 regions. It is expected that such FcRn sites will increase the serum half-life of the mimetibodies of the invention as well as improve other pharmacokinetic properties relative to a melanocortin receptor binding molecule, such as alpha-MSH alone. In the mimetibodies of the invention FcRn sites may be modified or added by removal, deletion, insertion or substitution of amino acids. Typically, such modifications are used to improve the binding of a given site to the FcRn. One example of a human FcRn binding sites is the sequence MISRTPTVLHQHNHY (SEQ. ID. NO.: 69) found in both IgG1 and IgG4 antibodies. Other FcRn binding sites are well known by those skilled in the art.
Antibodies with different isotypes, such as IgG4 and IgG1, may contain glycosylation sites. Glycosylation of these sites can alter the properties and activites of antibody molecules. Antibody molecules may be N-glycosylated or 0-glycosylated. N-glycosylation of antibody amino acid residue side chains containing nitrogen atoms (e.g., Asn) can modulate antibody Fc effector functions such as ADCC
by conferring a cytolytic activity to N-glycosylated antibody molecules. This ADCC associated cytolytic activity causes the lysis of cells effected by such N-glycosylated antibodies. Alternatively, an antibody molecule may be 0-glycosylated by modification of amino acid residue side chains containing oxygen atoms (e.g., Ser or Thr).
0-glycosylation can decrease the serum half-life of an antibody molecule through increased lectin mediated clearance of 0-glycosylated antibody molecules from the serum. Additionally, 0-glycosylation can cause undesirable increases in antibody heterogeneity due to differing extents of 0-glycosylation between various antibody molecules. Lastly, both 0-glycosylation and N-glycosylation can alter the structure dependent properties of antibody molecules such as binding affinity and immunogenicity.
Like the antibody molecules they mimic, the mimetibody polypeptides of the invention may also be post-translationally modified by N-glycosylation and 0-glycosylation. In most instances, it is desirable to limit the N-glycosylation of the mimetibodies of the invention to minimize cytolytic activity. N-glycosylation can be limited by the removal or substitution of amino acid residues, such as Asn, which are typically N-glycosylated. It is also desirable to limit mimetibody 0-glycosylation to minimize lectin-mediated clearance, mimetibody heterogeneity and the alteration of structure dependent mimetibody properties such as binding affinity and immunogenicity. One way to minimize 0-linked glycosylation in the mimetibodies of the invention is to substitute Ala residues for Thr residues in the V2 portion of the polypeptides of the invention.
The V2 amino acid sequence TLVAVSS (SEQ ID NO: 34) is exemplary of such a Thr to Ala substitution; this particular V2 substitution can also be obtained by a Thr to Ala substitution at postion 47 of SEQ
ID NO: 62. Those skilled in the art also will recognize other ways to control N-linked and 0-linked glycosylation including modulation of glycosylase enzyme activity.
The monomeric structure Mp-Lk- (V2) y-Hg-CH2-CH3 of the mimetibody polypeptides of the invention can be linked to "t" other monomers where t is an integer from 1 to 10. Such linking can occur through non-covalent interactions or covalent linkages such as a Cys-Cys disulfide bond. In this way multimeric structures such as dimers and higher order multimers of the polypeptides of the invention can be formed. It is expected that dimerization of the polypeptides of the invention will increase the affinity of these polypeptides to melanocortin receptors such as MC4R. The term "multimers" as used herein means molecules that have quaternary structure and are formed by the association of two or more subunits.
The polypeptides of the invention can optionally comprise at the amino terminus, an amino terminal portion of an immunoglobulin variable region, designated V1 as shown in Formula II:
(V1-Mp-Lk- (V2) v-Hg-CH2-CH3) (t) (II) Exemplary V1 amino acid sequences include QIQ, QVQ, QIQGG (SEQ ID
NO: 113), and QIQGGGG (SEQ ID NO: 115).
The polypeptides of the invention may also comprise secretory signals necessary to facilitate protein secretion or other signals necessary for protein trafficking in the cell. An exemplary secretory signal sequence is MAWVWTLLFLMAAAQSIQA (SEQ ID NO: 69).
Those skilled in the art will recognize other secretory signals.
