WO2024173281A1 - Antibody-drug conjugate for cancer treatment - Google Patents
Antibody-drug conjugate for cancer treatment Download PDFInfo
- Publication number
- WO2024173281A1 WO2024173281A1 PCT/US2024/015450 US2024015450W WO2024173281A1 WO 2024173281 A1 WO2024173281 A1 WO 2024173281A1 US 2024015450 W US2024015450 W US 2024015450W WO 2024173281 A1 WO2024173281 A1 WO 2024173281A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- her2
- cancer
- antibody
- patients
- treatment
- Prior art date
Links
- 206010028980 Neoplasm Diseases 0.000 title claims abstract description 79
- 229940049595 antibody-drug conjugate Drugs 0.000 title claims abstract description 70
- 239000000611 antibody drug conjugate Substances 0.000 title claims abstract description 69
- 238000011282 treatment Methods 0.000 title claims description 68
- 201000011510 cancer Diseases 0.000 title claims description 50
- 229960003649 eribulin Drugs 0.000 claims abstract description 25
- 238000000034 method Methods 0.000 claims abstract description 9
- 229960000575 trastuzumab Drugs 0.000 claims abstract description 9
- UFNVPOGXISZXJD-XJPMSQCNSA-N eribulin Chemical compound C([C@H]1CC[C@@H]2O[C@@H]3[C@H]4O[C@H]5C[C@](O[C@H]4[C@H]2O1)(O[C@@H]53)CC[C@@H]1O[C@H](C(C1)=C)CC1)C(=O)C[C@@H]2[C@@H](OC)[C@@H](C[C@H](O)CN)O[C@H]2C[C@@H]2C(=C)[C@H](C)C[C@H]1O2 UFNVPOGXISZXJD-XJPMSQCNSA-N 0.000 claims abstract 2
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 claims description 57
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 claims description 56
- 206010006187 Breast cancer Diseases 0.000 claims description 33
- 208000026310 Breast neoplasm Diseases 0.000 claims description 31
- 239000003814 drug Substances 0.000 claims description 16
- 208000002154 non-small cell lung carcinoma Diseases 0.000 claims description 14
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 claims description 10
- 230000002496 gastric effect Effects 0.000 claims description 9
- 206010044412 transitional cell carcinoma Diseases 0.000 claims description 8
- 208000023747 urothelial carcinoma Diseases 0.000 claims description 8
- 210000003236 esophagogastric junction Anatomy 0.000 claims description 7
- 208000004091 Parotid Neoplasms Diseases 0.000 claims description 3
- 238000003745 diagnosis Methods 0.000 claims description 3
- 201000001219 parotid gland cancer Diseases 0.000 claims description 3
- 239000000203 mixture Substances 0.000 abstract description 15
- 210000004027 cell Anatomy 0.000 description 36
- 229940049679 trastuzumab deruxtecan Drugs 0.000 description 29
- UFNVPOGXISZXJD-JBQZKEIOSA-N eribulin Chemical compound C([C@H]1CC[C@@H]2O[C@@H]3[C@H]4O[C@@H]5C[C@](O[C@H]4[C@H]2O1)(O[C@@H]53)CC[C@@H]1O[C@H](C(C1)=C)CC1)C(=O)C[C@@H]2[C@@H](OC)[C@@H](C[C@H](O)CN)O[C@H]2C[C@@H]2C(=C)[C@H](C)C[C@H]1O2 UFNVPOGXISZXJD-JBQZKEIOSA-N 0.000 description 22
- 230000014509 gene expression Effects 0.000 description 15
- 208000017891 HER2 positive breast carcinoma Diseases 0.000 description 14
- 230000004614 tumor growth Effects 0.000 description 14
- 230000003902 lesion Effects 0.000 description 13
- 230000000694 effects Effects 0.000 description 12
- 208000029523 Interstitial Lung disease Diseases 0.000 description 10
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 10
- 230000035772 mutation Effects 0.000 description 10
- 230000004044 response Effects 0.000 description 10
- 229960001612 trastuzumab emtansine Drugs 0.000 description 10
- 230000037396 body weight Effects 0.000 description 9
- 201000010099 disease Diseases 0.000 description 9
- 229940079593 drug Drugs 0.000 description 8
- 241000282693 Cercopithecidae Species 0.000 description 7
- 241000282567 Macaca fascicularis Species 0.000 description 7
- 208000005718 Stomach Neoplasms Diseases 0.000 description 7
- 230000000259 anti-tumor effect Effects 0.000 description 7
- 238000011156 evaluation Methods 0.000 description 7
- 230000037449 immunogenic cell death Effects 0.000 description 7
- 208000020816 lung neoplasm Diseases 0.000 description 7
- 150000003839 salts Chemical class 0.000 description 7
- 230000001225 therapeutic effect Effects 0.000 description 7
- 238000002560 therapeutic procedure Methods 0.000 description 7
- 239000003053 toxin Substances 0.000 description 7
- 231100000765 toxin Toxicity 0.000 description 7
- 108700012359 toxins Proteins 0.000 description 7
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 6
- 230000000981 bystander Effects 0.000 description 6
- 206010017758 gastric cancer Diseases 0.000 description 6
- 238000001990 intravenous administration Methods 0.000 description 6
- 201000005202 lung cancer Diseases 0.000 description 6
- 230000007246 mechanism Effects 0.000 description 6
- 230000009467 reduction Effects 0.000 description 6
- 238000011160 research Methods 0.000 description 6
- 201000011549 stomach cancer Diseases 0.000 description 6
- 210000004881 tumor cell Anatomy 0.000 description 6
- 206010005003 Bladder cancer Diseases 0.000 description 5
- 206010055113 Breast cancer metastatic Diseases 0.000 description 5
- 206010009944 Colon cancer Diseases 0.000 description 5
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 5
- 101150029707 ERBB2 gene Proteins 0.000 description 5
- 206010053759 Growth retardation Diseases 0.000 description 5
- 241000699670 Mus sp. Species 0.000 description 5
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 5
- 230000009471 action Effects 0.000 description 5
- 230000003321 amplification Effects 0.000 description 5
- 239000000427 antigen Substances 0.000 description 5
- 108091007433 antigens Proteins 0.000 description 5
- 102000036639 antigens Human genes 0.000 description 5
- 230000008878 coupling Effects 0.000 description 5
- 238000010168 coupling process Methods 0.000 description 5
- 238000005859 coupling reaction Methods 0.000 description 5
- 239000003937 drug carrier Substances 0.000 description 5
- 210000000987 immune system Anatomy 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 238000003199 nucleic acid amplification method Methods 0.000 description 5
- 239000008194 pharmaceutical composition Substances 0.000 description 5
- 238000011301 standard therapy Methods 0.000 description 5
- 201000005112 urinary bladder cancer Diseases 0.000 description 5
- PLXLYXLUCNZSAA-QLXKLKPCSA-N CC[C@@]1(O)C(=O)OCC2=C1C=C1N(CC3=C1N=C1C=C(F)C(C)=C4CC[C@H](NC(=O)CO)C3=C14)C2=O Chemical compound CC[C@@]1(O)C(=O)OCC2=C1C=C1N(CC3=C1N=C1C=C(F)C(C)=C4CC[C@H](NC(=O)CO)C3=C14)C2=O PLXLYXLUCNZSAA-QLXKLKPCSA-N 0.000 description 4
