WO2021016451A1 - Cd63 agonist-related methods and compositions - Google Patents
Cd63 agonist-related methods and compositions Download PDFInfo
- Publication number
- WO2021016451A1 WO2021016451A1 PCT/US2020/043264 US2020043264W WO2021016451A1 WO 2021016451 A1 WO2021016451 A1 WO 2021016451A1 US 2020043264 W US2020043264 W US 2020043264W WO 2021016451 A1 WO2021016451 A1 WO 2021016451A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cell
- agonist
- inhibitor
- kit
- pharmaceutical composition
- Prior art date
Links
- 239000000556 agonist Substances 0.000 title claims abstract description 177
- 238000000034 method Methods 0.000 title claims abstract description 90
- 239000000203 mixture Substances 0.000 title description 35
- 101150001853 CD63 gene Proteins 0.000 title 1
- 101000934368 Homo sapiens CD63 antigen Proteins 0.000 claims abstract description 234
- 102100025222 CD63 antigen Human genes 0.000 claims abstract description 232
- 210000004027 cell Anatomy 0.000 claims abstract description 109
- 229940126530 T cell activator Drugs 0.000 claims abstract description 102
- 230000002062 proliferating effect Effects 0.000 claims abstract description 85
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 55
- 230000005867 T cell response Effects 0.000 claims abstract description 11
- 239000003795 chemical substances by application Substances 0.000 claims description 90
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 71
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 57
- 239000003112 inhibitor Substances 0.000 claims description 49
- 206010028980 Neoplasm Diseases 0.000 claims description 45
- 108090000695 Cytokines Proteins 0.000 claims description 36
- 102000004127 Cytokines Human genes 0.000 claims description 36
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 29
- 201000011510 cancer Diseases 0.000 claims description 28
- 230000002401 inhibitory effect Effects 0.000 claims description 23
- 230000006044 T cell activation Effects 0.000 claims description 22
- 238000002560 therapeutic procedure Methods 0.000 claims description 22
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 claims description 21
- 102000037984 Inhibitory immune checkpoint proteins Human genes 0.000 claims description 21
- 108091008026 Inhibitory immune checkpoint proteins Proteins 0.000 claims description 21
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 claims description 21
- 239000005557 antagonist Substances 0.000 claims description 18
- 150000003384 small molecules Chemical group 0.000 claims description 17
- 101001137987 Homo sapiens Lymphocyte activation gene 3 protein Proteins 0.000 claims description 15
- 102100020862 Lymphocyte activation gene 3 protein Human genes 0.000 claims description 15
- 239000003446 ligand Substances 0.000 claims description 15
- 229920001184 polypeptide Polymers 0.000 claims description 15
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 15
- 102000008203 CTLA-4 Antigen Human genes 0.000 claims description 14
- 108010021064 CTLA-4 Antigen Proteins 0.000 claims description 14
- 230000014509 gene expression Effects 0.000 claims description 14
- 108090000623 proteins and genes Proteins 0.000 claims description 13
- 102000050627 Glucocorticoid-Induced TNFR-Related Human genes 0.000 claims description 12
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 claims description 12
- 101710187882 Tumor necrosis factor receptor superfamily member 18 Proteins 0.000 claims description 12
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 claims description 12
- 102000027596 immune receptors Human genes 0.000 claims description 12
- 108091008915 immune receptors Proteins 0.000 claims description 12
- 102100024834 T-cell immunoreceptor with Ig and ITIM domains Human genes 0.000 claims description 11
- 101710090983 T-cell immunoreceptor with Ig and ITIM domains Proteins 0.000 claims description 11
- 102000004169 proteins and genes Human genes 0.000 claims description 11
- 108060003951 Immunoglobulin Proteins 0.000 claims description 10
- 102000018358 immunoglobulin Human genes 0.000 claims description 10
- 229940121655 pd-1 inhibitor Drugs 0.000 claims description 10
- 230000006028 immune-suppresssive effect Effects 0.000 claims description 9
- 108091008034 costimulatory receptors Proteins 0.000 claims description 8
- 229940121656 pd-l1 inhibitor Drugs 0.000 claims description 8
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 8
- IHWDSEPNZDYMNF-UHFFFAOYSA-N 1H-indol-2-amine Chemical compound C1=CC=C2NC(N)=CC2=C1 IHWDSEPNZDYMNF-UHFFFAOYSA-N 0.000 claims description 7
- 229940116741 CD137 agonist Drugs 0.000 claims description 7
- 229940123189 CD40 agonist Drugs 0.000 claims description 7
- 230000003389 potentiating effect Effects 0.000 claims description 7
- 102100038078 CD276 antigen Human genes 0.000 claims description 6
- 101710185679 CD276 antigen Proteins 0.000 claims description 6
- 108091006082 receptor inhibitors Proteins 0.000 claims description 6
- 239000003862 glucocorticoid Substances 0.000 claims description 2
- 102000016680 Dioxygenases Human genes 0.000 claims 1
- 108010028143 Dioxygenases Proteins 0.000 claims 1
- 208000035475 disorder Diseases 0.000 description 41
- 238000009739 binding Methods 0.000 description 24
- 230000027455 binding Effects 0.000 description 23
- 210000005007 innate immune system Anatomy 0.000 description 19
- 230000000284 resting effect Effects 0.000 description 14
- 239000000427 antigen Substances 0.000 description 13
- 108091007433 antigens Proteins 0.000 description 12
- 102000036639 antigens Human genes 0.000 description 12
- 235000018102 proteins Nutrition 0.000 description 10
- 238000000684 flow cytometry Methods 0.000 description 9
- 238000009472 formulation Methods 0.000 description 9
- 230000000694 effects Effects 0.000 description 8
- 239000012634 fragment Substances 0.000 description 8
- 238000004519 manufacturing process Methods 0.000 description 8
- 241000699666 Mus <mouse, genus> Species 0.000 description 7
- 230000004913 activation Effects 0.000 description 7
- 238000003556 assay Methods 0.000 description 7
- 239000000872 buffer Substances 0.000 description 7
- 230000000638 stimulation Effects 0.000 description 7
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 6
- 241000699670 Mus sp. Species 0.000 description 6
- 230000000139 costimulatory effect Effects 0.000 description 6
- 238000001727 in vivo Methods 0.000 description 6
- 230000000670 limiting effect Effects 0.000 description 6
- 238000002965 ELISA Methods 0.000 description 5
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 5
- 108010002350 Interleukin-2 Proteins 0.000 description 5
- 239000002202 Polyethylene glycol Substances 0.000 description 5
- 108091008874 T cell receptors Proteins 0.000 description 5
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 5
- 235000001014 amino acid Nutrition 0.000 description 5
- 150000001413 amino acids Chemical class 0.000 description 5
- 210000000612 antigen-presenting cell Anatomy 0.000 description 5
- 230000004663 cell proliferation Effects 0.000 description 5
- 150000001875 compounds Chemical class 0.000 description 5
- 230000028993 immune response Effects 0.000 description 5
- 210000000987 immune system Anatomy 0.000 description 5
- 230000001965 increasing effect Effects 0.000 description 5
- 229920001223 polyethylene glycol Polymers 0.000 description 5
- 239000003755 preservative agent Substances 0.000 description 5
- 238000012216 screening Methods 0.000 description 5
- 239000004094 surface-active agent Substances 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 5
- 229940046168 CpG oligodeoxynucleotide Drugs 0.000 description 4
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 4
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 4
- -1 TIM-3 Proteins 0.000 description 4
- 229940024606 amino acid Drugs 0.000 description 4
- 239000011230 binding agent Substances 0.000 description 4
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 4
- 230000001747 exhibiting effect Effects 0.000 description 4
- 230000003993 interaction Effects 0.000 description 4
- RLSSMJSEOOYNOY-UHFFFAOYSA-N m-cresol Chemical compound CC1=CC=CC(O)=C1 RLSSMJSEOOYNOY-UHFFFAOYSA-N 0.000 description 4
- 230000002018 overexpression Effects 0.000 description 4
- 238000002823 phage display Methods 0.000 description 4
- 229920001983 poloxamer Polymers 0.000 description 4
- 229920000136 polysorbate Polymers 0.000 description 4
- 230000035755 proliferation Effects 0.000 description 4
- 239000007787 solid Substances 0.000 description 4
- 239000000243 solution Substances 0.000 description 4
- 239000003381 stabilizer Substances 0.000 description 4
- 230000004936 stimulating effect Effects 0.000 description 4
- 108020000946 Bacterial DNA Proteins 0.000 description 3
- 229920002261 Corn starch Polymers 0.000 description 3
- 108020004414 DNA Proteins 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 102100039364 Metalloproteinase inhibitor 1 Human genes 0.000 description 3
- 206010033128 Ovarian cancer Diseases 0.000 description 3
- 206010061535 Ovarian neoplasm Diseases 0.000 description 3
- 239000012270 PD-1 inhibitor Substances 0.000 description 3
- 239000012668 PD-1-inhibitor Substances 0.000 description 3
- 239000002033 PVDF binder Substances 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 108020000411 Toll-like receptor Proteins 0.000 description 3
- 102000002689 Toll-like receptor Human genes 0.000 description 3
- 239000000654 additive Substances 0.000 description 3
- 230000000903 blocking effect Effects 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 239000000969 carrier Substances 0.000 description 3
- 239000008120 corn starch Substances 0.000 description 3
- 230000002596 correlated effect Effects 0.000 description 3
- 239000003814 drug Substances 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 239000008187 granular material Substances 0.000 description 3
- 201000010536 head and neck cancer Diseases 0.000 description 3
- 208000014829 head and neck neoplasm Diseases 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 102000006495 integrins Human genes 0.000 description 3
- 108010044426 integrins Proteins 0.000 description 3
- 238000002372 labelling Methods 0.000 description 3
- 210000000265 leukocyte Anatomy 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 210000004379 membrane Anatomy 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 3
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 238000000159 protein binding assay Methods 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 239000000758 substrate Substances 0.000 description 3
- 235000000346 sugar Nutrition 0.000 description 3
- 150000008163 sugars Chemical class 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 description 3
- OZFAFGSSMRRTDW-UHFFFAOYSA-N (2,4-dichlorophenyl) benzenesulfonate Chemical compound ClC1=CC(Cl)=CC=C1OS(=O)(=O)C1=CC=CC=C1 OZFAFGSSMRRTDW-UHFFFAOYSA-N 0.000 description 2
- CFKMVGJGLGKFKI-UHFFFAOYSA-N 4-chloro-m-cresol Chemical compound CC1=CC(O)=CC=C1Cl CFKMVGJGLGKFKI-UHFFFAOYSA-N 0.000 description 2
- CDEURGJCGCHYFH-UHFFFAOYSA-N 5-ethynyl-1-[4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]pyrimidine-2,4-dione Chemical compound C1C(O)C(CO)OC1N1C(=O)NC(=O)C(C#C)=C1 CDEURGJCGCHYFH-UHFFFAOYSA-N 0.000 description 2
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 206010005003 Bladder cancer Diseases 0.000 description 2
- 201000009030 Carcinoma Diseases 0.000 description 2
- 206010009944 Colon cancer Diseases 0.000 description 2
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 2
- 239000012591 Dulbecco’s Phosphate Buffered Saline Substances 0.000 description 2
- 238000011510 Elispot assay Methods 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 2
- 239000004471 Glycine Substances 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101000935043 Homo sapiens Integrin beta-1 Proteins 0.000 description 2
- 102100025304 Integrin beta-1 Human genes 0.000 description 2
- 108010002352 Interleukin-1 Proteins 0.000 description 2
- 108090000978 Interleukin-4 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
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 2
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 2
- 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 2
- 229930195725 Mannitol Natural products 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- RVGRUAULSDPKGF-UHFFFAOYSA-N Poloxamer Chemical compound C1CO1.CC1CO1 RVGRUAULSDPKGF-UHFFFAOYSA-N 0.000 description 2
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 2
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M Potassium chloride Chemical compound [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 2
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 2
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 2
- 208000006265 Renal cell carcinoma Diseases 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 2
- 208000000102 Squamous Cell Carcinoma of Head and Neck Diseases 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 229930006000 Sucrose Natural products 0.000 description 2
- 230000006052 T cell proliferation Effects 0.000 description 2
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 2
- 235000004279 alanine Nutrition 0.000 description 2
- 125000001931 aliphatic group Chemical group 0.000 description 2
- 150000005215 alkyl ethers Chemical class 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 239000013011 aqueous formulation Substances 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 229960000686 benzalkonium chloride Drugs 0.000 description 2
- CADWTSSKOVRVJC-UHFFFAOYSA-N benzyl(dimethyl)azanium;chloride Chemical compound [Cl-].C[NH+](C)CC1=CC=CC=C1 CADWTSSKOVRVJC-UHFFFAOYSA-N 0.000 description 2
- MSWZFWKMSRAUBD-UHFFFAOYSA-N beta-D-galactosamine Natural products NC1C(O)OC(CO)C(O)C1O MSWZFWKMSRAUBD-UHFFFAOYSA-N 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 239000001913 cellulose Substances 0.000 description 2
- 229920002678 cellulose Polymers 0.000 description 2
- 230000001472 cytotoxic effect Effects 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 239000001177 diphosphate Substances 0.000 description 2
- 231100000673 dose–response relationship Toxicity 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 239000000194 fatty acid Substances 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- 229920000159 gelatin Polymers 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 235000013922 glutamic acid Nutrition 0.000 description 2
- 239000004220 glutamic acid Substances 0.000 description 2
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 2
- 235000011187 glycerol Nutrition 0.000 description 2
- 210000003714 granulocyte Anatomy 0.000 description 2
- 201000000459 head and neck squamous cell carcinoma Diseases 0.000 description 2
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 2
- 102000057640 human CD63 Human genes 0.000 description 2
- 210000004408 hybridoma Anatomy 0.000 description 2
- 229940126533 immune checkpoint blocker Drugs 0.000 description 2
- 238000001361 intraarterial administration Methods 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 239000008101 lactose Substances 0.000 description 2
- 208000014018 liver neoplasm Diseases 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 239000000594 mannitol Substances 0.000 description 2
- 235000010355 mannitol Nutrition 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 229930182817 methionine Natural products 0.000 description 2
- 235000006109 methionine Nutrition 0.000 description 2
- 229960004452 methionine Drugs 0.000 description 2
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 2
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 2
- 229920002113 octoxynol Polymers 0.000 description 2
- 229940046166 oligodeoxynucleotide Drugs 0.000 description 2
- 150000007524 organic acids Chemical class 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- WVDDGKGOMKODPV-ZQBYOMGUSA-N phenyl(114C)methanol Chemical compound O[14CH2]C1=CC=CC=C1 WVDDGKGOMKODPV-ZQBYOMGUSA-N 0.000 description 2
- 230000036470 plasma concentration Effects 0.000 description 2
- 229960000502 poloxamer Drugs 0.000 description 2
- 229920001592 potato starch Polymers 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 235000010232 propyl p-hydroxybenzoate Nutrition 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 235000002639 sodium chloride Nutrition 0.000 description 2
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 2
- 230000009870 specific binding Effects 0.000 description 2
- 206010041823 squamous cell carcinoma Diseases 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 238000003860 storage Methods 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 2
- 239000003826 tablet Substances 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 210000001519 tissue Anatomy 0.000 description 2
- 239000012929 tonicity agent Substances 0.000 description 2
- 201000005112 urinary bladder cancer Diseases 0.000 description 2
- HDTRYLNUVZCQOY-UHFFFAOYSA-N α-D-glucopyranosyl-α-D-glucopyranoside Natural products OC1C(O)C(O)C(CO)OC1OC1C(O)C(O)C(O)C(CO)O1 HDTRYLNUVZCQOY-UHFFFAOYSA-N 0.000 description 1
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- LDDMACCNBZAMSG-BDVNFPICSA-N (2r,3r,4s,5r)-3,4,5,6-tetrahydroxy-2-(methylamino)hexanal Chemical compound CN[C@@H](C=O)[C@@H](O)[C@H](O)[C@H](O)CO LDDMACCNBZAMSG-BDVNFPICSA-N 0.000 description 1
- OWEGMIWEEQEYGQ-UHFFFAOYSA-N 100676-05-9 Natural products OC1C(O)C(O)C(CO)OC1OCC1C(O)C(O)C(O)C(OC2C(OC(O)C(O)C2O)CO)O1 OWEGMIWEEQEYGQ-UHFFFAOYSA-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
- MSWZFWKMSRAUBD-GASJEMHNSA-N 2-amino-2-deoxy-D-galactopyranose Chemical compound N[C@H]1C(O)O[C@H](CO)[C@H](O)[C@@H]1O MSWZFWKMSRAUBD-GASJEMHNSA-N 0.000 description 1
- MSWZFWKMSRAUBD-IVMDWMLBSA-N 2-amino-2-deoxy-D-glucopyranose Chemical compound N[C@H]1C(O)O[C@H](CO)[C@@H](O)[C@@H]1O MSWZFWKMSRAUBD-IVMDWMLBSA-N 0.000 description 1
- QRXMUCSWCMTJGU-UHFFFAOYSA-N 5-bromo-4-chloro-3-indolyl phosphate Chemical compound C1=C(Br)C(Cl)=C2C(OP(O)(=O)O)=CNC2=C1 QRXMUCSWCMTJGU-UHFFFAOYSA-N 0.000 description 1
- 208000003200 Adenoma Diseases 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 201000001320 Atherosclerosis Diseases 0.000 description 1
- 108010074708 B7-H1 Antigen Proteins 0.000 description 1
- 102000008096 B7-H1 Antigen Human genes 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 102100037904 CD9 antigen Human genes 0.000 description 1
- 239000012275 CTLA-4 inhibitor Substances 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 101100454807 Caenorhabditis elegans lgg-1 gene Proteins 0.000 description 1
- 101100454808 Caenorhabditis elegans lgg-2 gene Proteins 0.000 description 1
- 101100217502 Caenorhabditis elegans lgg-3 gene Proteins 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 102000019034 Chemokines Human genes 0.000 description 1
- 108010012236 Chemokines Proteins 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 208000001154 Dermoid Cyst Diseases 0.000 description 1
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 206010014759 Endometrial neoplasm Diseases 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 241000724791 Filamentous phage Species 0.000 description 1
- 229930091371 Fructose Natural products 0.000 description 1
- RFSUNEUAIZKAJO-ARQDHWQXSA-N Fructose Chemical compound OC[C@H]1O[C@](O)(CO)[C@@H](O)[C@@H]1O RFSUNEUAIZKAJO-ARQDHWQXSA-N 0.000 description 1
- 239000005715 Fructose Substances 0.000 description 1
- 206010017993 Gastrointestinal neoplasms Diseases 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 102000006354 HLA-DR Antigens Human genes 0.000 description 1
- 108010058597 HLA-DR Antigens Proteins 0.000 description 1
- 208000006933 Hermanski-Pudlak Syndrome Diseases 0.000 description 1
- 206010071775 Hermansky-Pudlak syndrome Diseases 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 208000017604 Hodgkin disease Diseases 0.000 description 1
- 208000021519 Hodgkin lymphoma Diseases 0.000 description 1
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 1
- 101000738354 Homo sapiens CD9 antigen Proteins 0.000 description 1
- 101000777628 Homo sapiens Leukocyte antigen CD37 Proteins 0.000 description 1
- 101000980823 Homo sapiens Leukocyte surface antigen CD53 Proteins 0.000 description 1
- 101000669513 Homo sapiens Metalloproteinase inhibitor 1 Proteins 0.000 description 1
- 101000946889 Homo sapiens Monocyte differentiation antigen CD14 Proteins 0.000 description 1
- 101000763579 Homo sapiens Toll-like receptor 1 Proteins 0.000 description 1
- 102000037982 Immune checkpoint proteins Human genes 0.000 description 1
- 108091008036 Immune checkpoint proteins Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 108090000174 Interleukin-10 Proteins 0.000 description 1
- 108090000172 Interleukin-15 Proteins 0.000 description 1
- 108050003558 Interleukin-17 Proteins 0.000 description 1
- 208000008839 Kidney Neoplasms Diseases 0.000 description 1
- LKDRXBCSQODPBY-AMVSKUEXSA-N L-(-)-Sorbose Chemical compound OCC1(O)OC[C@H](O)[C@@H](O)[C@@H]1O LKDRXBCSQODPBY-AMVSKUEXSA-N 0.000 description 1
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 1
- AHLPHDHHMVZTML-BYPYZUCNSA-N L-Ornithine Chemical compound NCCC[C@H](N)C(O)=O AHLPHDHHMVZTML-BYPYZUCNSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 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