In one embodiment the polypeptides of the invention comprise polypeptides having the sequences shown in SEQ ID NO: 60, 62, 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268. These sequences exemplify melanocortin receptor binding alpha-MSH polypetides either having, or lacking, an amino terminal secretory signal sequence. SEQ ID NO: 62 represents a (V1-Mp-Lk-(V2)y-Hg-CH2-CH3) (t) melanocortin receptor binding alpha-MSH polypetide of generic formula (II) which has the secretory signal MAWVWTLLFLMAAAQSIQA (SEQ ID NO: 69) fused to its amino terminus. SEQ ID NO: 60 represents a (Mp-Lk- (V2) y-Hg-CH2-CH3) (t) melanocortin receptor binding alpha-MSH polypetide of generic formula (I). No secretory signal is present in SEQ ID NO: 60.
Another aspect of the present invention is a polynucleotide comprising, complementary to or having significant identity with, a polynucleotide encoding at least one melanocortin receptor binding mimetibody. Other aspects of the present invention include vectors comprising at least one polynucleotide molecule encoding a melanocortin receptor binding mimetibody. In a different aspect the invention provides a cell comprising a vector of the invention or a cell expressing a mimetibody polypeptide of the invention. The polynucleotides, vectors and cells may be used to produce the mimetibody polypeptides of the invention.
In one embodiment, the polynucleotides of the invention comprise a polynucleotide having the sequence shown in SEQ ID NO:
59, 61, 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267 or a polynucleotide having a sequence complementary to the sequence shown in SEQ ID NO: 59, 61, 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267. SEQ ID NO: 59 is a cDNA encoding a (Mp-Lk- (V2) y-Hg-CH2-CH3) (t) melanocortin receptor binding alpha-MSH
polypetide of generic formula (I) which lacks a signal sequence.
SEQ ID NO: 61 is a cDNA encoding a (V1-Mp-Lk- (V2) Y-Hg-CH2-CH3) (t) melanocortin receptor binding alpha-MSH polypetide of generic formula (II) which has a secretory signal fused to its amino terminus.
In one embodiment, the polynucleotides of the invention comprise a polynucleotide encoding the polypeptide having the sequence shown in SEQ ID NO: 60, 62, 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268.
Exemplary nucleic acid sequences that encode the polypeptide sequences shown in SEQ ID NO 60, 62, 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268 are shown in SEQ ID NO: 59, 61, 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267, respectively.
Also provided are polynucleotides encoding polypeptides that are substantially identical to the polypeptides of the invention.
The term "substantially identical" in the context of polypeptides means that a given polypeptide sequence is identical to a polypeptide sequence of the invention, in particular the V1-Mp-Lk-V2-Hg region, in at least 50% or at least about 60% or at least about 70% or at least about 80% or at least about 90% or at least about 95-98% of the amino acid residues. Percent identity between two polypeptide sequences can be determined by pairwise alignment using the default settings of the AlignX module of Vector NTI
v.9Ø0 (Invitrogen Corp., Carlsbad, CA). Those skilled in the art would recognize polynucleotide sequences which would encode the above-described polypeptides.
Typically, the polynucleotides of the invention are used in expression vectors for the preparation of the mimetibody polypeptides of the invention. Vectors within the scope of the invention provide necessary elements for eukaryotic expression and include viral promoter driven vectors, such as CMV promoter driven vectors, e.g., pcDNA3.1, pCEP4, and their derivatives, Baculovirus expression vectors, Drosophila expression vectors, and expression vectors that are driven by mammalian gene promoters, such as human Ig gene promoters. Other examples include prokaryotic expression vectors, such as T7 promoter driven vectors, e.g. pET41, lactose promoter driven vectors and arabinose gene promoter driven vectors.
The present invention also relates to a cell that expresses a mimetibody of the invention or comprises a vector of the invention.
Open reading frames encoding the mimetibody polypeptides of the invention can be identified by translation of the positive strand reading frame beginning with nucleotide residue 1601 of SEQ ID NOs:
63, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 254, 259, 264, or 269. The various signal peptide, V1, Mp, V2, Hg, CH2, and CH3 portions of the mimetibody polypeptides of the invention, which have been exemplified herein, are also present in these open reading frames and can be identified using standard sequence analysis methods, such as multiple sequence alignment or other methods well know in the art. Such a cell can be prokaryotic or eukaryotic. Exemplary eukaryotic cells are mammalian cells, such as but not limited to, COS-1, COS-7, HEK293, BHK21, CHO, BSC-1, HepG2, 653, SP2/0, NSO, 293, HeLa, myeloma, lymphoma cells or any derivative thereof. Most preferably, the eukaryotic cell is a HEK293, NSO, SP2/0, or CHO cell. E. coli is an exemplary prokaryotic cell. A cell according to the invention may be generated by transfection, cell fusion, immortalization, or other procedures that are well known in the art. Polynucleotides transfected into a cell may be extrachromasomal or stably integrated into the chromosome of the cell.