- 206010057269 Mucoepidermoid carcinoma Diseases 0.000 description 4
- 210000000170 cell membrane Anatomy 0.000 description 4
- 208000006990 cholangiocarcinoma Diseases 0.000 description 4
- 230000001472 cytotoxic effect Effects 0.000 description 4
- 230000003013 cytotoxicity Effects 0.000 description 4
- 231100000135 cytotoxicity Toxicity 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- -1 for example Chemical class 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 206010023841 laryngeal neoplasm Diseases 0.000 description 4
- 210000004072 lung Anatomy 0.000 description 4
- 208000037821 progressive disease Diseases 0.000 description 4
- 230000004083 survival effect Effects 0.000 description 4
- 230000001988 toxicity Effects 0.000 description 4
- 231100000419 toxicity Toxicity 0.000 description 4
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 101100339431 Arabidopsis thaliana HMGB2 gene Proteins 0.000 description 3
- 102000004082 Calreticulin Human genes 0.000 description 3
- 108090000549 Calreticulin Proteins 0.000 description 3
- 206010008342 Cervix carcinoma Diseases 0.000 description 3
- 206010014733 Endometrial cancer Diseases 0.000 description 3
- 206010014759 Endometrial neoplasm Diseases 0.000 description 3
- 108700010013 HMGB1 Proteins 0.000 description 3
- 101150021904 HMGB1 gene Proteins 0.000 description 3
- 102100037907 High mobility group protein B1 Human genes 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- SJRJJKPEHAURKC-UHFFFAOYSA-N N-Methylmorpholine Chemical compound CN1CCOCC1 SJRJJKPEHAURKC-UHFFFAOYSA-N 0.000 description 3
- 206010033128 Ovarian cancer Diseases 0.000 description 3
- 206010061535 Ovarian neoplasm Diseases 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 102000004243 Tubulin Human genes 0.000 description 3
- 108090000704 Tubulin Proteins 0.000 description 3
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- 150000001412 amines Chemical class 0.000 description 3
- 210000001185 bone marrow Anatomy 0.000 description 3
- 201000010881 cervical cancer Diseases 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 231100000433 cytotoxic Toxicity 0.000 description 3
- 230000034994 death Effects 0.000 description 3
- 231100000517 death Toxicity 0.000 description 3
- 230000007423 decrease Effects 0.000 description 3
- 238000000684 flow cytometry Methods 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 238000007918 intramuscular administration Methods 0.000 description 3
- 238000004020 luminiscence type Methods 0.000 description 3
- 210000004698 lymphocyte Anatomy 0.000 description 3
- 210000000440 neutrophil Anatomy 0.000 description 3
- 231100000191 repeated dose toxicity Toxicity 0.000 description 3
- 238000012552 review Methods 0.000 description 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 229940121358 tyrosine kinase inhibitor Drugs 0.000 description 3
- PAMIQIKDUOTOBW-UHFFFAOYSA-N 1-methylpiperidine Chemical compound CN1CCCCC1 PAMIQIKDUOTOBW-UHFFFAOYSA-N 0.000 description 2
- QGZKDVFQNNGYKY-UHFFFAOYSA-N Ammonia Chemical compound N QGZKDVFQNNGYKY-UHFFFAOYSA-N 0.000 description 2
- 102000005600 Cathepsins Human genes 0.000 description 2
- 108010084457 Cathepsins Proteins 0.000 description 2
- 241001146155 Emilia Species 0.000 description 2
- 235000002139 Emilia sonchifolia Nutrition 0.000 description 2
- QUSNBJAOOMFDIB-UHFFFAOYSA-N Ethylamine Chemical compound CCN QUSNBJAOOMFDIB-UHFFFAOYSA-N 0.000 description 2
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 2
- 206010066896 HER-2 positive gastric cancer Diseases 0.000 description 2
- ZBLLGPUWGCOJNG-UHFFFAOYSA-N Halichondrin B Natural products CC1CC2(CC(C)C3OC4(CC5OC6C(CC5O4)OC7CC8OC9CCC%10OC(CC(C(C9)C8=C)C%11%12CC%13OC%14C(OC%15CCC(CC(=O)OC7C6C)OC%15C%14O%11)C%13O%12)CC%10=C)CC3O2)OC%16OC(CC1%16)C(O)CC(O)CO ZBLLGPUWGCOJNG-UHFFFAOYSA-N 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- 206010023825 Laryngeal cancer Diseases 0.000 description 2
- 206010061523 Lip and/or oral cavity cancer Diseases 0.000 description 2
- 206010062038 Lip neoplasm Diseases 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 2
- 208000003445 Mouth Neoplasms Diseases 0.000 description 2
- 206010029098 Neoplasm skin Diseases 0.000 description 2
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- 206010060862 Prostate cancer Diseases 0.000 description 2
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 2
- 101710100968 Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 2
- 208000015634 Rectal Neoplasms Diseases 0.000 description 2
- 208000006265 Renal cell carcinoma Diseases 0.000 description 2
- 208000004337 Salivary Gland Neoplasms Diseases 0.000 description 2
- 206010061934 Salivary gland cancer Diseases 0.000 description 2
- 208000000453 Skin Neoplasms Diseases 0.000 description 2
- 208000000102 Squamous Cell Carcinoma of Head and Neck Diseases 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- 229940123237 Taxane Drugs 0.000 description 2
- 206010062129 Tongue neoplasm Diseases 0.000 description 2
- 206010044002 Tonsil cancer Diseases 0.000 description 2
- 208000006842 Tonsillar Neoplasms Diseases 0.000 description 2
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 2
- 208000006593 Urologic Neoplasms Diseases 0.000 description 2
- 208000002495 Uterine Neoplasms Diseases 0.000 description 2
- IEDXPSOJFSVCKU-HOKPPMCLSA-N [4-[[(2S)-5-(carbamoylamino)-2-[[(2S)-2-[6-(2,5-dioxopyrrolidin-1-yl)hexanoylamino]-3-methylbutanoyl]amino]pentanoyl]amino]phenyl]methyl N-[(2S)-1-[[(2S)-1-[[(3R,4S,5S)-1-[(2S)-2-[(1R,2R)-3-[[(1S,2R)-1-hydroxy-1-phenylpropan-2-yl]amino]-1-methoxy-2-methyl-3-oxopropyl]pyrrolidin-1-yl]-3-methoxy-5-methyl-1-oxoheptan-4-yl]-methylamino]-3-methyl-1-oxobutan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]-N-methylcarbamate Chemical compound CC[C@H](C)[C@@H]([C@@H](CC(=O)N1CCC[C@H]1[C@H](OC)[C@@H](C)C(=O)N[C@H](C)[C@@H](O)c1ccccc1)OC)N(C)C(=O)[C@@H](NC(=O)[C@H](C(C)C)N(C)C(=O)OCc1ccc(NC(=O)[C@H](CCCNC(N)=O)NC(=O)[C@@H](NC(=O)CCCCCN2C(=O)CCC2=O)C(C)C)cc1)C(C)C IEDXPSOJFSVCKU-HOKPPMCLSA-N 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 230000002927 anti-mitotic effect Effects 0.000 description 2
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 2
- 210000000013 bile duct Anatomy 0.000 description 2
- 210000003445 biliary tract Anatomy 0.000 description 2
- 229960000106 biosimilars Drugs 0.000 description 2
- 238000012512 characterization method Methods 0.000 description 2
- 238000003776 cleavage reaction Methods 0.000 description 2