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 229940125563 LAG3 inhibitor Drugs 0.000 description 1
- 240000007472 Leucaena leucocephala Species 0.000 description 1
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- 102100031586 Leukocyte antigen CD37 Human genes 0.000 description 1
- 102100024221 Leukocyte surface antigen CD53 Human genes 0.000 description 1
- 206010024612 Lipoma Diseases 0.000 description 1
- 206010025219 Lymphangioma Diseases 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 108010009489 Lysosomal-Associated Membrane Protein 3 Proteins 0.000 description 1
- 108010064171 Lysosome-Associated Membrane Glycoproteins Proteins 0.000 description 1
- 102000014944 Lysosome-Associated Membrane Glycoproteins Human genes 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 1
- GUBGYTABKSRVRQ-PICCSMPSSA-N Maltose Natural products O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@@H](CO)OC(O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-PICCSMPSSA-N 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 108050006599 Metalloproteinase inhibitor 1 Proteins 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 102100035877 Monocyte differentiation antigen CD14 Human genes 0.000 description 1
- 208000004304 Myofibromatosis Diseases 0.000 description 1
- 241000232901 Nephroma Species 0.000 description 1
- 208000007256 Nevus Diseases 0.000 description 1
- AHLPHDHHMVZTML-UHFFFAOYSA-N Orn-delta-NH2 Natural products NCCCC(N)C(O)=O AHLPHDHHMVZTML-UHFFFAOYSA-N 0.000 description 1
- UTJLXEIPEHZYQJ-UHFFFAOYSA-N Ornithine Natural products OC(=O)C(C)CCCN UTJLXEIPEHZYQJ-UHFFFAOYSA-N 0.000 description 1
- 239000012271 PD-L1 inhibitor Substances 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 241000282579 Pan Species 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 201000004681 Psoriasis Diseases 0.000 description 1
- MUPFEKGTMRGPLJ-RMMQSMQOSA-N Raffinose Natural products O(C[C@H]1[C@@H](O)[C@H](O)[C@@H](O)[C@@H](O[C@@]2(CO)[C@H](O)[C@@H](O)[C@@H](CO)O2)O1)[C@@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 MUPFEKGTMRGPLJ-RMMQSMQOSA-N 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 206010061934 Salivary gland cancer Diseases 0.000 description 1
- 206010039491 Sarcoma Diseases 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 102000007562 Serum Albumin Human genes 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- 206010041067 Small cell lung cancer Diseases 0.000 description 1
- 101100289792 Squirrel monkey polyomavirus large T gene Proteins 0.000 description 1
- 230000020385 T cell costimulation Effects 0.000 description 1
- 210000000662 T-lymphocyte subset Anatomy 0.000 description 1
- 229940125555 TIGIT inhibitor Drugs 0.000 description 1
- 108700012920 TNF Proteins 0.000 description 1
- 206010043276 Teratoma Diseases 0.000 description 1
- 108700031126 Tetraspanins Proteins 0.000 description 1
- 102000043977 Tetraspanins Human genes 0.000 description 1
- 208000024770 Thyroid neoplasm Diseases 0.000 description 1
- 108010031374 Tissue Inhibitor of Metalloproteinase-1 Proteins 0.000 description 1
- 102100027010 Toll-like receptor 1 Human genes 0.000 description 1
- HDTRYLNUVZCQOY-WSWWMNSNSA-N Trehalose Natural products O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-WSWWMNSNSA-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
- MUPFEKGTMRGPLJ-UHFFFAOYSA-N UNPD196149 Natural products OC1C(O)C(CO)OC1(CO)OC1C(O)C(O)C(O)C(COC2C(C(O)C(O)C(CO)O2)O)O1 MUPFEKGTMRGPLJ-UHFFFAOYSA-N 0.000 description 1
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 206010047741 Vulval cancer Diseases 0.000 description 1
- 239000008351 acetate buffer Substances 0.000 description 1
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 238000004220 aggregation Methods 0.000 description 1
- HDTRYLNUVZCQOY-LIZSDCNHSA-N alpha,alpha-trehalose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-LIZSDCNHSA-N 0.000 description 1
- WQZGKKKJIJFFOK-PHYPRBDBSA-N alpha-D-galactose Chemical compound OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@H]1O WQZGKKKJIJFFOK-PHYPRBDBSA-N 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 239000002246 antineoplastic agent Substances 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 235000006708 antioxidants Nutrition 0.000 description 1
- 239000003125 aqueous solvent Substances 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 206010003246 arthritis Diseases 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 108010030694 avidin-horseradish peroxidase complex Proteins 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- GUBGYTABKSRVRQ-QUYVBRFLSA-N beta-maltose Chemical compound OC[C@H]1O[C@H](O[C@H]2[C@H](O)[C@@H](O)[C@H](O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@@H]1O GUBGYTABKSRVRQ-QUYVBRFLSA-N 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 201000000053 blastoma Diseases 0.000 description 1
- 210000001772 blood platelet Anatomy 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 239000008366 buffered solution Substances 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 208000019425 cirrhosis of liver Diseases 0.000 description 1
- 235000015165 citric acid Nutrition 0.000 description 1
- 229960004106 citric acid Drugs 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 238000012875 competitive assay Methods 0.000 description 1
- 230000002860 competitive effect Effects 0.000 description 1
- 239000012141 concentrate Substances 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 230000000875 corresponding effect Effects 0.000 description 1
- 230000001351 cycling effect Effects 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 229960002433 cysteine Drugs 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000000368 destabilizing effect Effects 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 238000000502 dialysis Methods 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 150000002016 disaccharides Chemical class 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 210000003162 effector t lymphocyte Anatomy 0.000 description 1
- 201000008184 embryoma Diseases 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 230000001804 emulsifying effect Effects 0.000 description 1
- 201000003914 endometrial carcinoma Diseases 0.000 description 1
- 230000002357 endometrial effect Effects 0.000 description 1
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 1
- 235000020776 essential amino acid Nutrition 0.000 description 1
- 239000003797 essential amino acid Substances 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 150000002170 ethers Chemical class 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 229930195729 fatty acid Natural products 0.000 description 1
- 230000003176 fibrotic effect Effects 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 229930182830 galactose Natural products 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 229960002442 glucosamine Drugs 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 229960002743 glutamine Drugs 0.000 description 1
- 235000003969 glutathione Nutrition 0.000 description 1
- 229960003180 glutathione Drugs 0.000 description 1
- 125000005456 glyceride group Chemical group 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 229940093915 gynecological organic acid Drugs 0.000 description 1
- 201000011066 hemangioma Diseases 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 230000002440 hepatic effect Effects 0.000 description 1
- 238000013537 high throughput screening Methods 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 125000000487 histidyl group Chemical group [H]N([H])C(C(=O)O*)C([H])([H])C1=C([H])N([H])C([H])=N1 0.000 description 1
- 239000000819 hypertonic solution Substances 0.000 description 1
- 239000000815 hypotonic solution Substances 0.000 description 1
- 239000012216 imaging agent Substances 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000000899 immune system response Effects 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 238000010874 in vitro model Methods 0.000 description 1
- 238000011503 in vivo imaging Methods 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 201000011489 infantile myofibromatosis Diseases 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 230000003960 inflammatory cascade Effects 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 238000000185 intracerebroventricular administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- BQINXKOTJQCISL-GRCPKETISA-N keto-neuraminic acid Chemical compound OC(=O)C(=O)C[C@H](O)[C@@H](N)[C@@H](O)[C@H](O)[C@H](O)CO BQINXKOTJQCISL-GRCPKETISA-N 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 201000007270 liver cancer Diseases 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 201000005249 lung adenocarcinoma Diseases 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 238000012792 lyophilization process Methods 0.000 description 1
- 230000002101 lytic effect Effects 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 239000012092 media component Substances 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 210000003584 mesangial cell Anatomy 0.000 description 1
- 239000003226 mitogen Substances 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 230000004001 molecular interaction Effects 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 150000002772 monosaccharides Chemical class 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- DAZSWUUAFHBCGE-KRWDZBQOSA-N n-[(2s)-3-methyl-1-oxo-1-pyrrolidin-1-ylbutan-2-yl]-3-phenylpropanamide Chemical compound N([C@@H](C(C)C)C(=O)N1CCCC1)C(=O)CCC1=CC=CC=C1 DAZSWUUAFHBCGE-KRWDZBQOSA-N 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000009826 neoplastic cell growth Effects 0.000 description 1
- 208000025351 nephroma Diseases 0.000 description 1
- CERZMXAJYMMUDR-UHFFFAOYSA-N neuraminic acid Natural products NC1C(O)CC(O)(C(O)=O)OC1C(O)C(O)CO CERZMXAJYMMUDR-UHFFFAOYSA-N 0.000 description 1
- 239000002736 nonionic surfactant Substances 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 229960003104 ornithine Drugs 0.000 description 1
- 230000000242 pagocytic effect Effects 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 1
- 201000002628 peritoneum cancer Diseases 0.000 description 1
- 230000003285 pharmacodynamic effect Effects 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 150000004713 phosphodiesters Chemical group 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 230000010118 platelet activation Effects 0.000 description 1
- 229920001993 poloxamer 188 Polymers 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 229920002503 polyoxyethylene-polyoxypropylene Polymers 0.000 description 1
- 229920005606 polypropylene copolymer Polymers 0.000 description 1
- 229940068977 polysorbate 20 Drugs 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 229940068968 polysorbate 80 Drugs 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 239000001103 potassium chloride Substances 0.000 description 1
- 235000011164 potassium chloride Nutrition 0.000 description 1
- 230000001376 precipitating effect Effects 0.000 description 1
- 244000062645 predators Species 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 238000011809 primate model Methods 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 238000002818 protein evolution Methods 0.000 description 1
- 230000006916 protein interaction Effects 0.000 description 1
- 239000002510 pyrogen Substances 0.000 description 1
- 238000003127 radioimmunoassay Methods 0.000 description 1
- MUPFEKGTMRGPLJ-ZQSKZDJDSA-N raffinose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO[C@@H]2[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O2)O)O1 MUPFEKGTMRGPLJ-ZQSKZDJDSA-N 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 210000003289 regulatory T cell Anatomy 0.000 description 1
- 208000037803 restenosis Diseases 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 201000003804 salivary gland carcinoma Diseases 0.000 description 1
- 239000012266 salt solution Substances 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 208000000587 small cell lung carcinoma Diseases 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 229940054269 sodium pyruvate Drugs 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 235000010356 sorbitol Nutrition 0.000 description 1
- 238000001179 sorption measurement Methods 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 210000004988 splenocyte Anatomy 0.000 description 1
- 208000017572 squamous cell neoplasm Diseases 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 239000008174 sterile solution Substances 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-N succinic acid Chemical compound OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 241001515965 unidentified phage Species 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 206010046766 uterine cancer Diseases 0.000 description 1
- 208000012991 uterine carcinoma Diseases 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 201000005102 vulva cancer Diseases 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2896—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2809—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2818—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2827—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2878—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
- A61K2039/507—Comprising a combination of two or more separate antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/545—Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/74—Inducing cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/75—Agonist effect on antigen
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
Definitions
- the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist, where the CD63 agonist is administered to the subject to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder.
- the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist and a therapeutically effective amount of a T cell activator.
- pharmaceutical compositions and kits that find use, e.g., in practicing the methods.
- FIG. 1 A graph showing co-stimulation of human T cells via CD63 and CD3 using a first anti-CD63 antibody agonist.
- FIG. 2 A graph showing co-stimulation of human T cells via CD63 and CD3 using a second anti-CD63 antibody agonist.
- FIG. 3 Flow cytometry data showing CD63 expression levels on T cells in human ovarian cancer.
- FIG. 4 Flow cytometry data showing CD63 expression levels on tumor infiltrating lymphocytes (TILs) relative to blood in a human head and neck cancer patient.
- TILs tumor infiltrating lymphocytes
- FIG. 5 Data demonstrating the in vivo efficacy of a CD63 agonist in treating cancer.
- aspects of the present disclosure include methods of treating cell proliferative disorders.
- the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist, where the CD63 agonist is administered to the subject to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder.
- the methods of the present disclosure are based in part on the inventors’ surprising discovery that CD63 agonists enhance T cell responses to abnormally proliferating cells of a cell proliferative disorder (e.g., cancer), independent of whether the abnormally proliferating cells express or overexpress CD63.
- the CD63 agonist is administered to the subject independent of the level of expression of CD63 on the abnormally proliferating cells of the cell proliferative disorder.
- the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist, where at the time of the administering, abnormally proliferating cells of the cell proliferative disorder are not suspected of exhibiting overexpression of CD63.
- CD63 is not suspected to be expressed at higher levels on the abnormally proliferating cells (e.g., cancer cells) compared to a second cell population (e.g., non-abnormally proliferating (e.g., non-cancer) cells of the same cell/tissue type as the abnormally proliferating cells).
- a second cell population e.g., non-abnormally proliferating (e.g., non-cancer) cells of the same cell/tissue type as the abnormally proliferating cells.
- the methods include making such a determination.
- the subject to whom the CD63 agonist is administered is receiving a T cell activation therapy.
- a non-limiting example of such a T cell activation therapy is an immune checkpoint inhibitor therapy, e.g., a therapy where the subject is receiving one or more inhibitors of PD-1 , PD-L1 , CTLA-4, LAG-3, TIM-3, TIGIT, VISTA, B7-H3, and/or the like.
- the methods of the present disclosure may further include administering the T cell activation therapy to the subject.
- aspects of the present disclosure include methods of treating cell proliferative disorders, the methods including administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist and a therapeutically effective amount of a T cell activator. Details regarding embodiments of the methods of the present disclosure will be now be described.
- the tetraspanins are integral membrane proteins expressed on cell surface and granular membranes of hematopoietic cells and are components of multimolecular complexes with specific integrins.
- the tetraspanin CD63 also known as LAMP-3, melanoma-associated antigen ME491 , TSPAN30, MLA1 and OMA81 H
- CD63 (UniProtKB - P08962) functions as cell surface receptor for TIMP1 and plays a role in the activation of ITGB1 and integrin signaling, leading to the activation of AKT, FAK/PTK2 and MAP kinases.
- CD63 is expressed on activated platelets, monocytes and macrophages, and is weakly expressed on granulocytes, T cell and B cells. It is located on the basophilic granule membranes and on the plasma membranes of lymphocytes and granulocytes.
- CD63 is a member of the TM4 superfamily of leukocyte glycoproteins that includes CD9, CD37 and CD53, which contain four transmembrane regions. CD63 may play a role in phagocytic and intracellular lysosome-phagosome fusion events. CD63 deficiency is associated with Hermansky-Pudlak syndrome.
- the CD63 agonist may be administered to any of a variety of subjects.
- the subject is a“mammal” or“mammalian,” where these terms are used broadly to describe organisms which are within the class mammalia, including the orders carnivore (e.g., dogs and cats), rodentia (e.g., mice, guinea pigs, and rats), and primates (e.g., humans, chimpanzees, and monkeys).
- the subject is a human.
- the subject is an animal model (e.g., a mouse model, a primate model, or the like) of a cellular proliferative disorder, e.g., cancer.
- cell proliferative disorder is meant a disorder wherein unwanted cell proliferation of one or more subset(s) of cells in a multicellular organism occurs, resulting in harm, for example, pain or decreased life expectancy to the organism.
- Cell proliferative disorders include, but are not limited to, cancer, pre-cancer, benign tumors, blood vessel proliferative disorders (e.g., arthritis, restenosis, and the like), fibrotic disorders (e.g., hepatic cirrhosis, atherosclerosis, and the like), psoriasis, epidermic and dermoid cysts, lipomas, adenomas, capillary and cutaneous hemangiomas, lymphangiomas, nevi lesions, teratomas, nephromas, myofibromatosis, osteoplastic tumors, dysplastic masses, mesangial cell proliferative disorders, and the like.
- blood vessel proliferative disorders e.g., arthritis, restenosis, and the like
- fibrotic disorders e.g., hepatic cirrhosis, atherosclerosis, and the like
- psoriasis e.g., epidermic and dermoid cysts
- the subject has cancer.
- the subject methods may be employed for the treatment of a large variety of cancers by virtue of the enhanced anti cancer T cell response achieved.
- the subject has a cancer suspected of evading the immune system (e.g., effector T cells), e.g., by co-opting one or more immune checkpoint pathways.
- Tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
- cancer and“cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation.
- cancers that may be treated using the subject methods include, but are not limited to, carcinoma, lymphoma, blastoma, and sarcoma. More particular examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, various types of head and neck cancer, and the like.
- the subject has a cancer selected from melanoma, Hodgkin lymphoma, renal cell carcinoma (RCC), bladder cancer, non-small cell lung cancer (NSCLC), and head and neck squamous cell carcinoma (HNSCC).
- a cancer selected from melanoma, Hodgkin lymphoma, renal cell carcinoma (RCC), bladder cancer, non-small cell lung cancer (NSCLC), and head and neck squamous cell carcinoma (HNSCC).
- the subject has a cancer for which administration of a T cell activator (e.g., an immune checkpoint inhibitor (e.g., a CTLA-4 inhibitor, a PD-1 inhibitor, a PD-L1 inhibitor, a LAG-3 inhibitor, a TIM-3 inhibitor, an IDO inhibitor, a TIGIT inhibitor, a VISTA inhibitor, a B7/H3 inhibitor, and/or the like), an agonist of a T cell co-stimulatory receptor, an antagonist of a T cell inhibitory signal, and/or the like) to treat the cancer (alone or in combination with second anti-cancer agent) has been approved by the Food and Drug Administration (FDA) and/or the European Medicines Agency (EMA).
- a T cell activator e.g., an immune checkpoint inhibitor (e.g., a CTLA-4 inhibitor, a PD-1 inhibitor, a PD-L1 inhibitor, a LAG-3 inhibitor, a TIM-3 inhibitor, an IDO inhibitor, a TIGIT inhibitor, a
- CD63 agonist is meant an agent determined to have activity in agonizing CD63.