The mimetibodies of the invention can be made more compatible with a given host cell by modification of the Hg-CH2-CH3 portion of the polypeptide. For example, when a mimetibody of the invention is expressed recombinantly in a bacterial cell such as E. coli, the Pro-Ala sequence in the Hg element may be removed to prevent digestion by the E. coli enzyme proline iminopeptidase. Similarly, a portion of the Hg element can be deleted or substituted with other amino acids in the mimetibodies of the invention to prevent heterogeneity in the products expressed in a selected host cell.
The present invention further provides a method to produce a mimetibody polypeptide comprising the steps of culturing a cell of the invention and purifying an expressed mimetibody polypeptide of the invention. Cell components, such as those necessary for in vitro transcription and translation, may also be used to express the polypeptides of the invention. The present invention encompasses mimetibodies produced by both methods. Expressed mimetibody polypeptides can be recovered and purified from cells or cell component based systems by methods well known in the art including, but not limited to, protein A purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylatpatite chromatography and lectin chromatography. High performance liquid chroatography (HPLC) can also be employed for purification. Typically purfication will require a combination of several different methods.
Another aspect of the present invention is a pharmaceutical composition comprising an effective amount of at least one mimetibody polypeptide and a pharmaceutically acceptable carrier or diluent. The term "effective amount" generally refers to the quantity of mimetibody necessary for effective therapy, i.e., the partial or complete alleviation of the symptom or disorder for which treatment was sought. The composition can optionally comprise at least one further compound, protein or composition useful for treating obesity and the other conditions described below. The parmaceutically acceptable carrier or diluent in the compositions can be a solution, suspension, emulsion, colloid or powder. Those skilled in the art will recognize other parmaceutically acceptable carriers and diluents.
Another aspect of the present invention is a method of modifying the biological activity of a melanocortin receptor in a cell, tissue or organ comprising contacting the pharmaceutical compositions of the invention with the cell, tissue or organ. The method may be used to modify melanocortin receptor activity in the brain, brain tissue, or brain cells. Alternatively, the method of the invention may be used to modify melanocortin receptor activity in other peripheral cells or tissues such as muscle, or other organs such as the stomach. Those skilled in the art will recognize other cells, tissues or organs, which may be used.
Another aspect of the invention is a method of modulating at least one melanocortin receptor-mediated condition comprising administering a pharmaceutical composition of the invention to a patient in need thereof. The pharmaceutical compositions of the invention can be administered by any suitable route. Such routes may be intrathecal, intranasal, peripheral (e.g., subcutaneous, intramuscular, intradermal, intravenous) or by any other means known in the art. As described previously, abnormal melanocortin receptor activity has been implicated in a number of pathological conditions, such as obesity and Type 2 diabetes. Stimulation of MC4R can cause weight loss while inhibition may cause weight gain. The mimetibody polypeptides of the invention may be also be used to modulate other melanocortin receptor mediated conditions such as male and female erectile dysfunction, inflammation, congestive heart failure, central nervous system disorders, nerve damage, infectious disease, pulmonary disease, skin disease, fever and pain.
The present invention is further described with reference to the following examples. These examples are merely to illustrate aspects of the present invention and are not intended as limitations of this invention.
Example 1 Alpha-MSH Mimetibody and Expression Vector Construction An alpha-MSH mimetibody protein comprising a secretory signal sequence, an alpha-MSH peptide sequence, a linker sequence, VH
sequence, a hinge sequence, a human IgG1 CH2 sequence and a human IgG1 CH3 sequence was designed (Fig. 3 and SEQ ID NO. 62) Analytical data, e.g., mass spectroscopy, has confirmed that a mature polypeptide is generated (61,344.6 for G1/G1 form). Nucleic acid sequences encoding this alpha-MSH mimetibody protein (Fig. 3; SEQ ID
NO: 61) were generated using standard molecular biology techniques.
Nucleic acid sequences encoding the alpha-MSH mimetibody sequence were subcloned into the p2389 expression vector to generate an alpha-MSH mimetibody expression vector (SEQ ID NO: 63).