- 238000000576 coating method Methods 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 239000002254 cytotoxic agent Substances 0.000 description 2
- XBDQKXXYIPTUBI-UHFFFAOYSA-N dimethylselenoniopropionate Natural products CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 2
- 229940121550 disitamab vedotin Drugs 0.000 description 2
- 230000007783 downstream signaling Effects 0.000 description 2
- QAMYWGZHLCQOOJ-PWIVHLLHSA-N eribulin mesylate Chemical compound CS(O)(=O)=O.C([C@H]1CC[C@@H]2O[C@@H]3[C@H]4O[C@H]5C[C@](O[C@H]4[C@H]2O1)(O[C@@H]53)CC[C@@H]1O[C@H](C(C1)=C)CC1)C(=O)C[C@@H]2[C@@H](OC)[C@@H](C[C@H](O)CN)O[C@H]2C[C@@H]2C(=C)[C@H](C)C[C@H]1O2 QAMYWGZHLCQOOJ-PWIVHLLHSA-N 0.000 description 2
- 229960000439 eribulin mesylate Drugs 0.000 description 2
- 201000007492 gastroesophageal junction adenocarcinoma Diseases 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- FXNFULJVOQMBCW-VZBLNRDYSA-N halichondrin b Chemical compound O([C@@H]1[C@@H](C)[C@@H]2O[C@@H]3C[C@@]4(O[C@H]5[C@@H](C)C[C@@]6(C[C@@H]([C@@H]7O[C@@H](C[C@@H]7O6)[C@@H](O)C[C@@H](O)CO)C)O[C@H]5C4)O[C@@H]3C[C@@H]2O[C@H]1C[C@@H]1C(=C)[C@H](C)C[C@@H](O1)CC[C@H]1C(=C)C[C@@H](O1)CC1)C(=O)C[C@H](O2)CC[C@H]3[C@H]2[C@H](O2)[C@@H]4O[C@@H]5C[C@@]21O[C@@H]5[C@@H]4O3 FXNFULJVOQMBCW-VZBLNRDYSA-N 0.000 description 2
- 208000014829 head and neck neoplasm Diseases 0.000 description 2
- 201000000459 head and neck squamous cell carcinoma Diseases 0.000 description 2
- 238000004191 hydrophobic interaction chromatography Methods 0.000 description 2
- 238000013388 immunohistochemistry analysis Methods 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 230000008595 infiltration Effects 0.000 description 2
- 238000001764 infiltration Methods 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 239000007951 isotonicity adjuster Substances 0.000 description 2
- 230000002147 killing effect Effects 0.000 description 2
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 description 2
- 201000004959 laryngeal benign neoplasm Diseases 0.000 description 2
- 201000004962 larynx cancer Diseases 0.000 description 2
- 210000000265 leukocyte Anatomy 0.000 description 2
- 238000010859 live-cell imaging Methods 0.000 description 2
- 201000007270 liver cancer Diseases 0.000 description 2
- 208000014018 liver neoplasm Diseases 0.000 description 2
- 210000001165 lymph node Anatomy 0.000 description 2
- 230000002132 lysosomal effect Effects 0.000 description 2
- 239000003120 macrolide antibiotic agent Substances 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 230000010534 mechanism of action Effects 0.000 description 2
- 230000001394 metastastic effect Effects 0.000 description 2
- 206010061289 metastatic neoplasm Diseases 0.000 description 2
- 201000010879 mucinous adenocarcinoma Diseases 0.000 description 2
- 208000023983 oral cavity neoplasm Diseases 0.000 description 2
- 210000003254 palate Anatomy 0.000 description 2
- 201000002528 pancreatic cancer Diseases 0.000 description 2
- 208000008443 pancreatic carcinoma Diseases 0.000 description 2
- 210000004303 peritoneum Anatomy 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 206010038038 rectal cancer Diseases 0.000 description 2
- 201000001275 rectum cancer Diseases 0.000 description 2
- 230000000306 recurrent effect Effects 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 230000007017 scission Effects 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 238000004904 shortening Methods 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 210000001541 thymus gland Anatomy 0.000 description 2
- 231100000607 toxicokinetics Toxicity 0.000 description 2
- 231100000041 toxicology testing Toxicity 0.000 description 2
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 2
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 2
- 208000029584 urinary system neoplasm Diseases 0.000 description 2
- 206010046766 uterine cancer Diseases 0.000 description 2
- BJEPYKJPYRNKOW-REOHCLBHSA-N (S)-malic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O BJEPYKJPYRNKOW-REOHCLBHSA-N 0.000 description 1
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 1
- LBLYYCQCTBFVLH-UHFFFAOYSA-N 2-Methylbenzenesulfonic acid Chemical compound CC1=CC=CC=C1S(O)(=O)=O LBLYYCQCTBFVLH-UHFFFAOYSA-N 0.000 description 1
- BMYNFMYTOJXKLE-UHFFFAOYSA-N 3-azaniumyl-2-hydroxypropanoate Chemical compound NCC(O)C(O)=O BMYNFMYTOJXKLE-UHFFFAOYSA-N 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- GAGWJHPBXLXJQN-UORFTKCHSA-N Capecitabine Chemical compound C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](C)O1 GAGWJHPBXLXJQN-UORFTKCHSA-N 0.000 description 1
- GAGWJHPBXLXJQN-UHFFFAOYSA-N Capecitabine Natural products C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1C1C(O)C(O)C(C)O1 GAGWJHPBXLXJQN-UHFFFAOYSA-N 0.000 description 1
- 108010078791 Carrier Proteins Proteins 0.000 description 1
- 102000004225 Cathepsin B Human genes 0.000 description 1
- 108090000712 Cathepsin B Proteins 0.000 description 1
- 230000004544 DNA amplification Effects 0.000 description 1
- 229940123780 DNA topoisomerase I inhibitor Drugs 0.000 description 1
- 206010011906 Death Diseases 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 1
- XBPCUCUWBYBCDP-UHFFFAOYSA-N Dicyclohexylamine Chemical compound C1CCCCC1NC1CCCCC1 XBPCUCUWBYBCDP-UHFFFAOYSA-N 0.000 description 1
- 206010059866 Drug resistance Diseases 0.000 description 1
- 102000001301 EGF receptor Human genes 0.000 description 1
- 108060006698 EGF receptor Proteins 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- PIICEJLVQHRZGT-UHFFFAOYSA-N Ethylenediamine Chemical compound NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 101150054472 HER2 gene Proteins 0.000 description 1
- 238000011460 HER2-targeted therapy Methods 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000851181 Homo sapiens Epidermal growth factor receptor Proteins 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
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- 239000002136 L01XE07 - Lapatinib Substances 0.000 description 1
- XNRVGTHNYCNCFF-UHFFFAOYSA-N Lapatinib ditosylate monohydrate Chemical compound O.CC1=CC=C(S(O)(=O)=O)C=C1.CC1=CC=C(S(O)(=O)=O)C=C1.O1C(CNCCS(=O)(=O)C)=CC=C1C1=CC=C(N=CN=C2NC=3C=C(Cl)C(OCC=4C=C(F)C=CC=4)=CC=3)C2=C1 XNRVGTHNYCNCFF-UHFFFAOYSA-N 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
- 229930126263 Maytansine Natural products 0.000 description 1
- 102000013013 Member 2 Subfamily G ATP Binding Cassette Transporter Human genes 0.000 description 1
- 108010090306 Member 2 Subfamily G ATP Binding Cassette Transporter Proteins 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 102000029749 Microtubule Human genes 0.000 description 1