- An example assay for determining whether a CD63 binding agent is a CD63 agonist is one in which co-stimulation of CD4 T cells is assessed for a plate-bound CD63 agonist and the corresponding soluble CD63 agonist. Co-stimulation by the agent when plate-bound but not when soluble (or greater co-stimulation by the agent when plate-bound as compared to when soluble) indicates that the agent is a CD63 agonist, e.g., because the plate-bound agent is able to concentrate CD63 molecules on the surface of the cell, thereby facilitating signaling through CD63.
- the CD63 agonist is identified, e.g., using a suitable approach for screening small molecules (e.g., by screening a combinatorial library of small molecules), antibodies (e.g., by phage or yeast display of antibody libraries), ligands, or the like for the ability to bind CD63 with subsequent screening for the ability to agonize CD63.
- a suitable approach for screening small molecules e.g., by screening a combinatorial library of small molecules
- antibodies e.g., by phage or yeast display of antibody libraries
- ligands e.g., by phage or yeast display of antibody libraries
- CD63 agonists described herein may specifically bind to CD63.
- a CD63 agonist“specifically binds” to CD63 if it binds to or associates with CD63 with an affinity or K a (that is, an equilibrium association constant of a particular binding interaction with units of 1/M) of, for example, greater than or equal to about 10 5 M 1 .
- the CD63 agonist binds to CD63 with a K a greater than or equal to about 10 6 M ⁇ 10 7 M ⁇ 10 8 M ⁇ 10 9 M ⁇ 10 10 M ⁇ 10 11 M ⁇ 10 12 M ⁇ or 10 13 M 1 .
- “High affinity” binding refers to binding with a K a of at least 10 7 M -1 , at least 10 8 M -1 , at least 10 9 M -1 , at least 10 10 M 1 , at least 10 11 M -1 , at least 10 12 M -1 , at least 10 13 M -1 , or greater.
- affinity may be defined as an equilibrium dissociation constant (KD) of a particular binding interaction with units of M (e.g., 10 _5 M to 10 _13 M, or less).
- the binding affinity of the CD63 agonist for CD63 can be readily determined using conventional techniques, e.g., by competitive ELISA (enzyme-linked immunosorbent assay), by equilibrium dialysis, by using surface plasmon resonance (SPR) technology (e.g., the BIAcore 2000 instrument, using general procedures outlined by the manufacturer); by radioimmunoassay; or the like. Methods are available for measuring the affinity of a candidate CD63 agonist for CD63 expressed on the surface of cells (e.g., T cells) using direct binding or competition binding assays.
- SPR surface plasmon resonance
- the equilibrium binding constant (Kb) may be measured using a candidate CD63 agonist conjugated to a fluorophore or radioisotope, or a candidate CD63 agonist that contains an N- or C-terminal epitope tag for detection by a labeled antibody. If labels or tags are not feasible or desired, a competition binding assay can be used to determine the half-maximal inhibitory concentration (IC50), the amount of unlabeled candidate CD63 agonist at which 50% of the maximal signal of the labeled competitor is detectable. A Kb value can then be calculated from the measured IC50 value. Ligand depletion will be more pronounced when measuring high-affinity interactions over a lower concentration range, and can be avoided or minimized by decreasing the number of cells added in the experiment or by increasing the binding reaction volumes.
- IC50 half-maximal inhibitory concentration
- the CD63 agonist administered to the subject may vary.
- the CD63 agonist is a small molecule.
- a“small molecule” is a compound having a molecular weight of 1000 atomic mass units (amu) or less. In some embodiments, the small molecule is 750 amu or less, 500 amu or less, 400 amu or less, 300 amu or less, or 200 amu or less. In certain aspects, the small molecule is not made of repeating molecular units such as are present in a polymer.
- the CD63 agonist is a small molecule known to bind CD63. In other aspects, a small molecule CD63 agonist is identified, e.g., using a suitable approach for screening small molecules, e.g., by screening a combinatorial library of small molecules.
- the CD63 agonist is a peptide or polypeptide.
- the agonist may be a CD63 ligand.
- CD63 ligands of interest include, but are not limited to, tissue inhibitor of metalloproteinases-1 (TIMP-1 ; UniProtKB - P01033), integrin b1 (ITGB1 ; UniProtKB - Q5T3E5), or CD63-binding derivatives thereof, including CD63-binding fragments thereof.
- a “derivative” peptide or polypeptide CD63 agonist refers to a peptide or polypeptide that binds to and agonizes CD63 but has fewer or more amino acids than the wild-type/parental CD63 agonist, has one or more amino acid substitutions relative to the wild-type/parental CD63 agonist, or any combination thereof.
- the CD63 agonist is an antibody that specifically binds CD63.
- antibody and“immunoglobulin” include antibodies or immunoglobulins of any isotype (e.g., IgG (e.g., lgG1 , lgG2, lgG3 or lgG4), IgE, IgD, IgA, IgM, etc.), whole antibodies (e.g., antibodies composed of a tetramer which in turn is composed of two dimers of a heavy and light chain polypeptide); single chain antibodies; fragments of antibodies (e.g., fragments of whole or single chain antibodies) which retain specific binding to CD63, including, but not limited to, Fv, single chain Fv (scFv), Fab, F(ab’)2, Fab’, (scFv’)2, and diabodies; chimeric antibodies; monoclonal antibodies, human antibodies, humanized antibodies (e.g., humanized whole antibodies, humanized antibody fragment
- the antibodies may be detectably labeled, e.g., with an in vivo imaging agent, or the like.
- the antibodies may be further conjugated to other moieties, such as, e.g., polyethylene glycol (PEG), etc. Fusion to an antibody Fc region (or a fragment thereof), conjugation to PEG, etc. may find use, e.g., for increasing serum half-life of the antibody upon administration to the subject.
- PEG polyethylene glycol
- the CD63 agonist may be a known CD63-binding agent.
- the CD63 agonist is an antibody known to specifically bind CD63 and determined to be an agonist of CD63.
- the CD63 agonist may be an antibody having the binding properties of the MX-49.129.5 anti-human CD63 monoclonal antibody (Santa Cruz Biotechnology).
- the CD63 agonist may be an antibody that competes for binding to human CD63 with the MX-49.129.5 anti-human CD63 antibody. Whether a first antibody“competes with” a second antibody for binding to the compound may be readily determined using competitive binding assays known in the art. Competing antibodies may be identified, for example, via an antibody competition assay.
- a sample of a first antibody can be bound to a solid support. Then, a sample of a second antibody suspected of being able to compete with such first antibody is then added.
- One of the two antibodies is labelled. If the labeled antibody and the unlabeled antibody bind to separate and discrete sites on the compound, the labeled antibody will bind to the same level whether or not the suspected competing antibody is present. However, if the sites of interaction are identical or overlapping, the unlabeled antibody will compete, and the amount of labeled antibody bound to the antigen will be lowered. If the unlabeled antibody is present in excess, very little, if any, labeled antibody will bind.
- competing antibodies are those that decrease the binding of an antibody to the compound by about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or about 99% or more. Details of procedures for carrying out such competition assays are well known in the art and can be found, for example, in Harlow and Lane, Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1988, 567-569, 1988, ISBN 0-87969-314-2. Such assays can be made quantitative by using purified antibodies. A standard curve may be established by titrating one antibody against itself, i.e., the same antibody is used for both the label and the competitor. The capacity of an unlabeled competing antibody to inhibit the binding of the labeled antibody to the plate may be titrated. The results may be plotted, and the concentrations necessary to achieve the desired degree of binding inhibition may be compared.
- Antibodies that specifically bind CD63 can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, phage display technologies, or a combination thereof.
- an antibody may be made and isolated using methods of phage display.
- Phage display is used for the high-throughput screening of protein interactions. Phages may be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage expressing an antigen binding domain that binds CD63 can be selected or identified with CD63, e.g., using labeled CD63 bound or captured to a solid surface or bead.
- Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv (individual Fv region from light or heavy chains) or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein.
- Exemplary methods are set forth, for example, in U.S. Pat. No. 5,969,108, Hoogenboom, H. R. and Chames, Immunol. Today 2000, 21 :371 ; Nagy et al. Nat. Med. 2002, 8:801 ; Huie et al., Proc. Natl. Acad. Sci. USA 2001 , 98:2682; Lui et al., J.
- ribosomal display can be used to replace bacteriophage as the display platform (see, e.g., Hanes et al., Nat. Biotechnol. 2000, 18:1287; Wilson et al., Proc. Natl. Acad. Sci.
- Cell surface libraries may be screened for antibodies (Boder et al., Proc. Natl. Acad. Sci. USA 2000, 97:10701 ; Daugherty et al., J. Immunol. Methods 2000, 243:21 1 ). Such procedures provide alternatives to traditional hybridoma techniques for the isolation and subsequent cloning of monoclonal antibodies.
- the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria.
- techniques to recombinantly produce Fv, scFv, Fab, F(ab')2, and Fab' fragments may be employed using methods known in the art.
- the subject to whom the therapeutically effective amount of a CD63 agonist is administered may be receiving a T cell activation therapy.
- the methods further include administering such a T cell activation therapy to the subject.
- the CD63 agonist is administered to the subject to potentiate the T cell activation therapy.
- T cell activation therapy is meant a therapy that includes administration of a T cell activator to the subject.
- a “T cell activator” is an agent that stimulates an immune response in a T cell or group of T cells.
- TCR T cell receptor
- APC antigen presenting cell
- MHC major histocompatibility complex
- HLA human leukocyte antigen
- TCR is specific for a given epitope; however, the costimulatory protein is not epitope specific and instead is generally expressed on all T cells or on large T cell subsets.
- stimulation of an immune response in T cells can be determined by measuring antigen-induced production of cytokines by T cells.
- stimulation of an immune response in T cells can be determined by measuring antigen-induced production of IFNy, IL-4, IL-2, IL-10, IL-17 and/or TNFa by T cells.
- antigen-produced production of cytokines by T cells can be measured by intracellular cytokine staining followed by flow cytometry.
- antigen-induced production of cytokines by T cells can be measured by surface capture staining followed by flow cytometry. In some embodiments, antigen- induced production of cytokines by T cells can be measured by determining cytokine concentration in supernatants of activated T cell cultures. In some embodiments, this can be measured by ELISA.
- antigen-produced production of cytokines by T cells can be measured by ELISPOT assay.
- ELISPOT assays employ a technique very similar to the sandwich enzyme-linked immunosorbent assay (ELISA) technique.
- An antibody e.g. monoclonal antibody, polyclonal antibody, etc.
- An antibody is coated aseptically onto a PVDF (polyvinylidene fluoride)-backed microplate.
- Antibodies are chosen for their specificity for the cytokine in question. The plate is blocked (e.g. with a serum protein that is non-reactive with any of the antibodies in the assay).
- Cells of interest are plated out at varying densities, along with antigen or mitogen, and then placed in a humidified 37° C. CO2 incubator for a specified period of time.
- Cytokine secreted by activated cells is captured locally by the coated antibody on the high surface area PVDF membrane.
- a secondary antibody e.g., a biotinylated polyclonal antibody
- This antibody is reactive with a distinct epitope of the target cytokine and thus is employed to detect the captured cytokine.
- the detected cytokine is then visualized using an avidin-HRP, and a precipitating substrate (e.g., AEC, BCIP/NBT).
- a precipitating substrate e.g., AEC, BCIP/NBT.
- the colored end product typically represents an individual cytokine-producing cell. Spots can be counted manually (e.g., with a dissecting microscope) or using an automated reader to capture the microwell images and to analyze spot number and size. In some embodiments, each spot correlates to a single cytokine- producing cell.
- T cells activated by the T cell activator are specific for an epitope present on abnormally proliferative cells underlying the cellular proliferative disorder (e.g., cancer cells in an individual having cancer), and contacting such T cells with the T cell activator increases cytotoxic activity of the T cells toward the abnormally proliferating cells, increases the number of such epitope-specific T cells, or a combination thereof.
- the increase in cytotoxic activity is further potentiated by the CD63 agonist.
- the T cell activator is an immune checkpoint inhibitor.
- an “immune checkpoint inhibitor” is any agent (e.g., small molecule, nucleic acid, protein (e.g., antibody)) that prevents the suppression of any component in the immune system such as MHC class presentation, T cell presentation and/or differentiation, any cytokine, chemokine or signaling for immune cell proliferation and/or differentiation.
- the immune checkpoint inhibitor is selected from a cytotoxic T-lymphocyte- associated antigen 4 (CTLA-4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)-dioxygenase (IDO) inhibitor, a T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitor, a V-domain Ig suppressor of T cell activation (VISTA) inhibitor, a B7-H3 inhibitor, and any combination thereof.
- CTLA-4 cytotoxic T-lymphocyte- associated antigen 4
- PD-1 programmed cell death-1
- PD-L1 programmed cell death ligand-1
- LAG-3 lymphocyte activation gene-3
- TIM-3 T-cell immunoglobulin domain and mucin domain 3
- the T cell activator is an agonist of a T cell co stimulatory receptor.
- agonists include an 0X40 agonist, a glucocorticoid-induced TNFR-related protein (GITR) agonist, a CD137 agonist, and a CD40 agonist.
- GITR glucocorticoid-induced TNFR-related protein
- the T cell activator is an antagonist of a T cell inhibitory signal.
- Inhibitory antibodies to T cell inhibitors such as PD-1 or CTLA-4 increase the activation of T cells.
- These inhibitory T cell signals play a role in the natural tolerance to auto-antigens but also can play a role to limit T cell function against tumor antigens.
- blocking Abs when administered to tumor-bearing hosts they increase T cell activation/responses against tumors leading to increased immune-mediated tumor destruction.
- the T cell activator is a cytokine.
- Cytokines of interest in the context of the present disclosure are those that promote T cell activation (e.g., IL-1 , and the like), promote proliferation of activated T cells (e.g., IL-2, and the like), etc.
- Non-limiting examples of cytokines that may be administered with the CD63 agonist include IL-1 , IL-2, IL-4, IL-15, and any combination thereof.
- the T cell activator is an agent that blocks immune suppressive cytokines.
- a non-limiting example of such a T cell activator is a TGF-b receptor inhibitor.
- the T cell activator is an antagonist of an inhibitory immune receptor.
- such an antagonist binds directly to the inhibitory immune receptor, thereby blocking activation of the inhibitory immune receptor, e.g., by preventing binding of the receptor to its ligand.
- such an antagonist binds to the ligand of an inhibitory immune receptor, thereby blocking activation of the inhibitory immune receptor by preventing binding of the receptor to its ligand.
- Antagonists of inhibitory immune receptors that may be administered with the CD63 agonist include, but are not limited to, TGF-b.
- the subject to whom the CD63 agonist is administered is receiving an innate immune system stimulator therapy.
- the methods further include administering the innate immune system stimulator therapy to the subject.
- the administering e.g., the initial administration of the CD63 agonist, the innate immune system stimulator, or both (if present in a single formulation)
- abnormally proliferating cells of the cell proliferative disorder are not suspected of exhibiting overexpression of CD63.
- an“innate immune system stimulator” is an agent that stimulates an innate immune system response in the subject.
- agents that may be employed to stimulate the innate immune system include, but are not limited to, agents that bind to one or more Toll-like receptors (TLRs), e.g., one or more of TLR1 -TLR9.
- TLRs Toll-like receptors
- Most mammalian species have 10-13 types of TLRs and each receptor recognizes specific ligands and induces a wide array of inflammatory cascades.
- the innate immune system stimulator is an agent that includes unmethylated CpG dinucleotides. It is now understood that the immune stimulatory effects of bacterial DNA are a result of the presence of unmethylated CpG dinucleotides in particular base contexts (CpG motifs), which are common in bacterial DNA, but methylated and underrepresented in vertebrate DNA (Krieg et al, 1995 Nature 374:546-549; Krieg, 1999 Biochim. Biophys. Acta 93321 :1 -10). The immune stimulatory effects of bacterial DNA can be mimicked with synthetic oligodeoxynucleotides (ODN) containing these CpG motifs.
- ODN synthetic oligodeoxynucleotides
- Such CpG ODN have highly stimulatory effects on human and murine leukocytes, inducing B cell proliferation; cytokine and immunoglobulin secretion; natural killer (NK) cell lytic activity and IFN-g secretion; and activation of dendritic cells (DCs) and other antigen presenting cells to express costimulatory molecules and secrete cytokines, especially the Th1 -like cytokines that are important in promoting the development of Th1 -like T cell responses.
- DCs dendritic cells
- the CD63 agonist and, if also administered, a T cell activator and/or innate immune system stimulator, are administered in a therapeutically effective amount.
- therapeutically effective amount is meant a dosage sufficient to produce a desired result, e.g., an amount sufficient to effect beneficial or desired therapeutic (including preventative) results, such as a reduction in a symptom of the proliferative disorder, as compared to a control.
- the therapeutically effective amount is sufficient to slow the growth of a tumor, reduce the size of a tumor, and/or the like.
- An effective amount can be administered in one or more administrations.
- the methods include administering a combination of the CD63 agonist and a second agent (e.g., a T cell activator and/or innate immune system stimulator), the CD63 agonist and the second agent may be administered concurrently (e.g., in the same or separate formulations), sequentially, or both.
- the second agent is administered to the individual prior to administration of the CD63 agonist, concurrently with administration of the CD63 agonist, or both.
- the CD63 agonist is administered to the individual prior to administration of the second agent, concurrently with administration of the second agent, or both.
- the methods include administering the CD63 agonist to a subject to whom a T cell activator (e.g., immune checkpoint blocker (such as a PD1 inhibitor, etc.) or other T cell activator) has already been administered, and the CD63 agonist is administered to potentiate the efficacy of the prior administered T cell activator.
- a T cell activator e.g., immune checkpoint blocker (such as a PD1 inhibitor, etc.) or other T cell activator
- the one or more agents are administered according to a dosing regimen approved for individual use.
- the administration of the CD63 agonist permits the second agent to be administered according to a dosing regimen that involves one or more lower and/or less frequent doses, and/or a reduced number of cycles as compared with that utilized when the second agent is administered without administration of the CD63 agonist.
- the administration of the second agent permits the CD63 agonist to be administered according to a dosing regimen that involves one or more lower and/or less frequent doses, and/or a reduced number of cycles as compared with that utilized when the CD63 agonist is administered without administration of the second agent.
- one or more doses of the CD63 agonist and second agent are administered at the same time; in some such embodiments, such agents may be administered present in the same pharmaceutical composition.
- the CD63 agonist and second agent are administered to the individual in different compositions and/or at different times.
- the CD63 agonist may be administered prior to administration of the second agent (e.g., in a particular cycle).
- the second agent may be administered prior to administration of the CD63 agonist (e.g., in a particular cycle).
- the second agent to be administered may be administered a period of time that starts at least 1 hour, 3 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, or up to 5 days or more prior to or after the administration of the CD63 agonist.
- the CD63 agonist is administered to the individual for a desirable period of time prior to administration of the second agent.
- a regimen“primes” the immune system e.g., for T cell activation by the T cell activator and/or innate immune system stimulation by the innate immune system stimulator.