Example 2 Alpha-MSH Mimetibody Expression The alpha-MSH mimetibody was transiently expressed in HEK293E
cells. Cells were cultured using standard conditions and transiently transfected with the alpha-MSH mimetibody expression vector using Lipofectamine 2000 (Invitrogen, Carlsbad, CA) as directed by the manufacturer. 24 h after transfection cells were transferred to a serum free media formulation and cultured for 5 days. The culture media was then removed and centrifuged to remove debris. Clarified media was incubated with Protein A-SepharoseTM
(HiTrap rProtein A FF, Amersham Biosciencies, Piscataway, NJ) and proteins were eluted from the Protein A-SepharoseTM conjugate as directed by the manufacturer. The eluted protein solution was then further purified via SuperoseTM 12 size exclusion chromatography (Superose 12 10/300 GL, Amersham Biosciencies, Piscataway, NJ) using standard methods. Column eluant was then subjected to SDS-PAGE and visualized by silver and Coomassie blue staining. Western blots were then prepared and the blots were probed with either an Fc specific primary antibody or an alpha-MSH specific primary antibody.
Together, the Western Blot and SDS-PAGE staining results indicated that a purified alpha-MSH mimetibody, composed of two polypeptide chains, had been obtained from the transiently transfected HEK293 cells.
Example 3 Alpha-MSH Mimetibody Binds MC4R
The alpha-MSH mimetibody binds to MC4R and can compete with radiolabeled [Nle(4), D-Phe(7)]-alpha-MSH (NDP-alpha-MSH) agonist molecules for MC4R binding (Fig. 4). MC4R is a receptor for alpha-MSH. alpha-MSH binding to recombinantly expressed MC4R in HEK293 cell membranes (Perkin Elmer Life and Analytical Sciences, Boston, MA) was examined by competive binding assays in which increasing amounts of unlabeled MC4R agonists (positive controls) and the Fc domain of a human antibody (negative control) were added to assay cocktails containing [125I]-NDP-alpha-MSH as indicated in Fig. 4.
The unlabeled MC4R agonists were melanotan II (MTII; an alpha MSH
analog), alpha-MSH, and NDP-alpha-MSH. Alpha-MSH mimetibody binding to MC4R was stable after two weeks of storage at 4 C, -20 C, and -80 C in PBS (phosphate buffered saline) as assessed by competive binding assays.
Competivive binding assays were performed using Scintillation Proximity Assays (Amersham Biosciences Corp, Piscataway, NJ) as directed by the assay manufacturer. Assay cocktails contained [1251]-NDP-alpha-MSH at EC80, i.e., -0.5 nM, 0.1 pg of MC4R
membranes, 1 mM MgS04, 1.5 mM CaC12, 25 mM Hepes, 0.2% BSA, 1 mM
1,10-phenthroline, an assay manufacturer recommended quantity of protease inhibitor cocktail (Roche Diagnostics Corp., Indianapolis, IN) and SPA beads. Light emission from Scintillation Proximity Assay beads was measured with a Packard Top Count NXT Instrument (Perkin Elmer Life and Analytical Sciences, Boston, MA) for 5 minutes.
Example 4 Alpha-MSH Mimetibody Activates MC4R
The alpha-MSH mimetibody can activate MC4R signalling to increase cAMP production in CHOK1 cells expressing MC4R (Fig. 5 and Fig. 6). MC4R is a seven transmembrane (7TM) G-protein coupled receptor. Activation of MC4R by ligand or agonist results in an increase in cyclic AMP levels (cAMP).