- 108091022875 Microtubule Proteins 0.000 description 1
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 1
- UEEJHVSXFDXPFK-UHFFFAOYSA-N N-dimethylaminoethanol Chemical compound CN(C)CCO UEEJHVSXFDXPFK-UHFFFAOYSA-N 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 206010035664 Pneumonia Diseases 0.000 description 1
- 206010035742 Pneumonitis Diseases 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- OFOBLEOULBTSOW-UHFFFAOYSA-N Propanedioic acid Natural products OC(=O)CC(O)=O OFOBLEOULBTSOW-UHFFFAOYSA-N 0.000 description 1
- 206010061924 Pulmonary toxicity Diseases 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric acid Natural products [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Triethanolamine Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 description 1
- 229940122429 Tubulin inhibitor Drugs 0.000 description 1
- 229940122530 Tubulin polymerization inhibitor Drugs 0.000 description 1
- 229940122803 Vinca alkaloid Drugs 0.000 description 1
- 239000003070 absorption delaying agent Substances 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 229910052783 alkali metal Inorganic materials 0.000 description 1
- 229910052784 alkaline earth metal Inorganic materials 0.000 description 1
- BJEPYKJPYRNKOW-UHFFFAOYSA-N alpha-hydroxysuccinic acid Natural products OC(=O)C(O)CC(O)=O BJEPYKJPYRNKOW-UHFFFAOYSA-N 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 229910021529 ammonia Inorganic materials 0.000 description 1
- 150000003863 ammonium salts Chemical class 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 229940124691 antibody therapeutics Drugs 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 239000002246 antineoplastic agent 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
- 238000003556 assay Methods 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 239000002585 base Substances 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- SRSXLGNVWSONIS-UHFFFAOYSA-N benzenesulfonic acid Chemical compound OS(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-N 0.000 description 1
- 229940092714 benzenesulfonic acid Drugs 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- 208000036815 beta tubulin Diseases 0.000 description 1
- 201000009036 biliary tract cancer Diseases 0.000 description 1
- 208000020790 biliary tract neoplasm Diseases 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 229960000074 biopharmaceutical Drugs 0.000 description 1
- 206010005084 bladder transitional cell carcinoma Diseases 0.000 description 1
- 201000001528 bladder urothelial carcinoma Diseases 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 238000010241 blood sampling Methods 0.000 description 1
- 201000010386 breast liposarcoma Diseases 0.000 description 1
- 159000000007 calcium salts Chemical class 0.000 description 1
- 229960004117 capecitabine Drugs 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 125000004432 carbon atom Chemical group C* 0.000 description 1
- 150000001735 carboxylic acids Chemical class 0.000 description 1
- 230000009084 cardiovascular function Effects 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 238000002512 chemotherapy Methods 0.000 description 1
- 229940044683 chemotherapy drug Drugs 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- 238000011210 chromatographic step Methods 0.000 description 1
- 235000015165 citric acid Nutrition 0.000 description 1
- 229960002173 citrulline Drugs 0.000 description 1
- 230000015271 coagulation Effects 0.000 description 1
- 238000005345 coagulation Methods 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 150000001875 compounds Chemical class 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
- 210000000172 cytosol Anatomy 0.000 description 1
- 229940127089 cytotoxic agent Drugs 0.000 description 1
- 231100000599 cytotoxic agent Toxicity 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 229960002887 deanol Drugs 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- ZBCBWPMODOFKDW-UHFFFAOYSA-N diethanolamine Chemical compound OCCNCCO ZBCBWPMODOFKDW-UHFFFAOYSA-N 0.000 description 1
- HPNMFZURTQLUMO-UHFFFAOYSA-N diethylamine Chemical compound CCNCC HPNMFZURTQLUMO-UHFFFAOYSA-N 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- LFQCJSBXBZRMTN-OAQYLSRUSA-N diflomotecan Chemical compound CC[C@@]1(O)CC(=O)OCC(C2=O)=C1C=C1N2CC2=CC3=CC(F)=C(F)C=C3N=C21 LFQCJSBXBZRMTN-OAQYLSRUSA-N 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 239000006185 dispersion Substances 0.000 description 1
- 239000002612 dispersion medium Substances 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 238000009513 drug distribution Methods 0.000 description 1
- 230000036267 drug metabolism Effects 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 description 1
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 108700020302 erbB-2 Genes Proteins 0.000 description 1
- 210000000267 erythroid cell Anatomy 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 210000003754 fetus Anatomy 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 235000012631 food intake Nutrition 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 239000001530 fumaric acid Substances 0.000 description 1
- 235000011087 fumaric acid Nutrition 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 231100000226 haematotoxicity Toxicity 0.000 description 1
- 230000009931 harmful effect Effects 0.000 description 1
- 229940022353 herceptin Drugs 0.000 description 1
- 102000051957 human ERBB2 Human genes 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 229940127121 immunoconjugate Drugs 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 230000005917 in vivo anti-tumor Effects 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000009533 lab test Methods 0.000 description 1
- 239000004310 lactic acid Substances 0.000 description 1
- 235000014655 lactic acid Nutrition 0.000 description 1
- 229960004891 lapatinib Drugs 0.000 description 1
- 238000009092 lines of therapy Methods 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 159000000003 magnesium salts Chemical class 0.000 description 1
- 210000004995 male reproductive system Anatomy 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- 239000011976 maleic acid Substances 0.000 description 1
- 239000001630 malic acid Substances 0.000 description 1