- the second agent is administered to the individual for a desirable period of time prior to administration of the CD63 agonist.
- administration of one agent is specifically timed relative to administration of another agent.
- a first agent is administered so that a particular effect is observed (or expected to be observed, for example based on population studies showing a correlation between a given dosing regimen and the particular effect of interest).
- desired relative dosing regimens for agents administered in combination may be assessed or determined empirically, for example using ex vivo, in vivo and/or in vitro models; in some embodiments, such assessment or empirical determination is made in vivo, in a patient population (e.g., so that a correlation is established), or alternatively in a particular subject of interest.
- the CD63 agonist may be administered a period of time after administration of a T cell activator and/or innate immune system stimulator.
- the period of time may be selected to be correlated with an increase in T cell activation and/or innate immune system stimulation by the T cell activator and/or innate immune system stimulator, respectively.
- the relevant period of time permits (e.g., is correlated with) T cell activation and/or innate immune system stimulation that is 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, 100% or more, 150% or more, 200% or more, 250% or more, 300% or more, 350% or more, 400% or more, 450% or more, or 500% or more, than that observed prior to (or at the moment of) the administration of a T cell activator and/or innate immune system stimulator, respectively.
- T cell activation and/or innate immune system stimulation that is 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, 100% or more, 150% or more, 200% or more, 250% or more, 300% or more, 350% or more, 400% or more, 450% or more, or 500% or more, than that observed prior to (or at the moment
- the CD63 agonist and second agent are administered according to an intermittent dosing regimen including at least two cycles. Where two or more agents are administered in combination, and each by such an intermittent, cycling, regimen, individual doses of different agents may be interdigitated with one another.
- one or more doses of the second agent is administered a period of time after a dose of the first agent.
- each dose of the second agent is administered a period of time after a dose of the first agent.
- each dose of the first agent is followed after a period of time by a dose of the second agent.
- two or more doses of the first agent are administered between at least one pair of doses of the second agent; in certain aspects, two or more doses of the second agent are administered between al least one pair of doses of the first agent.
- different doses of the same agent are separated by a common interval of time; in some embodiments, the interval of time between different doses of the same agent varies.
- different doses of the different agents are separated from one another by a common interval of time; in some embodiments, different doses of the different agents are separated from one another by different intervals of time.
- One exemplary protocol for interdigitating two intermittent, cycled dosing regimens may include: (a) a first dosing period during which a therapeutically effective amount a first agent is administered to an individual; (b) a first resting period; (c) a second dosing period during which a therapeutically effective amount of a second agent and, optionally, a third agent, is administered to the individual; and (d) a second resting period.
- the first resting period and second resting period may correspond to an identical number of hours or days. Alternatively, in some embodiments, the first resting period and second resting period are different, with either the first resting period being longer than the second one or, vice versa. In some embodiments, each of the resting periods corresponds to 120 hours, 96 hours, 72 hours, 48 hours, 24 hours, 12 hours, 6 hours, 30 hours, 1 hour, or less. In some embodiments, if the second resting period is longer than the first resting period, it can be defined as a number of days or weeks rather than hours (for instance 1 day, 3 days, 5 days, 1 week, 2, weeks, 4 weeks or more).
- the second resting period’s length may be determined on the basis of different factors, separately or in combination. Exemplary such factors may include type and/or stage of a cancer against which the agents are administered; identity and/or properties (e.g., pharmacokinetic properties) of the first agent, and/or one or more features of the patient’s response to therapy with the first agent. In some embodiments, length of one or both resting periods may be adjusted in light of pharmacokinetic properties (e.g., as assessed via plasma concentration levels) of one or the other (or both) of the administered agents.
- pharmacokinetic properties e.g., as assessed via plasma concentration levels
- a relevant resting period might be deemed to be completed when plasma concentration of the relevant agent is below about 1 pg/ml, 0.1 pg/ml, 0.01 pg/ml or 0.001 pg/ml, optionally upon evaluation or other consideration of one or more features of the individual’s response.
- the number of cycles for which a particular agent is administered may be determined empirically. Also, in some embodiments, the precise regimen followed (e.g., number of doses, spacing of doses (e.g., relative to each other or to another event such as administration of another therapy), amount of doses, etc.) may be different for one or more cycles as compared with one or more other cycles.
- the CD63 agonist, and if also administered, a second agent may be administered via a route of administration independently selected from oral, parenteral (e.g., by intravenous, intra-arterial, subcutaneous, intramuscular, or epidural injection), topical, or nasal administration.
- a route of administration independently selected from oral, parenteral (e.g., by intravenous, intra-arterial, subcutaneous, intramuscular, or epidural injection), topical, or nasal administration.
- the CD63 agonist and a second agent are both administered parenterally, either concurrently (in the same pharmaceutical composition or separate pharmaceutical compositions) or sequentially.
- aspects of the present disclosure include methods for treating a cell proliferative disorder (e.g., cancer).
- treatment is meant at least an amelioration of one or more symptoms associated with the cell proliferative disorder of the individual, where amelioration is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the cell proliferative disorder (e.g., cancer) being treated.
- a parameter e.g. symptom
- treatment also includes situations where the cell proliferative disorder, or at least one or more symptoms associated therewith, are completely inhibited, e.g., prevented from happening, or stopped, e.g., terminated, such that the individual no longer suffers from the cell proliferative disorder, or at least the symptoms that characterize the cell proliferative disorder.
- “treating” the cancer may include slowing the rate of tumor growth in the subject.
- a pharmaceutical composition of the present disclosure includes a CD63 agonist (e.g., any of the CD63 agonists described above), and a pharmaceutically acceptable excipient.
- a pharmaceutical composition of the present disclosure includes a CD63 agonist (e.g., any of the CD63 agonists described above), a T cell activator (e.g., any of the T cell activators described above), and a pharmaceutically acceptable excipient.
- a pharmaceutical composition of the present disclosure includes a CD63 agonist (e.g., any of the CD63 agonists described above), an innate immune system stimulator (e.g., any of the innate immune system stimulators described above), and a pharmaceutically acceptable excipient.
- a CD63 agonist e.g., any of the CD63 agonists described above
- an innate immune system stimulator e.g., any of the innate immune system stimulators described above
- a pharmaceutically acceptable excipient e.g., any of the CD63 agonists described above
- compositions of the present disclosure may include any of the agents and features described above in the section relating to the subject methods, which are incorporated but not reiterated in detail herein for purposes of brevity.
- the CD63 agonist present in the pharmaceutical composition is a small molecule.
- the CD63 agonist present in the pharmaceutical composition is a peptide or polypeptide.
- the CD63 agonist may be a CD63 ligand, an antibody that specifically binds CD63, or the like.
- the CD63 agonist is an antibody, the antibody may be selected from an IgG, Fv, scFv, Fab, F(ab')2, and Fab'.
- the T cell activator is an immune checkpoint inhibitor.
- the immune checkpoint inhibitor is selected from a cytotoxic T-lymphocyte- associated antigen 4 (CTLA-4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)-dioxygenase (IDO) inhibitor, a T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitor, a V-domain Ig suppressor of T cell activation (VISTA) inhibitor, a B7-H3 inhibitor, and any combination thereof.
- CTLA-4 cytotoxic T-lymphocyte- associated antigen 4
- PD-1 programmed cell death-1
- PD-L1 programmed cell death ligand-1
- LAG-3 lymphocyte activation gene-3
- TIM-3
- the T cell activator is an agonist of a T cell co-stimulatory receptor, such as but not limited to, an 0X40 agonist, a glucocorticoid-induced TNFFt- related protein (GITR) agonist, a CD137 agonist, a CD40 agonist, and/or the like.
- a T cell co-stimulatory receptor such as but not limited to, an 0X40 agonist, a glucocorticoid-induced TNFFt- related protein (GITR) agonist, a CD137 agonist, a CD40 agonist, and/or the like.
- a pharmaceutical composition of the present disclosure includes a T cell activator
- the T cell activator is an antagonist of a T cell inhibitory signal, a cytokine, an agent that blocks immune suppressive cytokines (e.g., a TGF-b receptor inhibitor), and/or an antagonist of an inhibitory immune receptor.
- a pharmaceutical composition of the present disclosure includes the CD63 agonist and a combination (that is, two or more) of T cell activators, including but not limited to any combination of the T cell activators described herein.
- the CD63 agonist, T cell activator, or both can be incorporated into a variety of formulations for therapeutic administration. More particularly, the agent(s) (that is, CD63 agonist and/or T cell activator) can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable excipients or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, injections, inhalants and aerosols.
- Formulations of the agents for administration to the subject are generally sterile and may further be free of detectable pyrogens or other contaminants contraindicated for administration to a patient according to a selected route of administration.
- the agent(s) can be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
- the following methods and carriers/excipients are merely examples and are in no way limiting.
- the agent(s) can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
- conventional additives such as lactose, mannitol, corn starch or potato starch
- binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
- disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
- lubricants such as talc or magnesium stearate
- the agent(s) can be formulated for parenteral (e.g., intravenous, intra-arterial, intraosseous, intramuscular, intracerebral, intracerebroventricular, intrathecal, subcutaneous, etc.) administration.
- parenteral e.g., intravenous, intra-arterial, intraosseous, intramuscular, intracerebral, intracerebroventricular, intrathecal, subcutaneous, etc.
- the agent(s) are formulated for injection by dissolving, suspending or emulsifying the agent(s) in an aqueous or non- aqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
- an aqueous or non- aqueous solvent such as vegetable or
- compositions that include the agent(s) may be prepared by mixing the agent(s) having the desired degree of purity with optional physiologically acceptable carriers, excipients, stabilizers, surfactants, buffers and/or tonicity agents.
- Acceptable carriers, excipients and/or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid, glutathione, cysteine, methionine and citric acid; preservatives (such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, or combinations thereof); amino acids such as arginine, glycine, ornithine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine
- the pharmaceutical composition may be in a liquid form, a lyophilized form or a liquid form reconstituted from a lyophilized form, wherein the lyophilized preparation is to be reconstituted with a sterile solution prior to administration.
- the standard procedure for reconstituting a lyophilized composition is to add back a volume of pure water (typically equivalent to the volume removed during lyophilization); however solutions comprising antibacterial agents may be used for the production of pharmaceutical compositions for parenteral administration.
- An aqueous formulation of the agent(s) may be prepared in a pH-buffered solution, e.g., at pH ranging from about 4.0 to about 7.0, or from about 5.0 to about 6.0, or alternatively about 5.5.
- buffers that are suitable for a pH within this range include phosphate-, histidine-, citrate-, succinate-, acetate-buffers and other organic acid buffers.
- the buffer concentration can be from about 1 mM to about 100 mM, or from about 5 mM to about 50 mM, depending, e.g., on the buffer and the desired tonicity of the formulation.
- a tonicity agent may be included to modulate the tonicity of the formulation.
- Example tonicity agents include sodium chloride, potassium chloride, glycerin and any component from the group of amino acids, sugars as well as combinations thereof.
- the aqueous formulation is isotonic, although hypertonic or hypotonic solutions may be suitable.
- the term "isotonic" denotes a solution having the same tonicity as some other solution with which it is compared, such as physiological salt solution or serum.
- Tonicity agents may be used in an amount of about 5 mM to about 350 mM, e.g., in an amount of 100 mM to 350 mM.
- a surfactant may also be added to the formulation to reduce aggregation and/or minimize the formation of particulates in the formulation and/or reduce adsorption.
- Example surfactants include polyoxyethylensorbitan fatty acid esters (Tween), polyoxyethylene alkyl ethers (Brij), alkylphenylpolyoxyethylene ethers (Triton-X), polyoxyethylene- polyoxypropylene copolymer (Poloxamer, Pluronic), and sodium dodecyl sulfate (SDS).
- suitable polyoxyethylenesorbitan-fatty acid esters are polysorbate 20, (sold under the trademark Tween 20TM) and polysorbate 80 (sold under the trademark Tween 80TM).
- Suitable polyethylene-polypropylene copolymers are those sold under the names Pluronic® F68 or Poloxamer 188TM.
- suitable Polyoxyethylene alkyl ethers are those sold under the trademark BrijTM.
- Example concentrations of surfactant may range from about 0.001 % to about 1 % w/v.
- a lyoprotectant may also be added in order to protect the CD63 agonist and/or T cell activator against destabilizing conditions during a lyophilization process.
- known lyoprotectants include sugars (including glucose and sucrose); polyols (including mannitol, sorbitol and glycerol); and amino acids (including alanine, glycine and glutamic acid). Lyoprotectants can be included in an amount of about 10 mM to 500 nM.
- the pharmaceutical composition includes the CD63 agonist and/or T cell activator, and one or more of the above-identified components (e.g., a surfactant, a buffer, a stabilizer, a tonicity agent) and is essentially free of one or more preservatives, such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, and combinations thereof.
- a preservative is included in the formulation, e.g., at concentrations ranging from about 0.001 to about 2% (w/v).
- kits find use, e.g., in practicing the methods of the present disclosure.
- a subject kit includes a pharmaceutical composition that includes a CD63 agonist, e.g., any of the CD63 agonists described elsewhere herein.
- kits that include any of the pharmaceutical compositions described herein, including any of the pharmaceutical compositions described above in the section relating to the compositions of the present disclosure.
- Kits of the present disclosure may include instructions for administering the pharmaceutical composition to a subject having a cell proliferative disorder, e.g., cancer.
- a kit of the present disclosure includes instructions for administering the pharmaceutical composition to the subject to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder.
- the instructions may include instructions for administering the pharmaceutical composition to the subject independent of the level of expression of CD63 on the abnormally proliferating cells of the cell proliferative disorder.
- a kit of the present disclosure includes instructions for administering the pharmaceutical composition to a subject having a cell proliferative disorder in which abnormally proliferating cells of the cell proliferative disorder are not suspected of exhibiting expression or overexpression of CD63.
- a kit of the present disclosure includes instructions for administering the pharmaceutical composition to a subject receiving a T cell activation therapy, e.g., to potentiate the T cell activation therapy.
- kits include a CD63 agonist, a T cell activator, and instructions for administering the CD63 agonist and T cell activator to a subject having a cell proliferative disorder, e.g., cancer.
- the instructions may further include any of the instructions for the kits described above, e.g., for administering the pharmaceutical composition to a subject having a cell proliferative disorder to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder (e.g., cancer), for administering the pharmaceutical composition to the subject independent of the level of expression of CD63 on the abnormally proliferating cells of the cell proliferative disorder, and/or the like.
- the CD63 agonist and T cell activator may be present in the same container, or may be present in separate containers.
- the kit may include instructions to administer the CD63 agonist prior to and/or subsequent to administering the T cell activator (e.g., immune checkpoint blocker (such as a PD1 inhibitor, etc.) or other T cell activator) to potentiate the efficacy of the T cell activator.
- the T cell activator e.g., immune checkpoint blocker (such as a PD1 inhibitor, etc.) or other T cell activator
- kits may include a quantity of the compositions, present in unit dosages, e.g., ampoules, or a multi-dosage format.
- the kits may include one or more (e.g., two or more) unit dosages (e.g., ampoules) of a composition that includes a CD63 agonist, a T cell activator, or both.
- unit dosage refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of the composition calculated in an amount sufficient to produce the desired effect.
- kits may include a single multi dosage amount of the composition.
- kits of the present disclosure may include any of the agents and features described above in the section relating to the subject methods and compositions, which are incorporated but not reiterated in detail herein for purposes of brevity.
- the CD63 agonist present in the kit is a small molecule. In certain aspects, the CD63 agonist present in the kit is a peptide or polypeptide. When the CD63 agonist is a peptide or polypeptide, the CD63 agonist may be a CD63 ligand, an antibody that specifically binds CD63, or the like. When the CD63 agonist is an antibody, the antibody may be selected from an IgG, Fv, scFv, Fab, F(ab')2, and Fab'.
- the T cell activator is an immune checkpoint inhibitor.
- the immune checkpoint inhibitor is selected from a cytotoxic T-lymphocyte-associated antigen 4 (CTLA- 4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)-dioxygenase (IDO) inhibitor, a T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitor, a V-domain Ig suppressor of T cell activation (VISTA) inhibitor, a B7-H3 inhibitor, and any combination thereof.
- CTLA-4 cytotoxic T-lymphocyte-associated antigen 4
- PD-1 programmed cell death-1
- PD-L1 programmed cell death ligand-1
- LAG-3 lymphocyte activation gene-3
- the T cell activator is an agonist of a T cell co-stimulatory receptor, such as but not limited to, an 0X40 agonist, a glucocorticoid-induced TNFR-related protein (GITR) agonist, a CD137 agonist, a CD40 agonist, and/or the like.
- a T cell co-stimulatory receptor such as but not limited to, an 0X40 agonist, a glucocorticoid-induced TNFR-related protein (GITR) agonist, a CD137 agonist, a CD40 agonist, and/or the like.
- GITR glucocorticoid-induced TNFR-related protein
- kits of the present disclosure include a T cell activator
- the T cell activator is an antagonist of a T cell inhibitory signal, a cytokine, an agent that blocks immune suppressive cytokines (e.g., a TGF-b receptor inhibitor), and/or an antagonist of an inhibitory immune receptor.
- a kit of the present disclosure includes the CD63 agonist and a combination (that is, two or more) of T cell activators, including but not limited to a combination of any of the T cell activators described herein.
- the CD63 agonist present in the kit is a small molecule. In certain aspects, the CD63 agonist present in the kit is a peptide or polypeptide. When the CD63 agonist is a peptide or polypeptide, the CD63 agonist may be a CD63 ligand, an antibody that specifically binds CD63, or the like. When the CD63 agonist is an antibody, the antibody may be selected from an IgG, Fv, scFv, Fab, F(ab')2, and Fab'.
- kits of the present disclosure include a T cell activator
- the T cell activator is an immune checkpoint inhibitor.
- Immune checkpoint inhibitors of interest include, but are not limited to, an agonist of a T cell co-stimulatory receptor, an antagonist of a T cell inhibitory signal, and/or the like.
- the immune checkpoint inhibitor is selected from a cytotoxic T-lymphocyte-associated antigen 4 (CTLA- 4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)-dioxygenase (IDO) inhibitor, an 0X40 agonist, a glucocorticoid-induced TNFR-related protein (GITR) agonist, a CD137 agonist, and a CD40 agonist.
- CTLA-4 cytotoxic T-lymphocyte-associated antigen 4
- PD-1 programmed cell death-1
- PD-L1 programmed cell death ligand-1
- LAG-3 lymphocyte activation gene-3
- TIM-3 T-cell immunoglobulin domain and mucin domain 3
- IDO indo
- kits of the present disclosure include a T cell activator
- the T cell activator is a cytokine.
- the T cell activator is an antagonist of an inhibitory immune receptor.
- a kit of the present disclosure includes a combination (that is, two or more) of any of the T cell activators described herein.
- kits may be present in separate containers, or multiple components may be present in a single container.
- a suitable container includes a single tube (e.g., vial), ampoule, one or more wells of a plate (e.g., a 96-well plate, a 384-well plate, etc.), or the like.
- the instructions (e.g., instructions for use (IFU)) included in the kits may be recorded on a suitable recording medium.