MC4R receptor activation assays were performed using two different clonal CHOK1 cell lines stably transfected with a MC4R
expression vector and expressing MC4R. Clone 1 (Fig. 5) expressed MC4R at high levels relative to Clone 2 (Fig. 6). Clone 1 and Clone 2 cells were grown as a monolayer using standard culture conditions to a density of approximately 100,000 cells/well and then incubated with increasing amounts (0-100 pM) of alpha-MSH, MTII, or alpha-MSH
mimetibody for 15 minutes as indicated in Fig. 5 and Fig. 6. Cells were then lysed and cAMP assays were performed using the cAMP-Screen Direct'' Chemiluminescent Immunoassay System (Applied Biosystems, Foster City, CA) as directed by the manufacturer. EC50 values from cAMP assays using Clone 1 (Fig. 5) and Clone 2 (Fig. 6) are listed in Table 1 below Table 1 Clone 1 Clone 2 alpha-MSH peptide EC50 = 3.29 nM EC50 = 9.46 nM
(Positive control) MT II EC50 = 0.52 nM EC50 = 0.52 nM
(Positive control) alpha-MSH mimetibody EC50 = 14.36 nM EC50 = 52.4 nM
Example 5 Alpha-MSH Mimetibody Administration Decreases Animal Food Intake and Body Weight Alpha-MSH mimetibody administration to Rattus norvegicus brain ventricules decreases animal food intake (Fig. 7) and body weight (Fig. 8). Alpha-MSH mimetibody was supplied to brain ventricules by intracerebroventricular injections (ICV) via a cannula surgically inserted into the left lateral brain ventricle.
Cannulae were surgically inserted into male Sprague-Dawley or Wistar rats weighing 250 g to 350 g. Cannula placement coordinates were as follows: -0.8 mm from bregma, -4.5 mm ventral and -1.5 posterior-anterior. Animals recovered for 7 to 10 days after surgery. Animals were acclimatized to the experimental procedures by both daily handling and mock injection, in order to minimize stress. In addition animals were submitted to the reversal of dark-light cycle.
Proper cannula placement was confirmed by an angiotensin II
test. The test confirmed proper cannula placement if the ICV
administration of 10 ng of angiotensin II via the cannula caused the rats to drink 5-10 ml of water in 30 minutes. Only animals that passed this angiotensin II test were used in food intake experiments.
Animals were fasted for 18-24 hours and alpha-MSH mimetibody, alpha-MSH (positive control), or PBS (negative control) were then administered to the brain ventricles via the cannula at an injection rate of 9 pl/min. Each treatment group had a minimum of 7 animals.
Treatments and dosages were as indicated in Fig. 7 and Fig. 8.
Food and water was given to the animals after injection. The amount of food and water consumed was measured at 0 h, 4 h, 24 h, 48 h and 72 h (Fig. 7) after injection. Body weight at 72 hours post injection was measured as ahown in Fig. 8.
The present invention now being fully described, it will be apparent to one of ordinary skill in the art that many changes and modifications can be made thereto without departing from the spirit or scope of the appended claims.
Claims (20)
1. A polypeptide according to formula (I):
(Mp-Lk-(V2)y-Hg-C H2-C H3)(t) (I) where Mp is a biologically active melanocortin receptor binding molecule fragment of SEQ ID NO: 87, 89, 91, 93, 95, 97 or 282, Lk is a polypeptide or chemical linkage, V2 is a portion of a C-terminus of an immunoglobulin variable region, Hg is at least a portion of an immunoglobulin variable hinge region, C H2 is an immunoglobulin heavy chain C H2 constant region and C H3 is an immunoglobulin heavy chain C H3 constant region, y is 0 or 1, and t is independently an integer from 1 to 10.
(Mp-Lk-(V2)y-Hg-C H2-C H3)(t) (I) where Mp is a biologically active melanocortin receptor binding molecule fragment of SEQ ID NO: 87, 89, 91, 93, 95, 97 or 282, Lk is a polypeptide or chemical linkage, V2 is a portion of a C-terminus of an immunoglobulin variable region, Hg is at least a portion of an immunoglobulin variable hinge region, C H2 is an immunoglobulin heavy chain C H2 constant region and C H3 is an immunoglobulin heavy chain C H3 constant region, y is 0 or 1, and t is independently an integer from 1 to 10.
2. The polypeptide of claim 1 wherein Mp has the amino acid sequence shown in SEQ ID NO: 87, 89, 91, 93, 95, 97 or 282.
3. The polypeptide of claim 1 wherein the polypeptide binds to at least one melanocortin receptor.
4. The polypeptide of claim 3 wherein the melanocortin receptor is a melanocortin 4 receptor.
5. A polypeptide comprising a polypeptide having the sequence shown in SEQ ID NO: 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268.