- 235000011090 malic acid Nutrition 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- WKPWGQKGSOKKOO-RSFHAFMBSA-N maytansine Chemical group CO[C@@H]([C@@]1(O)C[C@](OC(=O)N1)([C@H]([C@@H]1O[C@@]1(C)[C@@H](OC(=O)[C@H](C)N(C)C(C)=O)CC(=O)N1C)C)[H])\C=C\C=C(C)\CC2=CC(OC)=C(Cl)C1=C2 WKPWGQKGSOKKOO-RSFHAFMBSA-N 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 230000008172 membrane trafficking Effects 0.000 description 1
- 229960005558 mertansine Drugs 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 229940098779 methanesulfonic acid Drugs 0.000 description 1
- 210000004688 microtubule Anatomy 0.000 description 1
- 230000008880 microtubule cytoskeleton organization Effects 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 201000010225 mixed cell type cancer Diseases 0.000 description 1
- 208000029638 mixed neoplasm Diseases 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 210000005087 mononuclear cell Anatomy 0.000 description 1
- 210000000066 myeloid cell Anatomy 0.000 description 1
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 description 1
- ZHDORMMHAKXTPT-UHFFFAOYSA-N n-benzoylbenzamide Chemical compound C=1C=CC=CC=1C(=O)NC(=O)C1=CC=CC=C1 ZHDORMMHAKXTPT-UHFFFAOYSA-N 0.000 description 1
- YZMHQCWXYHARLS-UHFFFAOYSA-N naphthalene-1,2-disulfonic acid Chemical compound C1=CC=CC2=C(S(O)(=O)=O)C(S(=O)(=O)O)=CC=C21 YZMHQCWXYHARLS-UHFFFAOYSA-N 0.000 description 1
- 230000003472 neutralizing effect Effects 0.000 description 1
- 239000002547 new drug Substances 0.000 description 1
- 231100000956 nontoxicity Toxicity 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 244000309459 oncolytic virus Species 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 208000033808 peripheral neuropathy Diseases 0.000 description 1
- 210000001986 peyer's patch Anatomy 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 231100000374 pneumotoxicity Toxicity 0.000 description 1
- 102000054765 polymorphisms of proteins Human genes 0.000 description 1
- 238000010837 poor prognosis Methods 0.000 description 1
- 159000000001 potassium salts Chemical class 0.000 description 1
- 230000036515 potency Effects 0.000 description 1
- 231100001271 preclinical toxicology Toxicity 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 235000019260 propionic acid Nutrition 0.000 description 1
- 108090000623 proteins and genes Proteins 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- 230000007047 pulmonary toxicity Effects 0.000 description 1
- IUVKMZGDUIUOCP-BTNSXGMBSA-N quinbolone Chemical compound O([C@H]1CC[C@H]2[C@H]3[C@@H]([C@]4(C=CC(=O)C=C4CC3)C)CC[C@@]21C)C1=CCCC1 IUVKMZGDUIUOCP-BTNSXGMBSA-N 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 108010054624 red fluorescent protein Proteins 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000004202 respiratory function Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 229950000143 sacituzumab govitecan Drugs 0.000 description 1
- ULRUOUDIQPERIJ-PQURJYPBSA-N sacituzumab govitecan Chemical compound N([C@@H](CCCCN)C(=O)NC1=CC=C(C=C1)COC(=O)O[C@]1(CC)C(=O)OCC2=C1C=C1N(C2=O)CC2=C(C3=CC(O)=CC=C3N=C21)CC)C(=O)COCC(=O)NCCOCCOCCOCCOCCOCCOCCOCCOCCN(N=N1)C=C1CNC(=O)C(CC1)CCC1CN1C(=O)CC(SC[C@H](N)C(O)=O)C1=O ULRUOUDIQPERIJ-PQURJYPBSA-N 0.000 description 1
- 238000005070 sampling Methods 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 159000000000 sodium salts Chemical class 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 230000010473 stable expression Effects 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000011272 standard treatment Methods 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 150000003460 sulfonic acids Chemical class 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- DKPFODGZWDEEBT-QFIAKTPHSA-N taxane Chemical class C([C@]1(C)CCC[C@@H](C)[C@H]1C1)C[C@H]2[C@H](C)CC[C@@H]1C2(C)C DKPFODGZWDEEBT-QFIAKTPHSA-N 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- 230000008719 thickening Effects 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 206010043554 thrombocytopenia Diseases 0.000 description 1
- 230000036962 time dependent Effects 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 229960000303 topotecan Drugs 0.000 description 1
- UCFGDBYHRUNTLO-QHCPKHFHSA-N topotecan Chemical compound C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 UCFGDBYHRUNTLO-QHCPKHFHSA-N 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 231100000816 toxic dose Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 231100000155 toxicity by organ Toxicity 0.000 description 1
- 230000007675 toxicity by organ Effects 0.000 description 1
- 231100000440 toxicity profile Toxicity 0.000 description 1
- 231100000027 toxicology Toxicity 0.000 description 1
- 102000040811 transporter activity Human genes 0.000 description 1
- 108091092194 transporter activity Proteins 0.000 description 1
- 230000004565 tumor cell growth Effects 0.000 description 1
- 230000005760 tumorsuppression Effects 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6801—Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
- A61K47/6803—Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6835—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6851—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
- A61K47/6855—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6835—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6851—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
- A61K47/6857—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from lung cancer cell
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6835—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6851—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
- A61K47/6863—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from stomach or intestines cancer cell
-
- 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
- C07K16/32—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
-
- 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
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
-
- 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
- C07K2317/77—Internalization into the cell
Definitions
- HER2 a member of the epidermal growth factor receptor (EGFR/ErbB) family encoded by the ERBB2 gene, is frequently amplified and overexpressed in various types of cancer, including approximately 20-25% of breast cancers, 20-30% of bladder, 10-30% of gastric esophageal, and 20% of lung cancers(1-3). This gene amplification and overexpression is associated with a poor prognosis of the relevant malignancies. Enhanced or constitutive HER2 activity is believed to play a driving role in the malignancy of HER2-expressing human tumor cells.