- the instructions may be printed on a substrate, such as paper or plastic, etc.
- the instructions may be present in the kits as a package insert, in the labeling of the container of the kit or components thereof (i.e., associated with the packaging or sub-packaging) etc.
- the instructions are present as an electronic storage data file present on a suitable computer readable storage medium, e.g., portable flash drive, DVD, CD-ROM, diskette, etc.
- the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source, e.g. via the internet, are provided.
- An example of this embodiment is a kit that includes a web address where the instructions can be viewed and/or from which the instructions can be downloaded.
- the means for obtaining the instructions is recorded on a suitable substrate.
- the binding agents to TCR (such as anti-CD3 antibodies) and to a potential co stimulation target may be provided in tandem to purified T cells.
- the resulting T cell proliferation may be quantified and used to gauge the efficacy of the co-stimulation target.
- FIG. 1 Shown in FIG. 1 is a graph depicting the results of T cell co-stimulation assays using anti- CD3 along with an anti-human CD63 co-stimulatory antibody (clone MX-49.129.5).
- Anti- 0X40 Providence, 9B12
- 1 % BSA/DPBS as a negative control.
- CD4+ T cells were purified from frozen PBMCs and resuspended in RPMI media supplemented with penicillin, streptomycin, glutamine, non-essential amino acids, HEPES buffer, and sodium pyruvate.
- PHA-L (2 pg/ml) and IL2 (60 lll/ml) were added to the media to activate the cells, which were then incubated at 37° under 5% CO2 for 96 hours.
- Ninety- six-well plates were coated with goat anti-mouse IgG and then blocked with BSA.
- Mouse anti-human CD3 (OKT3) was prepared at 2 ng/ml in 1 % BSA/DPBS, and added to the plates.
- Mouse anti-human CD63 was then added in serial, two-fold dilutions between 160 ng/ml and 1 .25 ng/ml or mouse anti-human 0X40 between 320 and 2.5 ng/ml.
- Activated CD4+ T cells were then added to each well at 50,000 cells/well in 200 mI of media, and then incubated at 37° for 48 hours.
- Cells were then labeled with 5-ethynyl-2’-deoxyuridine (EdU) at a final concentration of 10 mM, using the Click-it DNA labeling kit (Invitrogen) as per the manufacturer’s instructions for 18 hours. Cells were then treated with live-dead stain (E450; eBiosciences) and developed using the Alex647 agent (Invitrogen).
- Proliferating cells were then quantified via flow cytometry.
- the MX-49.129.5 anti-CD63 clone stimulated proliferation of CD4+ T cells in a dose-dependent manner, with proliferation observed at concentrations as low as 2.5 ng/ml of anti-CD63.
- proliferation via the classic CD4 T cell co stimulator, 0X40 was observed at concentrations as low as 10-20 ng/ml.
- mouse splenocytes from a C57BI/6 mouse were enriched for CD4 T cells using a negative selection kit.
- Cells were treated with CD3 250 ng/ml + CD63 agonist (the MAS- 24169 antibody) at a range from 0.125-2 pg/ml.
- Cells were then labeled with 5-ethynyl-2’- deoxyuridine (EdU) at a final concentration of 10 mM, using the Click-it DNA labeling kit (Invitrogen) as per the manufacturer’s instructions.
- CD63 on T cells in the tumor context were assessed by flow cytometry. Shown in FIG. 3 is flow cytometry data showing CD63 expression levels on T cells in human ovarian cancer. Increases in CD63 levels on T cells correlated with increased HLA-DR expression on conventional CD4+, Tregs, CD8+ and CD14+ cells in the tumor.
- FIG. 4 Shown in FIG. 4 is flow cytometry data showing CD63 expression levels on tumor infiltrating lymphocytes (TILs) relative to blood in a human head and neck cancer patient. An upregulation of CD63 is seen in the TILs as compared to blood from the same patient.
- TILs tumor infiltrating lymphocytes
- mice were inoculated subcutaneously with 4e5 MCA205 tumor cells on day 0. T umor sizes were measured 2 to 3 times per week.
- FIG. 5, panel A shows slight efficacy of the CD63 agonist when administered as a single therapeutic agent. Mice were treated on days 9, 12 and 15 with either 200 pg of hamster IgG control antibody or anti mouse CD63 i.p. (ThermoFisher Clone (MA5-24169) at 50 pg, 100 pg or 200 pg dose levels. With the single agent treatment of anti-CD63 0/5 mice reject their tumors.
- FIG. 5, panel B shows CD63 treatment, when administered following anti-PD1 treatment starting at day 12, leads to enhanced clearance with 1 1/15 mice rejecting tumors. Administration of the anti-PD1 treatment alone led to 3/15 cures, demonstrating that the CD63 agonist potentiates the anti-PD1 treatment. No mice treated with control IgG rejected their tumors.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Genetics & Genomics (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
Provided are methods of treating cell proliferative disorders. In some embodiments, the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist, where the CD63 agonist is administered to the subject to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder. In certain embodiments, the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist and a therapeutically effective amount of a T cell activator. Also provided are pharmaceutical compositions and kits that find use, e.g., in practicing the methods.
Description
CD63 AGONIST-RELATED METHODS AND COMPOSITIONS
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Patent Application No. 62/878,069, filed July 24, 2019, which application is incorporated herein by reference in its entirety.
SUMMARY
Provided are methods of treating cell proliferative disorders. In some embodiments, the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist, where the CD63 agonist is administered to the subject to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder. In certain embodiments, the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist and a therapeutically effective amount of a T cell activator. Also provided are pharmaceutical compositions and kits that find use, e.g., in practicing the methods.
BRIEF DESCRI PTION OF THE FIGURES
FIG. 1 : A graph showing co-stimulation of human T cells via CD63 and CD3 using a first anti-CD63 antibody agonist.
FIG. 2: A graph showing co-stimulation of human T cells via CD63 and CD3 using a second anti-CD63 antibody agonist.
FIG. 3: Flow cytometry data showing CD63 expression levels on T cells in human ovarian cancer.
FIG. 4: Flow cytometry data showing CD63 expression levels on tumor infiltrating lymphocytes (TILs) relative to blood in a human head and neck cancer patient.
FIG. 5: Data demonstrating the in vivo efficacy of a CD63 agonist in treating cancer.
DETAILED DESCRIPTION
Before the methods, compositions and kits of the present disclosure are described in greater detail, it is to be understood that the methods, compositions and kits are not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular
embodiments only, and is not intended to be limiting, since the scope of the methods, compositions and kits will be limited only by the appended claims.
Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the methods, compositions and kits. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges and are also encompassed within the methods, compositions and kits, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the methods, compositions and kits.
Certain ranges are presented herein with numerical values being preceded by the term“about.” The term“about” is used herein to provide literal support for the exact number that it precedes, as well as a number that is near to or approximately the number that the term precedes. In determining whether a number is near to or approximately a specifically recited number, the near or approximating unrecited number may be a number which, in the context in which it is presented, provides the substantial equivalent of the specifically recited number.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the methods, compositions and kits belong. Although any methods, compositions and kits similar or equivalent to those described herein can also be used in the practice or testing of the methods, compositions and kits, representative illustrative methods, compositions and kits are now described.
All publications and patents cited in this specification are herein incorporated by reference as if each individual publication or patent were specifically and individually indicated to be incorporated by reference and are incorporated herein by reference to disclose and describe the materials and/or methods in connection with which the publications are cited. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present methods, compositions and kits are not entitled to antedate such publication, as the date of publication provided may be different from the actual publication date which may need to be independently confirmed.
It is noted that, as used herein and in the appended claims, the singular forms“a”, “an”, and“the” include plural referents unless the context clearly dictates otherwise. It is further noted that the claims may be drafted to exclude any optional element. As such, this
statement is intended to serve as antecedent basis for use of such exclusive terminology as“solely,”“only” and the like in connection with the recitation of claim elements, or use of a“negative” limitation.
It is appreciated that certain features of the methods, compositions and kits, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the methods, compositions and kits, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination. All combinations of the embodiments are specifically embraced by the present disclosure and are disclosed herein just as if each and every combination was individually and explicitly disclosed, to the extent that such combinations embrace operable processes and/or compositions. In addition, all sub-combinations listed in the embodiments describing such variables are also specifically embraced by the present methods, compositions and kits and are disclosed herein just as if each and every such sub-combination was individually and explicitly disclosed herein.
As will be apparent to those of skill in the art upon reading this disclosure, each of the individual embodiments described and illustrated herein has discrete components and features which may be readily separated from or combined with the features of any of the other several embodiments without departing from the scope or spirit of the present methods. Any recited method can be carried out in the order of events recited or in any other order that is logically possible.
METHODS
Aspects of the present disclosure include methods of treating cell proliferative disorders. According to some embodiments, the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist, where the CD63 agonist is administered to the subject to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder. The methods of the present disclosure are based in part on the inventors’ surprising discovery that CD63 agonists enhance T cell responses to abnormally proliferating cells of a cell proliferative disorder (e.g., cancer), independent of whether the abnormally proliferating cells express or overexpress CD63. As such, in certain embodiments, the CD63 agonist is administered to the subject independent of the level of expression of CD63 on the abnormally proliferating cells of the cell proliferative disorder. According to some embodiments, the methods include administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist, where at the time of the administering, abnormally
proliferating cells of the cell proliferative disorder are not suspected of exhibiting overexpression of CD63. By“not suspected of exhibiting overexpression of CD63” is meant that CD63 is not suspected to be expressed at higher levels on the abnormally proliferating cells (e.g., cancer cells) compared to a second cell population (e.g., non-abnormally proliferating (e.g., non-cancer) cells of the same cell/tissue type as the abnormally proliferating cells). In certain embodiments, at the time of the administering, it has been determined that abnormally proliferating cells of the cell proliferative disorder do not overexpress CD63. According to some embodiments, the methods include making such a determination. In certain embodiments, the subject to whom the CD63 agonist is administered is receiving a T cell activation therapy. A non-limiting example of such a T cell activation therapy is an immune checkpoint inhibitor therapy, e.g., a therapy where the subject is receiving one or more inhibitors of PD-1 , PD-L1 , CTLA-4, LAG-3, TIM-3, TIGIT, VISTA, B7-H3, and/or the like. The methods of the present disclosure may further include administering the T cell activation therapy to the subject. Accordingly, aspects of the present disclosure include methods of treating cell proliferative disorders, the methods including administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist and a therapeutically effective amount of a T cell activator. Details regarding embodiments of the methods of the present disclosure will be now be described.
The tetraspanins are integral membrane proteins expressed on cell surface and granular membranes of hematopoietic cells and are components of multimolecular complexes with specific integrins. The tetraspanin CD63 (also known as LAMP-3, melanoma-associated antigen ME491 , TSPAN30, MLA1 and OMA81 H) is a lysosomal membrane glycoprotein that translocates to the plasma membrane after platelet activation. CD63 (UniProtKB - P08962) functions as cell surface receptor for TIMP1 and plays a role in the activation of ITGB1 and integrin signaling, leading to the activation of AKT, FAK/PTK2 and MAP kinases. CD63 is expressed on activated platelets, monocytes and macrophages, and is weakly expressed on granulocytes, T cell and B cells. It is located on the basophilic granule membranes and on the plasma membranes of lymphocytes and granulocytes. CD63 is a member of the TM4 superfamily of leukocyte glycoproteins that includes CD9, CD37 and CD53, which contain four transmembrane regions. CD63 may play a role in phagocytic and intracellular lysosome-phagosome fusion events. CD63 deficiency is associated with Hermansky-Pudlak syndrome.
The CD63 agonist may be administered to any of a variety of subjects. In certain embodiments, the subject is a“mammal” or“mammalian,” where these terms are used broadly to describe organisms which are within the class mammalia, including the orders
carnivore (e.g., dogs and cats), rodentia (e.g., mice, guinea pigs, and rats), and primates (e.g., humans, chimpanzees, and monkeys). According to some embodiments, the subject is a human. In certain embodiments, the subject is an animal model (e.g., a mouse model, a primate model, or the like) of a cellular proliferative disorder, e.g., cancer.
As summarized above, the subject has a cell proliferative disorder. By “cell proliferative disorder” is meant a disorder wherein unwanted cell proliferation of one or more subset(s) of cells in a multicellular organism occurs, resulting in harm, for example, pain or decreased life expectancy to the organism. Cell proliferative disorders include, but are not limited to, cancer, pre-cancer, benign tumors, blood vessel proliferative disorders (e.g., arthritis, restenosis, and the like), fibrotic disorders (e.g., hepatic cirrhosis, atherosclerosis, and the like), psoriasis, epidermic and dermoid cysts, lipomas, adenomas, capillary and cutaneous hemangiomas, lymphangiomas, nevi lesions, teratomas, nephromas, myofibromatosis, osteoplastic tumors, dysplastic masses, mesangial cell proliferative disorders, and the like.
In some embodiments, the subject has cancer. The subject methods may be employed for the treatment of a large variety of cancers by virtue of the enhanced anti cancer T cell response achieved. In some embodiments, the subject has a cancer suspected of evading the immune system (e.g., effector T cells), e.g., by co-opting one or more immune checkpoint pathways. “Tumor”, as used herein, refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues. The terms“cancer” and“cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation. Examples of cancers that may be treated using the subject methods include, but are not limited to, carcinoma, lymphoma, blastoma, and sarcoma. More particular examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, various types of head and neck cancer, and the like. In certain aspects, the subject has a cancer selected from melanoma, Hodgkin lymphoma, renal cell carcinoma (RCC), bladder cancer, non-small cell lung cancer (NSCLC), and head and neck squamous cell carcinoma (HNSCC). In some embodiments, the subject has a cancer for which administration of a T cell activator (e.g., an immune checkpoint inhibitor (e.g., a CTLA-4 inhibitor, a PD-1 inhibitor, a PD-L1 inhibitor, a LAG-3 inhibitor, a TIM-3 inhibitor, an IDO
inhibitor, a TIGIT inhibitor, a VISTA inhibitor, a B7/H3 inhibitor, and/or the like), an agonist of a T cell co-stimulatory receptor, an antagonist of a T cell inhibitory signal, and/or the like) to treat the cancer (alone or in combination with second anti-cancer agent) has been approved by the Food and Drug Administration (FDA) and/or the European Medicines Agency (EMA).
By“CD63 agonist” is meant an agent determined to have activity in agonizing CD63. An example assay for determining whether a CD63 binding agent is a CD63 agonist is one in which co-stimulation of CD4 T cells is assessed for a plate-bound CD63 agonist and the corresponding soluble CD63 agonist. Co-stimulation by the agent when plate-bound but not when soluble (or greater co-stimulation by the agent when plate-bound as compared to when soluble) indicates that the agent is a CD63 agonist, e.g., because the plate-bound agent is able to concentrate CD63 molecules on the surface of the cell, thereby facilitating signaling through CD63.
In certain aspects, the CD63 agonist is identified, e.g., using a suitable approach for screening small molecules (e.g., by screening a combinatorial library of small molecules), antibodies (e.g., by phage or yeast display of antibody libraries), ligands, or the like for the ability to bind CD63 with subsequent screening for the ability to agonize CD63.
Any of the CD63 agonists described herein may specifically bind to CD63. Such a CD63 agonist“specifically binds” to CD63 if it binds to or associates with CD63 with an affinity or Ka (that is, an equilibrium association constant of a particular binding interaction with units of 1/M) of, for example, greater than or equal to about 105 M 1. In certain embodiments, the CD63 agonist binds to CD63 with a Ka greater than or equal to about 106 M \ 107 M \ 108 M \ 109 M \ 1010 M \ 1011 M \ 1012 M \ or 1013 M 1. “High affinity” binding refers to binding with a Ka of at least 107 M-1 , at least 108 M-1 , at least 109 M-1 , at least 1010 M 1 , at least 1011 M-1 , at least 1012 M-1 , at least 1013 M-1 , or greater. Alternatively, affinity may be defined as an equilibrium dissociation constant (KD) of a particular binding interaction with units of M (e.g., 10_5 M to 10_13 M, or less). In certain aspects, specific binding means the CD63 agonist binds to CD63 with a KD of less than or equal to about 10- 5 M, less than or equal to about 10-6 M, less than or equal to about 10-7 M, less than or equal to about 10 8 M, or less than or equal to about 10-9 M, 10_1° M, 10-11 M, or 10-12 M or less. The binding affinity of the CD63 agonist for CD63 can be readily determined using conventional techniques, e.g., by competitive ELISA (enzyme-linked immunosorbent assay), by equilibrium dialysis, by using surface plasmon resonance (SPR) technology (e.g., the BIAcore 2000 instrument, using general procedures outlined by the manufacturer); by radioimmunoassay; or the like.
Methods are available for measuring the affinity of a candidate CD63 agonist for CD63 expressed on the surface of cells (e.g., T cells) using direct binding or competition binding assays. In a direct binding assay, the equilibrium binding constant (Kb) may be measured using a candidate CD63 agonist conjugated to a fluorophore or radioisotope, or a candidate CD63 agonist that contains an N- or C-terminal epitope tag for detection by a labeled antibody. If labels or tags are not feasible or desired, a competition binding assay can be used to determine the half-maximal inhibitory concentration (IC50), the amount of unlabeled candidate CD63 agonist at which 50% of the maximal signal of the labeled competitor is detectable. A Kb value can then be calculated from the measured IC50 value. Ligand depletion will be more pronounced when measuring high-affinity interactions over a lower concentration range, and can be avoided or minimized by decreasing the number of cells added in the experiment or by increasing the binding reaction volumes.
The CD63 agonist administered to the subject may vary. In certain aspects, the CD63 agonist is a small molecule. As used herein, a“small molecule” is a compound having a molecular weight of 1000 atomic mass units (amu) or less. In some embodiments, the small molecule is 750 amu or less, 500 amu or less, 400 amu or less, 300 amu or less, or 200 amu or less. In certain aspects, the small molecule is not made of repeating molecular units such as are present in a polymer. In some embodiments, the CD63 agonist is a small molecule known to bind CD63. In other aspects, a small molecule CD63 agonist is identified, e.g., using a suitable approach for screening small molecules, e.g., by screening a combinatorial library of small molecules.
In some embodiments, the CD63 agonist is a peptide or polypeptide. When the CD63 agonist is a peptide or polypeptide, the agonist may be a CD63 ligand. CD63 ligands of interest include, but are not limited to, tissue inhibitor of metalloproteinases-1 (TIMP-1 ; UniProtKB - P01033), integrin b1 (ITGB1 ; UniProtKB - Q5T3E5), or CD63-binding derivatives thereof, including CD63-binding fragments thereof. As used herein, a “derivative” peptide or polypeptide CD63 agonist (e.g., a ligand, an antibody, or the like) refers to a peptide or polypeptide that binds to and agonizes CD63 but has fewer or more amino acids than the wild-type/parental CD63 agonist, has one or more amino acid substitutions relative to the wild-type/parental CD63 agonist, or any combination thereof.