6. A polypeptide having the sequence shown in SEQ ID NO: 212.
7. A polynucleotide encoding a polypeptide according to any one of claims 1 to 6.
8. A polynucleotide comprising a polynucleotide having the sequence shown in SEQ ID NO: 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267 or a polynucleotide having a sequence complementary to the sequence shown in SEQ ID NO: 120, 122, 126, 128, 131, 133, 136, 138, 141, 143, 146, 148, 151, 153, 156, 158, 161, 163, 166, 168, 171, 173, 176, 178, 181, 183, 186, 188, 191, 193, 196, 198, 201, 203, 206, 208, 211, 213, 216, 218, 221, 223, 226, 228, 231, 233, 236, 238, 241, 243, 250, 252, 255, 257, 260, 262, 265, or 267.
9. A polynucleotide comprising a polynucleotide encoding the polypeptide having the sequence shown in SEQ ID NO: 121, 123, 127, 129, 132, 134, 137, 139, 142, 144, 147, 149, 152, 154, 157, 159, 162, 164, 167, 169, 172, 174, 177, 179, 182, 184, 187, 189, 192, 194, 197, 199, 202, 204, 207, 209, 212, 214, 217, 219, 222, 224, 227, 229, 232, 234, 237, 239, 242, 244, 251, 253, 256, 258, 261, 263, 266, or 268.
10. A vector comprising the polynucleotide of claim 8 or 9.
11. The vector of claim 10 comprising a polynucleotide having the sequence shown in SEQ ID NO: 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 254, 259, 264, or 269.
12. A cell expressing a polypeptide according to any one of claims 1 to 6.
13. A cell comprising the vector of claim 10.
14. The cell of claim 13 wherein the cell is a Chinese hamster ovary (CHO)-derived cell.
15. A method to produce a polypeptide comprising the steps of culturing the cell of claim 13 and purifying the expressed polypeptide.
16. A pharmaceutical composition comprising an effective amount of at least one polypeptide according to any one of claims 1 to 6 and a pharmaceutically acceptable carrier or diluent.
17. A method of modifying the biological activity of a melanocortin receptor in a cell, tissue or organ comprising contacting the pharmaceutical composition of claim 16 with the cell, tissue or organ.
18. A method of modulating at least one melanocortin receptor mediated condition comprising administering the pharmaceutical composition of claim 16 to a patient in need thereof.
19. The method of claim 18 wherein the melanocortin receptor mediated condition is obesity, male erectile dysfunction, female sexual dysfunction, an inflammatory condition or fibrosis.
20. The method of claim 19 wherein the inflammatory condition is allergic inflammation, gouty arthritis, rheumatoid arthritis, inflammatory bowel disease, post-ischemia renal injury, liver inflammation, ischemic brain damage or peripheral neuropathies.
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/US2008/081734 WO2009134281A2 (en) | 2007-10-30 | 2008-10-30 | Melanocortin receptor binding mimetibodies, compositions, methods and uses |
Publications (1)
Publication Number | Publication Date |
---|---|
CA2704446A1 true CA2704446A1 (en) | 2009-11-05 |
Family
ID=42286672
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CA2704446A Abandoned CA2704446A1 (en) | 2008-10-30 | 2008-10-30 | Melanocortin receptor binding mimetibodies, compositions, methods and uses |
Country Status (1)
Country | Link |
---|---|
CA (1) | CA2704446A1 (en) |
-
2008
- 2008-10-30 CA CA2704446A patent/CA2704446A1/en not_active Abandoned
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2008355596B2 (en) | Melanocortin receptor binding mimetibodies, compositions, methods and uses | |
EP1812046B1 (en) | Melanocortin receptor binding mimetibodies, compositions, methods and uses | |
US8043616B2 (en) | sRAGE mimetibody, compositions, methods and uses | |
US8722615B2 (en) | Compositions and methods for increasing serum half-life | |
EP3060235B1 (en) | Endoglin peptides to treat fibrotic diseases | |
JP5751641B2 (en) | Melanocortin receptor binding conjugate | |
US20230045794A1 (en) | Nk cell-directed chimeric proteins | |
EP1792626B1 (en) | Treatment of obesity with alpha-MSH mimetibodies | |
WO2006138610A2 (en) | Interferon-alpha/beta binding fusion proteins and therapeutic uses thereof | |
CA2704446A1 (en) | Melanocortin receptor binding mimetibodies, compositions, methods and uses | |
WO2023125881A1 (en) | Fusion protein of glp-1 and gdf15 and use thereof | |
WO2023025123A1 (en) | Gdf15 fusion protein and use thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
EEER | Examination request |
Effective date: 20131028 |
|
FZDE | Discontinued |
Effective date: 20180523 |