- EGFR/ErbB epidermal growth factor receptor
- HER2- targeting ADCs utilize the high specificity of anti-HER2 antibodies to deliver potent cytotoxic agents to HER2-expressing cells and directly kill the target cancer cells(4-6).
- Kadcyla which consists of trastuzumab and a tubulin polymerization inhibitor, was the first HER2-targeting ADC approved for the treatment of breast cancer patients whose tumors had become resistant to other HER2-targeting therapies.
- ADCs containing new anti-HER2 antibodies or new toxin payloads such as Dxd or MMAE, Enhertu (Trastuzumab deruxtecan, DS-8201a) and AIDIXI® (RC48), have demonstrated promising responses in HER2-positive breast and gastric cancers, as well as in patient populations that do not typically respond well to other HER2-targeting therapies, such as low HER2-expressing breast cancers and urothelial carcinoma(8-11).
- These newer generations of ADCs significantly expand HER2-targeting indications, but still face challenges of resistance and/or severe side effects associated with their specific payload toxins.
- Enhertu has reported an overall response rate of 80% in HER2-positive breast cancer, though the response rate in other indications (gastric, lung, and other cancers) was significantly lower, indicating the presence of primary resistance in these patient populations. Enhertu also reported approximately 12-18% of interstitial lung disease (ILD) in the breast cancer clinical studies (DESTINY-Breast03 & 04) and 7% patients who presented with Grade 3- 4 ILD in DESTINY-Breast03 study and 0.5% patients who presented with Grade 5 ILD in DESTINY-Breast04 study (10-11). Therefore, ADCs containing toxins with different mechanisms of action and desired safety profiles may have an advantage in overcoming resistance to current treatments.
- ILD interstitial lung disease
- Eribulin is a synthetic analog of the marine macrolide halichondrin B that has been approved for the treatment of metastatic breast cancer and liposarcoma(12). Its unique mechanism of action involves binding to the interdimer interface or the ⁇ -tubulin subunit alone, inhibiting only the growth of microtubules with no effect on shortening. This distinguishes it from taxanes and vinca alkaloids, which suppress both the growth and shortening phases of microtubule dynamic instability. The unique tubulin-based mechanism of eribulin may contribute to its ability to overcome taxane resistance and display a wider range of anti-cancer activity(12,13).
- the ADC takes the form of Ab-(L-D)p, where Ab is an anti-HER2 antibody moiety (e.g., an antibody having the same structure as trastuzumab), L is a cleavable linker comprising maleimido-PEG2-valine-citrulline-p-aminobenzylcarbamyl (or Mal- (PEG)2-Val-Cit-pAB), D is eribulin, and p is an integer from 1 to 8 inclusive.
- Ab an anti-HER2 antibody moiety
- L is a cleavable linker comprising maleimido-PEG2-valine-citrulline-p-aminobenzylcarbamyl (or Mal- (PEG)2-Val-Cit-pAB)
- D is eribulin
- p is an integer from 1 to 8 inclusive.
- compositions comprising multiple copies of the described ADCs, wherein the average drug loading (average p) of the ADCs in the composition is between about 2 and 8, or between 3 and 5, or about 3.5 to about 4.5, or about 4.
- therapeutic uses of pharmaceutical compositions comprising the ADC described herein and a pharmaceutically acceptable carrier(s).
- the therapeutic uses include treating a proliferative disease, disorder, or condition in a subject or patient (a mammal, or a human being).
- the present disclosure provides methods of treating a cancer that expresses an antigen targeted by the antibody moiety of the ADC, such as HER2.
- methods are provided of killing or inhibiting the proliferation of tumor cells or cancer cells by administering a therapeutically effective amount and/or regimen of any one of the described ADCs or their compositions.
- the cancer is HER2-low expressing breast cancer, HER2-expressing or HER2 mutation non-small cell lung cancer, parotid cancer, urothelial carcinoma, gastric/esophagogastric junction cancer.
- the cancer can be HER2 breast cancer, bladder cancer, urothelial carcinoma salivary gland cancer, breast cancer, biliary tract (or bile duct) cancer, ovarian cancer, urologic cancer, endometrial cancer, cervical cancer, lung cancer, stomach cancer, pancreatic cancer, colorectal cancer, bladder cancer, prostate cancer, tonsil cancer, head and neck tumors, squamous cell carcinoma of the head and neck, liver cancer, mucoepidermoid carcinoma, rectal cancer, oral cavity tumors (or mouth cancer), cholangiocarcinoma, laryngeal tumors, colorectal cancer, urinary system tumors, lip tumors, larynx cancer, palate cancer, mucoepidermoid carcinoma, primary mucinous adenocarcinoma, recurrent cholangiocarcinoma, peritoneum malignant tumors, gastric cancer, tongue tumors, skin tumors, renal cell carcinoma, gastroesophageal junction adenocarcinoma,
- Figure 1 shows structure and characterization of BB-1701, an ADC of the present disclosure.