In certain aspects, the CD63 agonist is an antibody that specifically binds CD63. The terms "antibody" and“immunoglobulin” include antibodies or immunoglobulins of any isotype (e.g., IgG (e.g., lgG1 , lgG2, lgG3 or lgG4), IgE, IgD, IgA, IgM, etc.), whole antibodies (e.g., antibodies composed of a tetramer which in turn is composed of two dimers of a heavy and light chain polypeptide); single chain antibodies; fragments of antibodies (e.g., fragments of whole or single chain antibodies) which retain specific binding to CD63,
including, but not limited to, Fv, single chain Fv (scFv), Fab, F(ab’)2, Fab’, (scFv’)2, and diabodies; chimeric antibodies; monoclonal antibodies, human antibodies, humanized antibodies (e.g., humanized whole antibodies, humanized antibody fragments, etc.); and fusion proteins including an antigen-binding portion of an antibody and a non-antibody protein or fragment thereof, e.g., an antibody Fc region or fragment thereof. The antibodies may be detectably labeled, e.g., with an in vivo imaging agent, or the like. The antibodies may be further conjugated to other moieties, such as, e.g., polyethylene glycol (PEG), etc. Fusion to an antibody Fc region (or a fragment thereof), conjugation to PEG, etc. may find use, e.g., for increasing serum half-life of the antibody upon administration to the subject.
The CD63 agonist may be a known CD63-binding agent. For example, in some embodiments, the CD63 agonist is an antibody known to specifically bind CD63 and determined to be an agonist of CD63. In one non-limiting example, the CD63 agonist may be an antibody having the binding properties of the MX-49.129.5 anti-human CD63 monoclonal antibody (Santa Cruz Biotechnology). For example, the CD63 agonist may be an antibody that competes for binding to human CD63 with the MX-49.129.5 anti-human CD63 antibody. Whether a first antibody“competes with” a second antibody for binding to the compound may be readily determined using competitive binding assays known in the art. Competing antibodies may be identified, for example, via an antibody competition assay. For example, a sample of a first antibody can be bound to a solid support. Then, a sample of a second antibody suspected of being able to compete with such first antibody is then added. One of the two antibodies is labelled. If the labeled antibody and the unlabeled antibody bind to separate and discrete sites on the compound, the labeled antibody will bind to the same level whether or not the suspected competing antibody is present. However, if the sites of interaction are identical or overlapping, the unlabeled antibody will compete, and the amount of labeled antibody bound to the antigen will be lowered. If the unlabeled antibody is present in excess, very little, if any, labeled antibody will bind.
For purposes of the present disclosure, competing antibodies are those that decrease the binding of an antibody to the compound by about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, or about 99% or more. Details of procedures for carrying out such competition assays are well known in the art and can be found, for example, in Harlow and Lane, Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1988, 567-569, 1988, ISBN 0-87969-314-2. Such assays can be made quantitative by using purified antibodies. A standard curve may be established by titrating one antibody against itself, i.e., the same antibody is used for both the label and the competitor. The capacity of an unlabeled competing antibody to inhibit the binding of the
labeled antibody to the plate may be titrated. The results may be plotted, and the concentrations necessary to achieve the desired degree of binding inhibition may be compared.
Antibodies that specifically bind CD63 (e.g., human CD63; UniProtKB - P08962) can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, phage display technologies, or a combination thereof. For example, an antibody may be made and isolated using methods of phage display. Phage display is used for the high-throughput screening of protein interactions. Phages may be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage expressing an antigen binding domain that binds CD63 can be selected or identified with CD63, e.g., using labeled CD63 bound or captured to a solid surface or bead. Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv (individual Fv region from light or heavy chains) or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein. Exemplary methods are set forth, for example, in U.S. Pat. No. 5,969,108, Hoogenboom, H. R. and Chames, Immunol. Today 2000, 21 :371 ; Nagy et al. Nat. Med. 2002, 8:801 ; Huie et al., Proc. Natl. Acad. Sci. USA 2001 , 98:2682; Lui et al., J. Mol. Biol. 2002, 315:1063, each of which is incorporated herein by reference. Several publications (e.g., Marks et al., Bio/Technology 1992, 10:779-783) have described the production of high affinity human antibodies by chain shuffling, as well as combinatorial infection and in vivo recombination as a strategy for constructing large phage libraries. In another embodiment, ribosomal display can be used to replace bacteriophage as the display platform (see, e.g., Hanes et al., Nat. Biotechnol. 2000, 18:1287; Wilson et al., Proc. Natl. Acad. Sci. USA 2001 , 98:3750; or Irving et al., J. Immunol. Methods 2001 , 248:31 ). Cell surface libraries may be screened for antibodies (Boder et al., Proc. Natl. Acad. Sci. USA 2000, 97:10701 ; Daugherty et al., J. Immunol. Methods 2000, 243:21 1 ). Such procedures provide alternatives to traditional hybridoma techniques for the isolation and subsequent cloning of monoclonal antibodies.
After phage selection, the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria. For example, techniques to recombinantly produce Fv, scFv, Fab, F(ab')2, and Fab' fragments may be employed using methods known in the art.
As summarized above, the subject to whom the therapeutically effective amount of a CD63 agonist is administered may be receiving a T cell activation therapy. In certain
embodiments, the methods further include administering such a T cell activation therapy to the subject. According to some embodiments, the CD63 agonist is administered to the subject to potentiate the T cell activation therapy. By“T cell activation therapy” is meant a therapy that includes administration of a T cell activator to the subject. As used herein, a “T cell activator” is an agent that stimulates an immune response in a T cell or group of T cells. Such an immune response involves the engagement of the T cell receptor (TCR), present on the surface of a T cell, with a small peptide antigen non-covalently presented on the surface of an antigen presenting cell (APC) by a major histocompatibility complex (MHC; also referred to in humans as a human leukocyte antigen (HLA) complex). This engagement represents the immune system’s targeting mechanism and is a requisite molecular interaction for T cell activation and effector function. Following epitope-specific cell targeting, the targeted T cells are activated through engagement of costimulatory proteins found on the APC with counterpart costimulatory proteins on the T cells. Both signals - epitope/TCR binding and engagement of APC costimulatory proteins with T cell costimulatory proteins - are required to drive T cell specificity and activation. The TCR is specific for a given epitope; however, the costimulatory protein is not epitope specific and instead is generally expressed on all T cells or on large T cell subsets.
Various assays can be utilized in order to determine whether an immune response has been stimulated in a T cell or group of T cells, i.e., whether a T cell or group of T cells has become“activated”. In certain aspects, stimulation of an immune response in T cells can be determined by measuring antigen-induced production of cytokines by T cells. In some embodiments, stimulation of an immune response in T cells can be determined by measuring antigen-induced production of IFNy, IL-4, IL-2, IL-10, IL-17 and/or TNFa by T cells. In some embodiments, antigen-produced production of cytokines by T cells can be measured by intracellular cytokine staining followed by flow cytometry. In some embodiments, antigen-induced production of cytokines by T cells can be measured by surface capture staining followed by flow cytometry. In some embodiments, antigen- induced production of cytokines by T cells can be measured by determining cytokine concentration in supernatants of activated T cell cultures. In some embodiments, this can be measured by ELISA.
In some embodiments, antigen-produced production of cytokines by T cells can be measured by ELISPOT assay. In general, ELISPOT assays employ a technique very similar to the sandwich enzyme-linked immunosorbent assay (ELISA) technique. An antibody (e.g. monoclonal antibody, polyclonal antibody, etc.) is coated aseptically onto a PVDF (polyvinylidene fluoride)-backed microplate. Antibodies are chosen for their specificity for the cytokine in question. The plate is blocked (e.g. with a serum protein that is non-reactive
with any of the antibodies in the assay). Cells of interest are plated out at varying densities, along with antigen or mitogen, and then placed in a humidified 37° C. CO2 incubator for a specified period of time. Cytokine secreted by activated cells is captured locally by the coated antibody on the high surface area PVDF membrane. After washing the wells to remove cells, debris, and media components, a secondary antibody (e.g., a biotinylated polyclonal antibody) specific for the cytokine is added to the wells. This antibody is reactive with a distinct epitope of the target cytokine and thus is employed to detect the captured cytokine. Following a wash to remove any unbound biotinylated antibody, the detected cytokine is then visualized using an avidin-HRP, and a precipitating substrate (e.g., AEC, BCIP/NBT). The colored end product (a spot, usually a blackish blue) typically represents an individual cytokine-producing cell. Spots can be counted manually (e.g., with a dissecting microscope) or using an automated reader to capture the microwell images and to analyze spot number and size. In some embodiments, each spot correlates to a single cytokine- producing cell.
In some instances, T cells activated by the T cell activator are specific for an epitope present on abnormally proliferative cells underlying the cellular proliferative disorder (e.g., cancer cells in an individual having cancer), and contacting such T cells with the T cell activator increases cytotoxic activity of the T cells toward the abnormally proliferating cells, increases the number of such epitope-specific T cells, or a combination thereof. In some embodiments, the increase in cytotoxic activity is further potentiated by the CD63 agonist.
A wide variety of agents may be employed as the T cell activator. In certain embodiments, the T cell activator is an immune checkpoint inhibitor. As used herein, an “immune checkpoint inhibitor” is any agent (e.g., small molecule, nucleic acid, protein (e.g., antibody)) that prevents the suppression of any component in the immune system such as MHC class presentation, T cell presentation and/or differentiation, any cytokine, chemokine or signaling for immune cell proliferation and/or differentiation. According to some embodiments, the immune checkpoint inhibitor is selected from a cytotoxic T-lymphocyte- associated antigen 4 (CTLA-4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)-dioxygenase (IDO) inhibitor, a T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitor, a V-domain Ig suppressor of T cell activation (VISTA) inhibitor, a B7-H3 inhibitor, and any combination thereof.
According to some embodiments, the T cell activator is an agonist of a T cell co stimulatory receptor. Non-limiting examples of such agonists include an 0X40 agonist, a
glucocorticoid-induced TNFR-related protein (GITR) agonist, a CD137 agonist, and a CD40 agonist.
In certain embodiments, the T cell activator is an antagonist of a T cell inhibitory signal.
Inhibitory antibodies to T cell inhibitors such as PD-1 or CTLA-4 increase the activation of T cells. These inhibitory T cell signals play a role in the natural tolerance to auto-antigens but also can play a role to limit T cell function against tumor antigens. Hence, when blocking Abs are administered to tumor-bearing hosts they increase T cell activation/responses against tumors leading to increased immune-mediated tumor destruction.
According to some embodiments, the T cell activator is a cytokine. Cytokines of interest in the context of the present disclosure are those that promote T cell activation (e.g., IL-1 , and the like), promote proliferation of activated T cells (e.g., IL-2, and the like), etc. Non-limiting examples of cytokines that may be administered with the CD63 agonist include IL-1 , IL-2, IL-4, IL-15, and any combination thereof.
In certain embodiments, the T cell activator is an agent that blocks immune suppressive cytokines. A non-limiting example of such a T cell activator is a TGF-b receptor inhibitor.
According to some embodiments, the T cell activator is an antagonist of an inhibitory immune receptor. In certain embodiments, such an antagonist binds directly to the inhibitory immune receptor, thereby blocking activation of the inhibitory immune receptor, e.g., by preventing binding of the receptor to its ligand. In other aspects, such an antagonist binds to the ligand of an inhibitory immune receptor, thereby blocking activation of the inhibitory immune receptor by preventing binding of the receptor to its ligand. Antagonists of inhibitory immune receptors that may be administered with the CD63 agonist include, but are not limited to, TGF-b.
In certain embodiments, the subject to whom the CD63 agonist is administered is receiving an innate immune system stimulator therapy. According to some embodiments, the methods further include administering the innate immune system stimulator therapy to the subject. Optionally, at the time of the administering (e.g., the initial administration of the CD63 agonist, the innate immune system stimulator, or both (if present in a single formulation)), abnormally proliferating cells of the cell proliferative disorder are not suspected of exhibiting overexpression of CD63.
As used herein, an“innate immune system stimulator” is an agent that stimulates an innate immune system response in the subject. Examples of agents that may be employed to stimulate the innate immune system include, but are not limited to, agents that bind to
one or more Toll-like receptors (TLRs), e.g., one or more of TLR1 -TLR9. Most mammalian species have 10-13 types of TLRs and each receptor recognizes specific ligands and induces a wide array of inflammatory cascades.
In some embodiments, the innate immune system stimulator is an agent that includes unmethylated CpG dinucleotides. It is now understood that the immune stimulatory effects of bacterial DNA are a result of the presence of unmethylated CpG dinucleotides in particular base contexts (CpG motifs), which are common in bacterial DNA, but methylated and underrepresented in vertebrate DNA (Krieg et al, 1995 Nature 374:546-549; Krieg, 1999 Biochim. Biophys. Acta 93321 :1 -10). The immune stimulatory effects of bacterial DNA can be mimicked with synthetic oligodeoxynucleotides (ODN) containing these CpG motifs. Such CpG ODN have highly stimulatory effects on human and murine leukocytes, inducing B cell proliferation; cytokine and immunoglobulin secretion; natural killer (NK) cell lytic activity and IFN-g secretion; and activation of dendritic cells (DCs) and other antigen presenting cells to express costimulatory molecules and secrete cytokines, especially the Th1 -like cytokines that are important in promoting the development of Th1 -like T cell responses. These immune stimulatory effects of native phosphodiester backbone CpG ODN are highly CpG specific in that the effects are dramatically reduced if the CpG motif is methylated, changed to a GpC, or otherwise eliminated or altered (Krieg et al, 1995 Nature 374:546-549; Hartmann et al, 1999 Proc. Natl. Acad. Sci USA 96:9305-10).
The CD63 agonist and, if also administered, a T cell activator and/or innate immune system stimulator, are administered in a therapeutically effective amount. By “therapeutically effective amount” is meant a dosage sufficient to produce a desired result, e.g., an amount sufficient to effect beneficial or desired therapeutic (including preventative) results, such as a reduction in a symptom of the proliferative disorder, as compared to a control. When the cell proliferative disorder is cancer, in some embodiments, the therapeutically effective amount is sufficient to slow the growth of a tumor, reduce the size of a tumor, and/or the like. An effective amount can be administered in one or more administrations.
When the methods include administering a combination of the CD63 agonist and a second agent (e.g., a T cell activator and/or innate immune system stimulator), the CD63 agonist and the second agent may be administered concurrently (e.g., in the same or separate formulations), sequentially, or both. For example, according to certain embodiments, the second agent is administered to the individual prior to administration of the CD63 agonist, concurrently with administration of the CD63 agonist, or both. In some
embodiments, the CD63 agonist is administered to the individual prior to administration of the second agent, concurrently with administration of the second agent, or both.
According to some embodiments, the methods include administering the CD63 agonist to a subject to whom a T cell activator (e.g., immune checkpoint blocker (such as a PD1 inhibitor, etc.) or other T cell activator) has already been administered, and the CD63 agonist is administered to potentiate the efficacy of the prior administered T cell activator. A non-limiting example of such potentiation is demonstrated in the Experimental section below.
In certain embodiments, the one or more agents are administered according to a dosing regimen approved for individual use. In some embodiments, the administration of the CD63 agonist permits the second agent to be administered according to a dosing regimen that involves one or more lower and/or less frequent doses, and/or a reduced number of cycles as compared with that utilized when the second agent is administered without administration of the CD63 agonist. In some embodiments, the administration of the second agent permits the CD63 agonist to be administered according to a dosing regimen that involves one or more lower and/or less frequent doses, and/or a reduced number of cycles as compared with that utilized when the CD63 agonist is administered without administration of the second agent.
As noted above, in certain embodiments, one or more doses of the CD63 agonist and second agent are administered at the same time; in some such embodiments, such agents may be administered present in the same pharmaceutical composition. In some embodiments, however, the CD63 agonist and second agent are administered to the individual in different compositions and/or at different times. For example, the CD63 agonist may be administered prior to administration of the second agent (e.g., in a particular cycle). Alternatively, the second agent may be administered prior to administration of the CD63 agonist (e.g., in a particular cycle). The second agent to be administered may be administered a period of time that starts at least 1 hour, 3 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, or up to 5 days or more prior to or after the administration of the CD63 agonist.
In one example, the CD63 agonist is administered to the individual for a desirable period of time prior to administration of the second agent. In certain aspects, such a regimen“primes” the immune system, e.g., for T cell activation by the T cell activator and/or innate immune system stimulation by the innate immune system stimulator. In another example, the second agent is administered to the individual for a desirable period of time prior to administration of the CD63 agonist. In certain aspects, such a regimen“primes” the immune system for T cell activation by the CD63 agonist.
In some embodiments, administration of one agent is specifically timed relative to administration of another agent. For example, in some embodiments, a first agent is administered so that a particular effect is observed (or expected to be observed, for example based on population studies showing a correlation between a given dosing regimen and the particular effect of interest).
In certain aspects, desired relative dosing regimens for agents administered in combination may be assessed or determined empirically, for example using ex vivo, in vivo and/or in vitro models; in some embodiments, such assessment or empirical determination is made in vivo, in a patient population (e.g., so that a correlation is established), or alternatively in a particular subject of interest.
By way of example, the CD63 agonist may be administered a period of time after administration of a T cell activator and/or innate immune system stimulator. The period of time may be selected to be correlated with an increase in T cell activation and/or innate immune system stimulation by the T cell activator and/or innate immune system stimulator, respectively. In some embodiments, the relevant period of time permits (e.g., is correlated with) T cell activation and/or innate immune system stimulation that is 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, 100% or more, 150% or more, 200% or more, 250% or more, 300% or more, 350% or more, 400% or more, 450% or more, or 500% or more, than that observed prior to (or at the moment of) the administration of a T cell activator and/or innate immune system stimulator, respectively.
In some embodiments, the CD63 agonist and second agent are administered according to an intermittent dosing regimen including at least two cycles. Where two or more agents are administered in combination, and each by such an intermittent, cycling, regimen, individual doses of different agents may be interdigitated with one another. In certain aspects, one or more doses of the second agent is administered a period of time after a dose of the first agent. In some embodiments, each dose of the second agent is administered a period of time after a dose of the first agent. In certain aspects, each dose of the first agent is followed after a period of time by a dose of the second agent. In some embodiments, two or more doses of the first agent are administered between at least one pair of doses of the second agent; in certain aspects, two or more doses of the second agent are administered between al least one pair of doses of the first agent. In some embodiments, different doses of the same agent are separated by a common interval of time; in some embodiments, the interval of time between different doses of the same agent varies. In certain aspects, different doses of the different agents are separated from one
another by a common interval of time; in some embodiments, different doses of the different agents are separated from one another by different intervals of time.
One exemplary protocol for interdigitating two intermittent, cycled dosing regimens (e.g., for potentiating the effect of the T cell activator, CD63 agonist, or both), may include: (a) a first dosing period during which a therapeutically effective amount a first agent is administered to an individual; (b) a first resting period; (c) a second dosing period during which a therapeutically effective amount of a second agent and, optionally, a third agent, is administered to the individual; and (d) a second resting period.
In some embodiments, the first resting period and second resting period may correspond to an identical number of hours or days. Alternatively, in some embodiments, the first resting period and second resting period are different, with either the first resting period being longer than the second one or, vice versa. In some embodiments, each of the resting periods corresponds to 120 hours, 96 hours, 72 hours, 48 hours, 24 hours, 12 hours, 6 hours, 30 hours, 1 hour, or less. In some embodiments, if the second resting period is longer than the first resting period, it can be defined as a number of days or weeks rather than hours (for instance 1 day, 3 days, 5 days, 1 week, 2, weeks, 4 weeks or more).