- Figure 1A shows basic structure of BB-1701 and predicted eribulin release upon cathepsins digestion.
- Figures 1B & 1C shows plasma stability study of BB-1701: Tab (Trastuzumab, ⁇ ), ADC ( ⁇ ) and free eribulin ( ⁇ ) measurement in human (1B) and monkey (1C) plasma samples where BB-1701 was incubated for up to 21 days.
- Figure 2 shows BB-1701 binding to HER2.
- A Binding to HER2 (BIAcore).
- FIG. 3 shows BB-1701 binding to endogenous HER2 expressed on A: NCI-N87; B: BT- 474 or C: JIMT-1 cells.
- Figure 4 shows internalization of BB-1701 in BT-474 cancer cells.
- Figures 5 shows in vitro cytotoxicity. Cytotoxicity curves of cell lines of different HER2 expression levels treated with BB-1701 (T-eribulin) and DS-8201a (T-Dxd). Relative HER2 expression of each cell line was evaluate by flow cytometry, left peak and right peak represented vehicle and anti-HER2 antibody-stained cell populations, respectively.
- Figure 6 shows in vivo tumor suppression in xenograft models that were insensitive to DS- 8201a treatment: Each point represents the mean and standard error.
- Figures 7A-7C show the bystander effect of BB-1701. in vivo bystander effect with co- inoculation of N87 (HER2+++) and U87-RFP(HER2-null) cells.
- A tumor volume of the mixed tumor;
- B live imaging of the tumor bearing mice from dosing day (day0) to day 9;
- C Average luminescence in (B).
- Figures 8D-8F shows U87-mCherry only tumors volume and luminescence.
- FIG. 8A-8F show immunogenic cell death induced by BB-1701.
- A Calreticulin expression on the cell surface of BT-474 cells treated with eribulin or BB-1701, analyzed with flow cytometry.
- B ATP level was quantified in the culture medium of 2nM eribulin or 0.167nM BB-1701 treated BT-474 cells at indicated time points.
- Figures 10A is the Waterfall of target lesion changes in patients of HER2 positive breast cancer treated with BB-1701.
- Figure 10B is the Swimmer plot treatment duration of BB-1701 in patients with HER2 positive breast cancer.
- Figure 11A Waterfall plot of target lesion changes in patients of HER2 low expressing breast cancer treated with BB-1701.
- Figure 11B Swimmer plot treatment duration of BB-1701 in patients with HER2 low expressing breast cancer.
- Figure 12A Waterfall plot of target lesion changes in NSCLC patients with Her2 amplification or mutations treated with BB-1701 (data cutoff date Sep, 30 th 2023).
- Figure 12B Swimmer plot treatment duration of BB-1701 in NSCLC patients with HER2 amplification or mutations (data cutoff date Sep.
- an antibody-drug conjugate takes the form of Ab-(L-D) p , where Ab is an anti-HER2 antibody moiety (e.g., an antibody having the same structure as trastuzumab), L is a cleavable linker comprising maleimido-PEG2-valine-citrulline-p-aminobenzylcarbamyl (or Mal-(PEG)2-Val-Cit- pAB), D is eribulin, and p is an integer from 1 to 8 inclusive.
- Ab is an anti-HER2 antibody moiety (e.g., an antibody having the same structure as trastuzumab)
- L is a cleavable linker comprising maleimido-PEG2-valine-citrulline-p-aminobenzylcarbamyl (or Mal-(PEG)2-Val-Cit- pAB)
- D is eribulin
- p is an integer from 1 to 8 inclusive.
- compositions comprising multiple copies of the described ADCs, wherein the average drug loading (average p) of the ADCs in the composition is between about 2 and 8, or 3 and 5, or about 3.5 to about 4.5, or about 4.
- pharmaceutical compositions comprising an ADC and a pharmaceutically acceptable carrier.
- Another aspect of the present disclosure includes therapeutic uses for the described ADC compounds and compositions, e.g., in treating a cancer that expresses an antigen targeted by the antibody moiety of the ADC, such as HER2.
- methods are provided of killing or inhibiting the proliferation of tumor cells or cancer cells by administering a therapeutically effective amount and/or regimen of any one of the described ADCs.
- the cancer is HER2 breast cancer, HER2-low expressing breast cancer, HER2- expressing or HER2 mutation non-small cell lung cancer, bladder cancer, urothelial carcinoma, or gastric/esophagogastric junction cancer.
- the cancer includes salivary gland cancer, breast cancer, biliary tract (or bile duct) cancer, ovarian cancer, urologic cancer, endometrial cancer, cervical cancer, lung cancer, stomach cancer, pancreatic cancer, colorectal cancer, bladder cancer, prostate cancer, tonsil cancer, head and neck tumors, squamous cell carcinoma of the head and neck, liver cancer, mucoepidermoid carcinoma, rectal cancer, oral cavity tumors (or mouth cancer), cholangiocarcinoma, laryngeal tumors, colorectal cancer, urinary system tumors, lip tumors, larynx cancer, palate cancer, mucoepidermoid carcinoma, primary mucinous adenocarcinoma, recurrent cholangiocarcinoma, peritoneum malignant tumors, gastric cancer, tongue tumors, skin tumors, renal cell carcinoma, gastroesophageal junction adenocarcinoma, non-small cell lung cancer, uterine cancer, endometrio
- the patient is one whose cancer has demonstrated resistance to other HER2-targeting therapeutics including other anti-HER2 antibody drug conjugate treatment.
- Trastuzumab is a recombinant IgG1 kappa, humanized monoclonal antibody that selectively binds with high affinity to the extracellular domain of the human epidermal growth factor receptor protein.
- Herceptin it was first FDA approved in 1998 to treat HER2+ metastatic breast cancer, and in 2006, it was approved by the FDA to treat Her2+ early breast cancer, and in 2010, it was approved by the FDA to treat HER2+ metastatic stomach cancer.
- the drug-linker can be attached to the antibody moiety through cysteine coupling, amine coupling, terminal coupling, enzymatic coupling, or carbohydrate coupling.
- pharmaceutically acceptable salts of the ADCs or compositions described herein which include acid addition salts of inorganic acids, carboxylic acids and sulfonic acids, for example, salts of the following acids: hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, naphthalene disulfonic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
- the pharmaceutically acceptable salts of the antibody-drug conjugates of the present disclosure also include salts of conventional bases, for example alkali metal salts (e.g., sodium salts and potassium salts), alkaline earth metal salts (e.g., calcium salts and magnesium salts) and ammonium salts derived from ammonia or organic amines containing from 1 to 16 carbon atoms, in which the organic amines are, for example, ethylamine, diethylamine, triethylamine, ethyl diisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzamide, N-methylpiperidine, N-methylmorpholine, arginine, lysine and 1,2-ethylenediamine.