If the first resting period’s length is determined by existence or development of a particular biological or therapeutic event (e.g., increased T cell activation), then the second resting period’s length may be determined on the basis of different factors, separately or in combination. Exemplary such factors may include type and/or stage of a cancer against which the agents are administered; identity and/or properties (e.g., pharmacokinetic properties) of the first agent, and/or one or more features of the patient’s response to therapy with the first agent. In some embodiments, length of one or both resting periods may be adjusted in light of pharmacokinetic properties (e.g., as assessed via plasma concentration levels) of one or the other (or both) of the administered agents. For example, a relevant resting period might be deemed to be completed when plasma concentration of the relevant agent is below about 1 pg/ml, 0.1 pg/ml, 0.01 pg/ml or 0.001 pg/ml, optionally upon evaluation or other consideration of one or more features of the individual’s response.
In certain aspects, the number of cycles for which a particular agent is administered may be determined empirically. Also, in some embodiments, the precise regimen followed (e.g., number of doses, spacing of doses (e.g., relative to each other or to another event such as administration of another therapy), amount of doses, etc.) may be different for one or more cycles as compared with one or more other cycles.
The CD63 agonist, and if also administered, a second agent (e.g., a T cell activator and/or innate immune system stimulator), may be administered via a route of administration independently selected from oral, parenteral (e.g., by intravenous, intra-arterial,
subcutaneous, intramuscular, or epidural injection), topical, or nasal administration. According to certain embodiments, the CD63 agonist and a second agent are both administered parenterally, either concurrently (in the same pharmaceutical composition or separate pharmaceutical compositions) or sequentially.
As described above, aspects of the present disclosure include methods for treating a cell proliferative disorder (e.g., cancer). By treatment is meant at least an amelioration of one or more symptoms associated with the cell proliferative disorder of the individual, where amelioration is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the cell proliferative disorder (e.g., cancer) being treated. As such, treatment also includes situations where the cell proliferative disorder, or at least one or more symptoms associated therewith, are completely inhibited, e.g., prevented from happening, or stopped, e.g., terminated, such that the individual no longer suffers from the cell proliferative disorder, or at least the symptoms that characterize the cell proliferative disorder. When the subject has cancer,“treating” the cancer may include slowing the rate of tumor growth in the subject.
COMPOSITIONS
As summarized above, aspects of the present disclosure include pharmaceutical compositions. In certain embodiments, a pharmaceutical composition of the present disclosure includes a CD63 agonist (e.g., any of the CD63 agonists described above), and a pharmaceutically acceptable excipient. According to some embodiments, a pharmaceutical composition of the present disclosure includes a CD63 agonist (e.g., any of the CD63 agonists described above), a T cell activator (e.g., any of the T cell activators described above), and a pharmaceutically acceptable excipient. In certain embodiments, a pharmaceutical composition of the present disclosure includes a CD63 agonist (e.g., any of the CD63 agonists described above), an innate immune system stimulator (e.g., any of the innate immune system stimulators described above), and a pharmaceutically acceptable excipient.
As will be appreciated, the pharmaceutical compositions of the present disclosure may include any of the agents and features described above in the section relating to the subject methods, which are incorporated but not reiterated in detail herein for purposes of brevity.
In some embodiments, the CD63 agonist present in the pharmaceutical composition is a small molecule. In certain aspects, the CD63 agonist present in the pharmaceutical composition is a peptide or polypeptide. When the CD63 agonist is a peptide or polypeptide, the CD63 agonist may be a CD63 ligand, an antibody that specifically binds
CD63, or the like. When the CD63 agonist is an antibody, the antibody may be selected from an IgG, Fv, scFv, Fab, F(ab')2, and Fab'.
When a pharmaceutical composition of the present disclosure includes a T cell activator, in certain embodiments, the T cell activator is an immune checkpoint inhibitor. When the T cell activator is an immune checkpoint inhibitor, according to some embodiments, the immune checkpoint inhibitor is selected from a cytotoxic T-lymphocyte- associated antigen 4 (CTLA-4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)-dioxygenase (IDO) inhibitor, a T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitor, a V-domain Ig suppressor of T cell activation (VISTA) inhibitor, a B7-H3 inhibitor, and any combination thereof.
When a pharmaceutical composition of the present disclosure includes a T cell activator, in certain embodiments, the T cell activator is an agonist of a T cell co-stimulatory receptor, such as but not limited to, an 0X40 agonist, a glucocorticoid-induced TNFFt- related protein (GITR) agonist, a CD137 agonist, a CD40 agonist, and/or the like.
When a pharmaceutical composition of the present disclosure includes a T cell activator, in some embodiments, the T cell activator is an antagonist of a T cell inhibitory signal, a cytokine, an agent that blocks immune suppressive cytokines (e.g., a TGF-b receptor inhibitor), and/or an antagonist of an inhibitory immune receptor. In some embodiments, a pharmaceutical composition of the present disclosure includes the CD63 agonist and a combination (that is, two or more) of T cell activators, including but not limited to any combination of the T cell activators described herein.
The CD63 agonist, T cell activator, or both, can be incorporated into a variety of formulations for therapeutic administration. More particularly, the agent(s) (that is, CD63 agonist and/or T cell activator) can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable excipients or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, injections, inhalants and aerosols.
Formulations of the agents for administration to the subject (e.g., suitable for human administration) are generally sterile and may further be free of detectable pyrogens or other contaminants contraindicated for administration to a patient according to a selected route of administration.
In pharmaceutical dosage forms, the agent(s) can be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate
association, as well as in combination, with other pharmaceutically active compounds. The following methods and carriers/excipients are merely examples and are in no way limiting.
For oral preparations, the agent(s) can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
The agent(s) can be formulated for parenteral (e.g., intravenous, intra-arterial, intraosseous, intramuscular, intracerebral, intracerebroventricular, intrathecal, subcutaneous, etc.) administration. In certain aspects, the agent(s) are formulated for injection by dissolving, suspending or emulsifying the agent(s) in an aqueous or non- aqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
Pharmaceutical compositions that include the agent(s) may be prepared by mixing the agent(s) having the desired degree of purity with optional physiologically acceptable carriers, excipients, stabilizers, surfactants, buffers and/or tonicity agents. Acceptable carriers, excipients and/or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid, glutathione, cysteine, methionine and citric acid; preservatives (such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, or combinations thereof); amino acids such as arginine, glycine, ornithine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, tryptophan, methionine, serine, proline and combinations thereof; monosaccharides, disaccharides and other carbohydrates; low molecular weight (less than about 10 residues) polypeptides; proteins, such as gelatin or serum albumin; chelating agents such as EDTA; sugars such as trehalose, sucrose, lactose, glucose, mannose, maltose, galactose, fructose, sorbose, raffinose, glucosamine, N- methylglucosamine, galactosamine, and neuraminic acid; and/or non-ionic surfactants such as Tween, Brij Pluronics, Triton-X, or polyethylene glycol (PEG).
The pharmaceutical composition may be in a liquid form, a lyophilized form or a liquid form reconstituted from a lyophilized form, wherein the lyophilized preparation is to be reconstituted with a sterile solution prior to administration. The standard procedure for
reconstituting a lyophilized composition is to add back a volume of pure water (typically equivalent to the volume removed during lyophilization); however solutions comprising antibacterial agents may be used for the production of pharmaceutical compositions for parenteral administration.
An aqueous formulation of the agent(s) may be prepared in a pH-buffered solution, e.g., at pH ranging from about 4.0 to about 7.0, or from about 5.0 to about 6.0, or alternatively about 5.5. Examples of buffers that are suitable for a pH within this range include phosphate-, histidine-, citrate-, succinate-, acetate-buffers and other organic acid buffers. The buffer concentration can be from about 1 mM to about 100 mM, or from about 5 mM to about 50 mM, depending, e.g., on the buffer and the desired tonicity of the formulation.
A tonicity agent may be included to modulate the tonicity of the formulation. Example tonicity agents include sodium chloride, potassium chloride, glycerin and any component from the group of amino acids, sugars as well as combinations thereof. In some embodiments, the aqueous formulation is isotonic, although hypertonic or hypotonic solutions may be suitable. The term "isotonic" denotes a solution having the same tonicity as some other solution with which it is compared, such as physiological salt solution or serum. Tonicity agents may be used in an amount of about 5 mM to about 350 mM, e.g., in an amount of 100 mM to 350 mM.
A surfactant may also be added to the formulation to reduce aggregation and/or minimize the formation of particulates in the formulation and/or reduce adsorption. Example surfactants include polyoxyethylensorbitan fatty acid esters (Tween), polyoxyethylene alkyl ethers (Brij), alkylphenylpolyoxyethylene ethers (Triton-X), polyoxyethylene- polyoxypropylene copolymer (Poloxamer, Pluronic), and sodium dodecyl sulfate (SDS). Examples of suitable polyoxyethylenesorbitan-fatty acid esters are polysorbate 20, (sold under the trademark Tween 20™) and polysorbate 80 (sold under the trademark Tween 80™). Examples of suitable polyethylene-polypropylene copolymers are those sold under the names Pluronic® F68 or Poloxamer 188™. Examples of suitable Polyoxyethylene alkyl ethers are those sold under the trademark Brij™. Example concentrations of surfactant may range from about 0.001 % to about 1 % w/v.
A lyoprotectant may also be added in order to protect the CD63 agonist and/or T cell activator against destabilizing conditions during a lyophilization process. For example, known lyoprotectants include sugars (including glucose and sucrose); polyols (including mannitol, sorbitol and glycerol); and amino acids (including alanine, glycine and glutamic acid). Lyoprotectants can be included in an amount of about 10 mM to 500 nM.
In some embodiments, the pharmaceutical composition includes the CD63 agonist and/or T cell activator, and one or more of the above-identified components (e.g., a surfactant, a buffer, a stabilizer, a tonicity agent) and is essentially free of one or more preservatives, such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, and combinations thereof. In other embodiments, a preservative is included in the formulation, e.g., at concentrations ranging from about 0.001 to about 2% (w/v).
KITS
As summarized above, the present disclosure provides kits. The kits find use, e.g., in practicing the methods of the present disclosure. According to some embodiments, a subject kit includes a pharmaceutical composition that includes a CD63 agonist, e.g., any of the CD63 agonists described elsewhere herein. In certain embodiments, provided are kits that include any of the pharmaceutical compositions described herein, including any of the pharmaceutical compositions described above in the section relating to the compositions of the present disclosure. Kits of the present disclosure may include instructions for administering the pharmaceutical composition to a subject having a cell proliferative disorder, e.g., cancer. According to some embodiments, a kit of the present disclosure includes instructions for administering the pharmaceutical composition to the subject to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder. The instructions may include instructions for administering the pharmaceutical composition to the subject independent of the level of expression of CD63 on the abnormally proliferating cells of the cell proliferative disorder. In certain embodiments, a kit of the present disclosure includes instructions for administering the pharmaceutical composition to a subject having a cell proliferative disorder in which abnormally proliferating cells of the cell proliferative disorder are not suspected of exhibiting expression or overexpression of CD63. According to some embodiments, a kit of the present disclosure includes instructions for administering the pharmaceutical composition to a subject receiving a T cell activation therapy, e.g., to potentiate the T cell activation therapy.
In some embodiments, kits are provided that include a CD63 agonist, a T cell activator, and instructions for administering the CD63 agonist and T cell activator to a subject having a cell proliferative disorder, e.g., cancer. The instructions may further include any of the instructions for the kits described above, e.g., for administering the pharmaceutical composition to a subject having a cell proliferative disorder to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder (e.g., cancer), for administering the pharmaceutical composition to the subject independent of the level of
expression of CD63 on the abnormally proliferating cells of the cell proliferative disorder, and/or the like. The CD63 agonist and T cell activator may be present in the same container, or may be present in separate containers. When the CD63 agonist and T cell activator are present in separate containers, the kit may include instructions to administer the CD63 agonist prior to and/or subsequent to administering the T cell activator (e.g., immune checkpoint blocker (such as a PD1 inhibitor, etc.) or other T cell activator) to potentiate the efficacy of the T cell activator.
The subject kits may include a quantity of the compositions, present in unit dosages, e.g., ampoules, or a multi-dosage format. As such, in certain embodiments, the kits may include one or more (e.g., two or more) unit dosages (e.g., ampoules) of a composition that includes a CD63 agonist, a T cell activator, or both. The term“unit dosage”, as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of the composition calculated in an amount sufficient to produce the desired effect. The amount of the unit dosage depends on various factors, such as the particular CD63 agonist and/or T cell activator employed, the effect to be achieved, and the pharmacodynamics associated with the CD63 agonist and/or T cell activator, in the subject. In yet other embodiments, the kits may include a single multi dosage amount of the composition.
As will be appreciated, the kits of the present disclosure may include any of the agents and features described above in the section relating to the subject methods and compositions, which are incorporated but not reiterated in detail herein for purposes of brevity.
In some embodiments, the CD63 agonist present in the kit is a small molecule. In certain aspects, the CD63 agonist present in the kit is a peptide or polypeptide. When the CD63 agonist is a peptide or polypeptide, the CD63 agonist may be a CD63 ligand, an antibody that specifically binds CD63, or the like. When the CD63 agonist is an antibody, the antibody may be selected from an IgG, Fv, scFv, Fab, F(ab')2, and Fab'.
When a kit of the present disclosure includes a T cell activator, in certain embodiments, the T cell activator is an immune checkpoint inhibitor. When the T cell activator is an immune checkpoint inhibitor, according to some embodiments, the immune checkpoint inhibitor is selected from a cytotoxic T-lymphocyte-associated antigen 4 (CTLA- 4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)-dioxygenase (IDO) inhibitor, a T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitor, a V-domain
Ig suppressor of T cell activation (VISTA) inhibitor, a B7-H3 inhibitor, and any combination thereof.
When a kit of the present disclosure includes a T cell activator, in certain embodiments, the T cell activator is an agonist of a T cell co-stimulatory receptor, such as but not limited to, an 0X40 agonist, a glucocorticoid-induced TNFR-related protein (GITR) agonist, a CD137 agonist, a CD40 agonist, and/or the like.
When a kit of the present disclosure includes a T cell activator, in some embodiments, the T cell activator is an antagonist of a T cell inhibitory signal, a cytokine, an agent that blocks immune suppressive cytokines (e.g., a TGF-b receptor inhibitor), and/or an antagonist of an inhibitory immune receptor. In some embodiments, a kit of the present disclosure includes the CD63 agonist and a combination (that is, two or more) of T cell activators, including but not limited to a combination of any of the T cell activators described herein.
In some embodiments, the CD63 agonist present in the kit is a small molecule. In certain aspects, the CD63 agonist present in the kit is a peptide or polypeptide. When the CD63 agonist is a peptide or polypeptide, the CD63 agonist may be a CD63 ligand, an antibody that specifically binds CD63, or the like. When the CD63 agonist is an antibody, the antibody may be selected from an IgG, Fv, scFv, Fab, F(ab')2, and Fab'.
When a kit of the present disclosure includes a T cell activator, in certain aspects, the T cell activator is an immune checkpoint inhibitor. Immune checkpoint inhibitors of interest include, but are not limited to, an agonist of a T cell co-stimulatory receptor, an antagonist of a T cell inhibitory signal, and/or the like. In some embodiments, the immune checkpoint inhibitor is selected from a cytotoxic T-lymphocyte-associated antigen 4 (CTLA- 4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)-dioxygenase (IDO) inhibitor, an 0X40 agonist, a glucocorticoid-induced TNFR-related protein (GITR) agonist, a CD137 agonist, and a CD40 agonist. When a kit of the present disclosure includes a T cell activator, in certain aspects, the T cell activator is a cytokine. In some embodiments, when a kit of the present disclosure includes a T cell activator, the T cell activator is an antagonist of an inhibitory immune receptor. In certain aspects, a kit of the present disclosure includes a combination (that is, two or more) of any of the T cell activators described herein.
Components of the kits may be present in separate containers, or multiple components may be present in a single container. A suitable container includes a single
tube (e.g., vial), ampoule, one or more wells of a plate (e.g., a 96-well plate, a 384-well plate, etc.), or the like.
The instructions (e.g., instructions for use (IFU)) included in the kits may be recorded on a suitable recording medium. For example, the instructions may be printed on a substrate, such as paper or plastic, etc. As such, the instructions may be present in the kits as a package insert, in the labeling of the container of the kit or components thereof (i.e., associated with the packaging or sub-packaging) etc. In other embodiments, the instructions are present as an electronic storage data file present on a suitable computer readable storage medium, e.g., portable flash drive, DVD, CD-ROM, diskette, etc. In yet other embodiments, the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source, e.g. via the internet, are provided. An example of this embodiment is a kit that includes a web address where the instructions can be viewed and/or from which the instructions can be downloaded. As with the instructions, the means for obtaining the instructions is recorded on a suitable substrate.
The following examples are offered by way of illustration and not by way of limitation.
EXPERIMENTAL
Example 1 - Co-Stimulation of Human T Cells via CD63 and CD3
The binding agents to TCR (such as anti-CD3 antibodies) and to a potential co stimulation target may be provided in tandem to purified T cells. The resulting T cell proliferation may be quantified and used to gauge the efficacy of the co-stimulation target. Shown in FIG. 1 is a graph depicting the results of T cell co-stimulation assays using anti- CD3 along with an anti-human CD63 co-stimulatory antibody (clone MX-49.129.5). Anti- 0X40 (Providence, 9B12) is used as a positive control and 1 % BSA/DPBS as a negative control.
CD4+ T cells were purified from frozen PBMCs and resuspended in RPMI media supplemented with penicillin, streptomycin, glutamine, non-essential amino acids, HEPES buffer, and sodium pyruvate. PHA-L (2 pg/ml) and IL2 (60 lll/ml) were added to the media to activate the cells, which were then incubated at 37° under 5% CO2 for 96 hours. Ninety- six-well plates were coated with goat anti-mouse IgG and then blocked with BSA. Mouse anti-human CD3 (OKT3) was prepared at 2 ng/ml in 1 % BSA/DPBS, and added to the plates. Mouse anti-human CD63 was then added in serial, two-fold dilutions between 160 ng/ml and 1 .25 ng/ml or mouse anti-human 0X40 between 320 and 2.5 ng/ml. Activated CD4+ T cells were then added to each well at 50,000 cells/well in 200 mI of media, and then incubated at 37° for 48 hours. Cells were then labeled with 5-ethynyl-2’-deoxyuridine
(EdU) at a final concentration of 10 mM, using the Click-it DNA labeling kit (Invitrogen) as per the manufacturer’s instructions for 18 hours. Cells were then treated with live-dead stain (E450; eBiosciences) and developed using the Alex647 agent (Invitrogen).
Proliferating cells were then quantified via flow cytometry.
As shown in FIG. 1 , the MX-49.129.5 anti-CD63 clone stimulated proliferation of CD4+ T cells in a dose-dependent manner, with proliferation observed at concentrations as low as 2.5 ng/ml of anti-CD63. In comparison, proliferation via the classic CD4 T cell co stimulator, 0X40, was observed at concentrations as low as 10-20 ng/ml.
In a second study using the anti-mouse CD63 antibody MA5-24169 (ThermoFisher Scientific) (the antibody employed in the in vivo mouse studies presented below and shown in FIG. 5), mouse splenocytes from a C57BI/6 mouse were enriched for CD4 T cells using a negative selection kit. Cells were treated with CD3 250 ng/ml + CD63 agonist (the MAS- 24169 antibody) at a range from 0.125-2 pg/ml. Cells were then labeled with 5-ethynyl-2’- deoxyuridine (EdU) at a final concentration of 10 mM, using the Click-it DNA labeling kit (Invitrogen) as per the manufacturer’s instructions. Cells were treated with IL-2 (50U) and harvested at day 3. Cells were then treated with live-dead stain (E450; eBiosciences) and developed using the Alex647 agent (Invitrogen). Proliferating cells were then quantified via flow cytometry. Results are shown in FIG. 2.
The data indicates that both the MX-49.129.5 and MA5-24169 anti-CD63 antibodies result in dose-dependent co-stimulation/stimulation (mitogenesis) of the CD4+ T cells when co-cultured with anti-CD3, thereby demonstrating that co-stimulation with a CD63 agonist increases T cell proliferation.
Example 2 -CD63 expression levels in cancer
The expression levels of CD63 on T cells in the tumor context were assessed by flow cytometry. Shown in FIG. 3 is flow cytometry data showing CD63 expression levels on T cells in human ovarian cancer. Increases in CD63 levels on T cells correlated with increased HLA-DR expression on conventional CD4+, Tregs, CD8+ and CD14+ cells in the tumor.
Shown in FIG. 4 is flow cytometry data showing CD63 expression levels on tumor infiltrating lymphocytes (TILs) relative to blood in a human head and neck cancer patient. An upregulation of CD63 is seen in the TILs as compared to blood from the same patient.
Example 3 - In vivo efficacy of CD63 agonist in treating cancer
In this example, mice were inoculated subcutaneously with 4e5 MCA205 tumor cells on day 0. T umor sizes were measured 2 to 3 times per week. FIG. 5, panel A, shows slight efficacy of the CD63 agonist when administered as a single therapeutic agent. Mice were
treated on days 9, 12 and 15 with either 200 pg of hamster IgG control antibody or anti mouse CD63 i.p. (ThermoFisher Clone (MA5-24169) at 50 pg, 100 pg or 200 pg dose levels. With the single agent treatment of anti-CD63 0/5 mice reject their tumors. FIG. 5, panel B, shows CD63 treatment, when administered following anti-PD1 treatment starting at day 12, leads to enhanced clearance with 1 1/15 mice rejecting tumors. Administration of the anti-PD1 treatment alone led to 3/15 cures, demonstrating that the CD63 agonist potentiates the anti-PD1 treatment. No mice treated with control IgG rejected their tumors.
Accordingly, the preceding merely illustrates the principles of the present disclosure. It will be appreciated that those skilled in the art will be able to devise various arrangements which, although not explicitly described or shown herein, embody the principles of the invention and are included within its spirit and scope. Furthermore, all examples and conditional language recited herein are principally intended to aid the reader in understanding the principles of the invention and the concepts contributed by the inventors to furthering the art, and are to be construed as being without limitation to such specifically recited examples and conditions. Moreover, all statements herein reciting principles, aspects, and embodiments of the invention as well as specific examples thereof, are intended to encompass both structural and functional equivalents thereof. Additionally, it is intended that such equivalents include both currently known equivalents and equivalents developed in the future, i.e., any elements developed that perform the same function, regardless of structure. The scope of the present invention, therefore, is not intended to be limited to the exemplary embodiments shown and described herein.
Claims
1 . A method of treating a cell proliferative disorder, comprising:
administering to a subject having a cell proliferative disorder a therapeutically effective amount of a CD63 agonist,
wherein the CD63 agonist is administered to the subject to enhance a T cell
response to abnormally proliferating cells of the cell proliferative disorder.
2. The method according to claim 1 , wherein the CD63 agonist is administered to the subject independent of the level of expression of CD63 on the abnormally proliferating cells of the cell proliferative disorder.
3. The method according to claim 1 or claim 2, wherein the subject to whom the CD63 agonist is administered is receiving a T cell activation therapy.
4. The method according to claim 3, wherein the T cell activation therapy is an immune checkpoint inhibitor therapy.
5. The method according to claim 3 or claim 4, wherein the CD63 agonist is administered to the subject to potentiate the T cell activation therapy.
6. The method according to any one of claims 1 to 5, further comprising administering to the subject a T cell activator.
7. A method of treating a cell proliferative disorder, comprising:
administering to a subject having a cell proliferative disorder:
a therapeutically effective amount of a CD63 agonist; and
a therapeutically effective amount of a T cell activator.
8. The method according to claim 7, wherein the CD63 agonist is administered to the subject to enhance a T cell response to abnormally proliferating cells of the cell proliferative disorder.
9. The method according to any one of claims 1 to 8, wherein the CD63 agonist is a small molecule.
10. The method according to any one of claims 1 to 8, wherein the CD63 agonist is a peptide or polypeptide.
1 1 . The method according to claim 10, wherein the CD63 agonist is a CD63 ligand.
12. The method according to claim 10, wherein the CD63 agonist is an antibody that specifically binds CD63.
13. The method according to claim 12, wherein the antibody that specifically binds CD63 is selected from the group consisting of: an IgG, Fv, scFv, Fab, F(ab')2, and Fab'.
14. The method according to any one of claims 6 to 13, wherein the T cell activator is an immune checkpoint inhibitor.
15. The method according to claim 14, wherein the immune checkpoint inhibitor is selected from the group consisting of: a cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)- dioxygenase (IDO) inhibitor, a T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitor, a V-domain Ig suppressor of T cell activation (VISTA) inhibitor, a B7-H3 inhibitor, and any combination thereof.
16. The method according to any one of claims 6 to 13, wherein the T cell activator is an agonist of a T cell co-stimulatory receptor.
17. The method according to claim 16, wherein the T cell activator is selected from the group consisting of: an 0X40 agonist, a glucocorticoid-induced TNFR-related protein (GITR) agonist, a CD137 agonist, and a CD40 agonist.
18. The method according to any one of claims 6 to 13, wherein the T cell activator is an antagonist of a T cell inhibitory signal.
19. The method according to any one of claims 6 to 13, wherein the T cell activator is a cytokine.
20. The method according to any one of claims 6 to 13, wherein the T cell activator is an agent that blocks immune suppressive cytokines.
21 . The method according to claim 20, wherein the agent that blocks immune suppressive cytokines is a TGF-b receptor inhibitor.
22. The method according to any one of claims 6 to 13, wherein the T cell activator is an antagonist of an inhibitory immune receptor.
23. The method according to any one of claims 6 to 22, wherein the CD63 agonist and the T cell activator are administered concurrently.
24. The method according to any one of claims 6 to 22, wherein the CD63 agonist and the T cell activator are administered sequentially.
25. The method according to claim 24, wherein the T cell activator is administered prior to the CD63 agonist.
26. The method according to any one of claims 1 to 25, wherein the cell proliferative disorder is cancer.
27. A pharmaceutical composition, comprising:
a CD63 agonist;
a T cell activator; and
a pharmaceutically acceptable excipient.
28. The pharmaceutical composition of claim 27, wherein the CD63 agonist is a small molecule.
29. The pharmaceutical composition of claim 27, wherein the CD63 agonist is a peptide or polypeptide.
30. The pharmaceutical composition of claim 29, wherein the CD63 agonist is a CD63 ligand.
31 . The pharmaceutical composition of claim 29, wherein the CD63 agonist is an antibody that specifically binds CD63.
32. The pharmaceutical composition of 31 , wherein the antibody that specifically binds CD63 is selected from the group consisting of: an IgG, Fv, scFv, Fab, F(ab')2, and Fab'.
33. The pharmaceutical composition of any one of claims 27 to 32, wherein the T cell activator is an immune checkpoint inhibitor.
34. The pharmaceutical composition of claim 33, wherein the immune checkpoint inhibitor is selected from the group consisting of: a cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, an indoleamine (2,3)-dioxygenase (IDO) inhibitor, a T cell immunoreceptor with Ig and ITIM domains (TIGIT) inhibitor, a V-domain Ig suppressor of T cell activation (VISTA) inhibitor, a B7-H3 inhibitor, and any combination thereof.
35. The pharmaceutical composition of any one of claims 27 to 32, wherein the T cell activator is an agonist of a T cell co-stimulatory receptor.
36. The pharmaceutical composition of claim 35, wherein the T cell activator is selected from the group consisting of: an 0X40 agonist, a glucocorticoid-induced TNFFt- related protein (GITR) agonist, a CD137 agonist, and a CD40 agonist.
37. The pharmaceutical composition of any one of claims 27 to 32, wherein the T cell activator is an antagonist of a T cell inhibitory signal.
38. The pharmaceutical composition of any one of claims 27 to 32, wherein the T cell activator is a cytokine.
39. The pharmaceutical composition of any one of claims 27 to 32, wherein the T cell activator is an agent that blocks immune suppressive cytokines.
40. The pharmaceutical composition of claim 39, wherein the agent that blocks immune suppressive cytokines is a TGF-b receptor inhibitor.
41 . The pharmaceutical composition of any one of claims 27 to 32, wherein the T cell activator is an antagonist of an inhibitory immune receptor.
42. A kit, comprising:
a pharmaceutical composition comprising a CD63 agonist; and
instructions for administering the pharmaceutical composition to a subject having a cell proliferative disorder.
43. The kit of claim 42, comprising instructions for administering the pharmaceutical composition to a subject receiving a T cell activation therapy.
44. The kit of claim 43, comprising instructions for administering the CD63 agonist to the subject to potentiate the T cell activation therapy.
45. The kit of any one of claims 42 to 44, wherein the pharmaceutical composition further comprises a T cell activator.
46. A kit, comprising:
the pharmaceutical composition of any one of claims 27 to 41 ; and
instructions for administering the pharmaceutical composition to a subject having a cell proliferative disorder.
47. The kit of any one of claims 42 to 46, wherein the kit comprises the pharmaceutical composition in one or more unit dosages.
48. The kit of any one of claims 42 to 46, wherein the kit comprises the pharmaceutical composition in two or more unit dosages.
49. The kit of any one of claims 42 to 48, comprising instructions for administering the pharmaceutical composition to a subject having a cell proliferative disorder independent of the level of expression of CD63 on the abnormally proliferating cells of the cell proliferative disorder.
50. A kit, comprising:
a CD63 agonist;
a T cell activator; and
instructions for administering the CD63 agonist and T cell activator to a subject having a cell proliferative disorder.
51 . The kit of claim 50, wherein the kit comprises the CD63 agonist and the T cell activator in one or more unit dosages.
52. The kit of claim 50, wherein the kit comprises the CD63 agonist and the T cell activator in two or more unit dosages.
53. The kit of any one of claims 50 to 52, wherein the CD63 agonist and T cell activator are present in separate containers.
54. The kit of any one of claims 50 to 53, wherein the instructions are for administering the CD63 agonist and the T cell activator to a subject having a cell proliferative disorder independent of the level of expression of CD63 on the abnormally proliferating cells of the cell proliferative disorder.
55. The kit of any one of claims 42 to 54, wherein the CD63 agonist is a small molecule.
56. The kit of any one of claims 42 to 54, wherein the CD63 agonist is a peptide or polypeptide.
57. The kit of claim 56, wherein the CD63 agonist is a CD63 ligand.
58. The kit of claim 56, wherein the CD63 agonist is an antibody that specifically binds CD63.
59. The kit of claim 58, wherein the antibody that specifically binds CD63 is selected from the group consisting of: an IgG, Fv, scFv, Fab, F(ab')2, and Fab'.
60. The kit of any one of claims 45 to 59, wherein the T cell activator is an immune checkpoint inhibitor.
61 . The kit of claim 60, wherein the immune checkpoint inhibitor is selected from the group consisting of: a cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) inhibitor, a programmed cell death-1 (PD-1 ) inhibitor, a programmed cell death ligand-1 (PD-L1 ) inhibitor, a lymphocyte activation gene-3 (LAG-3) inhibitor, a T-cell immunoglobulin domain and mucin domain 3 (TIM-3) inhibitor, and an indoleamine (2,3)-dioxygenase (IDO) inhibitor.
62. The kit of any one of claims 45 to 59, wherein the T cell activator is an agonist of a T cell co-stimulatory receptor.
63. The kit of claim 62, wherein the T cell activator is selected from the group consisting of: an 0X40 agonist, a glucocorticoid-induced TNFR-related protein (GITR) agonist, a CD137 agonist, and a CD40 agonist.
64. The kit of any one of claims 45 to 59, wherein the T cell activator is an antagonist of a T cell inhibitory signal.
65. The kit of any one of claims 45 to 59, wherein the T cell activator is a cytokine.
66. The kit of any one of claims 45 to 59, wherein the T cell activator is an agent that blocks immune suppressive cytokines.
67. The kit of claim 66, wherein the agent that blocks immune suppressive cytokines is a TGF-b receptor inhibitor.
68. The kit of any one of claims 45 to 59, wherein the T cell activator is an antagonist of an inhibitory immune receptor.
69. The kit of any one of claims 42 to 68, wherein the cell proliferative disorder is cancer.
Priority Applications (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202080057761.4A CN114286686A (en) | 2019-07-24 | 2020-07-23 | Methods and compositions related to CD63 agonists |
US17/629,295 US20220242966A1 (en) | 2019-07-24 | 2020-07-23 | Cd63 agonist-related methods and compositions |
EP20844284.8A EP4003402A4 (en) | 2019-07-24 | 2020-07-23 | Cd63 agonist-related methods and compositions |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201962878069P | 2019-07-24 | 2019-07-24 | |
US62/878,069 | 2019-07-24 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2021016451A1 true WO2021016451A1 (en) | 2021-01-28 |
Family
ID=74192635
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2020/043264 WO2021016451A1 (en) | 2019-07-24 | 2020-07-23 | Cd63 agonist-related methods and compositions |
Country Status (4)
Country | Link |
---|---|
US (1) | US20220242966A1 (en) |
EP (1) | EP4003402A4 (en) |
CN (1) | CN114286686A (en) |
WO (1) | WO2021016451A1 (en) |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20040258693A1 (en) * | 2003-01-21 | 2004-12-23 | Young David S. F. | Cytotoxicity mediation of cells evidencing surface expression of CD63 |
US20160115229A1 (en) * | 2013-05-15 | 2016-04-28 | The Translational Genomics Research Intsitute | Hybridoma Clones, Monoclonal Antibodies, and Methods of Use |
US20190015508A1 (en) * | 2017-07-11 | 2019-01-17 | Compass Therapeutics Llc | Agonist antibodies that bind human cd137 and uses thereof |
WO2019100052A2 (en) * | 2017-11-20 | 2019-05-23 | Compass Therapeutics Llc | Cd137 antibodies and tumor antigen-targeting antibodies and uses thereof |
Family Cites Families (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7534429B2 (en) * | 2000-11-29 | 2009-05-19 | Hoffmann-La Roche Inc. | Cytotoxicity mediation of cells evidencing surface expression of CD63 |
-
2020
- 2020-07-23 EP EP20844284.8A patent/EP4003402A4/en active Pending
- 2020-07-23 WO PCT/US2020/043264 patent/WO2021016451A1/en active Application Filing
- 2020-07-23 US US17/629,295 patent/US20220242966A1/en active Pending
- 2020-07-23 CN CN202080057761.4A patent/CN114286686A/en active Pending
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20040258693A1 (en) * | 2003-01-21 | 2004-12-23 | Young David S. F. | Cytotoxicity mediation of cells evidencing surface expression of CD63 |
US20160115229A1 (en) * | 2013-05-15 | 2016-04-28 | The Translational Genomics Research Intsitute | Hybridoma Clones, Monoclonal Antibodies, and Methods of Use |
US20190015508A1 (en) * | 2017-07-11 | 2019-01-17 | Compass Therapeutics Llc | Agonist antibodies that bind human cd137 and uses thereof |
WO2019100052A2 (en) * | 2017-11-20 | 2019-05-23 | Compass Therapeutics Llc | Cd137 antibodies and tumor antigen-targeting antibodies and uses thereof |
Non-Patent Citations (2)
Title |
---|
"Atezolizumab", WIKIPEDIA, 4 June 2019 (2019-06-04), pages 1 - 4, XP055789495, Retrieved from the Internet <URL:https://en.wikipedia.org/w/index.php?title=Atezolizumab&oldid=900323869> * |
See also references of EP4003402A4 * |
Also Published As
Publication number | Publication date |
---|---|
CN114286686A (en) | 2022-04-05 |
EP4003402A4 (en) | 2023-11-22 |
EP4003402A1 (en) | 2022-06-01 |
US20220242966A1 (en) | 2022-08-04 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7565797B2 (en) | Anti-CD25 for tumor-specific cell depletion | |
US20210403589A1 (en) | Combination therapies with anti cd40 antibodies | |
US9828432B2 (en) | Cancer treatment and monitoring methods using OX40 agonists | |
TWI359671B (en) | Cd40 antibody formulation and methods | |
AU2010284846B2 (en) | IL1RAP expression on acute and chronic myeloid leukemia cells | |
JP2020530289A (en) | Methods of Using Anti-CD39 Antibodies, Compositions Containing Anti-CD39 Antibodies, and Anti-CD39 Antibodies | |
JP2020530495A (en) | Compositions and Methods for Incapacitating Bone Marrow Cells Expressing TREM1 | |
JP2016534090A (en) | Medical use of CD38 agonist | |
JP2019527204A (en) | Suppressive immunoreceptor inhibition method and composition | |
Jeong et al. | Novel anti-4-1BB× PD-L1 bispecific antibody augments anti-tumor immunity through tumor-directed T-cell activation and checkpoint blockade | |
JP2023116681A (en) | CD38 regulatory antibody | |
JP2018534266A (en) | Combination therapy using inhibitors of human growth differentiation factor 15 (GDF-15) and immune checkpoint blockers | |
JP2021524269A (en) | Mesoterin and CD137 binding molecules | |
KR102574071B1 (en) | Anti-GITR antigen-binding proteins and methods of use thereof | |
JP7237951B2 (en) | CD38 regulatory antibody | |
WO2020185739A1 (en) | Anti-icos antibodies for the treatment of cancer | |
JP2021518138A (en) | Anti-IL-27 antibody and its use | |
JP7299873B2 (en) | CD38 antibody | |
US20220242966A1 (en) | Cd63 agonist-related methods and compositions | |
JP7224338B2 (en) | CD38 regulatory antibody | |
US11299545B2 (en) | Anti-HLA-C antibodies and uses thereof | |
US20210070858A1 (en) | CD55-Binding Agent-Related Methods and Compositions | |
RU2795625C2 (en) | Antigen-binding proteins against gitr and methods of their use | |
WO2023222827A1 (en) | Anti-complement factor h antibodies |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 20844284 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2020844284 Country of ref document: EP |
|
ENP | Entry into the national phase |
Ref document number: 2020844284 Country of ref document: EP Effective date: 20220224 |