- alkali metal salts e.g., sodium salts and potassium salts
- alkaline earth metal salts e.g., calcium salts and magnesium salts
- subject refers to mammal, such as cattle, cats, dogs, and horses, primates, mice and rats. In certain embodiments, the mammal refers to a human.
- the term “about” as used herein with reference to a certain given value or quantity herein means a range of up to 25% deviation from (greater or smaller than) the given value.
- isolated antibody refers to an antibody that is substantially free of other antibodies having different antigenic specificities. An isolated antibody that specifically binds to an antigen is substantially free of antibodies that do not bind to that antigen.
- the term “monoclonal antibody” as used herein refer to a preparation of a population of antibody molecules of substantially homogeneous molecular composition, wherein the individual antibodies in the population of the antibody molecules are identical except for possible naturally occurring mutations that may be present in miniscule amounts.
- the present disclosure provides a pharmaceutical composition comprising the ADCs herein or the pharmaceutically acceptable salts thereof, together with a pharmaceutically acceptable carrier for the treatment of various HER2-expressing cancers described herein.
- pharmaceutically acceptable carrier includes pharmaceutically acceptable diluents, or stabilizers, or other excipients.
- compositions may comprise one or more additional pharmaceutically active ingredients, such as another antibody, a drug, e.g., a cytotoxic or anti-tumor agent.
- composition can be suitable for intravenous, intramuscular, subcutaneous, parenteral, epidermal, and other routes of administration.
- the active ingredient can be coated with a material or otherwise loaded in a material or structure to protect it from the action of acids and other natural conditions that may inactivate it.
- parenteral administration means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
- the composition of the invention can be administered via a non-parenteral route, such as a topical, epidermal or mucosal route of administration, e.g., intranasally, orally, vaginally, rectally, sublingually or topically.
- the present invention provides a method of treating cancer in a human subject, comprising administering an effective amount of the ADC (such as BB-1701) or the pharmaceutical composition herein containing the ADC.
- the cancer can be a cancer associated with expression of HER2.
- the patient is one whose cancer has demonstrated resistance to other HER2-targeting therapeutics including other anti-HER2 antibody drug conjugate treatment.
- the route of administration can be intravenous, intramuscular, subcutaneous, parenteral, epidermal, and other routes of administration.
- dosage regimens can be adjusted to provide the optimum desired response (e.g., a therapeutic response).
- Single bolus or divided doses can be administered based on the subject, the disease to be treated, etc.
- Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated. Each unit contains a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Sustained release formulation can be used in which case less frequent administration is required.
- the dosage may range from about 0.0001 to 100 mg/kg, and more usually 0.01 to 10 mg/kg, of the body weight of the subject.
- dosages can be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg.
- a suitable treatment regime can be once per week, once every two weeks, once every three weeks, once every four weeks, once a month, etc.
- Example dosage regimens for an anti-HER2 antibody of the invention can include 1 mg/kg body weight or 10 mg/kg body weight via intravenous administration.
- a “therapeutically effective amount” or “effective amount” of the ADCs or compositions herein preferably results in a decrease in severity of disease symptoms, an increase in frequency and/or duration of disease symptom-free periods, prevention or reduction of likelihood of impairment or disability due to the disease affliction, or inhibition or delaying of the progression of disease.
- a “therapeutically effective amount” of the ADC or compositions herein may inhibit tumor growth by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects.
- BB-1701 is composed of an anti-HER2 antibody linked to a cytotoxic payload of eribulin, a synthetic analog of the marine macrolide halichondrin B.
- the sequence of the anti-HER2 antibody used in BB-1701 is identical to that of the well-known trastuzumab which was produced by mammalian cell culture in CHO cells and purified with a process that includes three chromatography steps and a dedicated viral clearance step.
- the conjugation of the eribulin payload to the antibody is achieved through a maleimido-PEG2-valine-citrulline-p- aminobenzylcarbamyl linker.
- BB-1701 molecule can include 2, 4, 6, 8 linked drug portion, and in this study the drug distribution as determined by hydrophobic interaction chromatography (HIC) had an average Drug to Antibody Ratio (DAR) of about 4.
- the linker peptide (valine-citrulline, VC) can be decomposed by lysosomal cathepsins which are highly expressed in tumor cells.
- BB-1701 After binding to HER2 molecules on the cell membrane and internalized into tumor cells, BB-1701 is cleaved by lysosomal enzymes (14), the p- aminobenzycarbamyl (pAB) spacer undergoes self-immolation upon cleavage of VC linker, and free eribulin, not the eribulin-linker construct, is the primary form of free toxin after being released from the ADC (Fig. 1A).
- Study of in vitro plasma stability where BB-1701 was incubated in human or cynomolgus monkey plasma for up to 21 days revealed that both ADC and TAb appeared to be stable during the 21-day incubation (Fig. 1B and 1C,).
- Cytotoxicity activity of BB-1701 in comparison with DS-8201a The cytotoxic activity of BB-1701 was evaluated and compared with DS-8201a which had the same anti-HER2 antibody linked to a different toxin payload Dxd (exatecan derivative for ADC) in four cell lines: human breast cancer cell line BT-474, gastric cancer cell line NCI-N87, lung cancer cell line NCI-H1975 and A549, where HER2 expression of these cell lines were analyzed by flow cytometry and histogram graphs were shown in the corresponding kill curves graphs in Fig. 5.
- BT474 and NCI-N87 have previously been found to be sensitive to DS-8201a.
- Results showed that BB-1701 had higher potencies against NCI-N87 and BT-474 cells than DS- 8201a, with IC50 values that were 5-10-fold lower than the IC50 values of DS-8201a-treated cells (Fig. 5 upper panels).
- DS-8201a was unable to completely suppress cell growth, as indicated by the higher low-plateau in the kill curves compared with BB- 1701.
- BB-1701 retained some cytotoxicity against NCI-1975 and A549 cells, these two cell lines were largely resistant to DS-8201a treatment.
- BB- 1701 may be a more effective treatment option for certain cancer types compared with DS-8201a (Fig. 5 lower panels).
- Potent anti-tumor activity in tumors insensitive to DS-8201a The in vivo antitumor activity of BB-1701 was also evaluated in several animal models that had developed resistance to DS-8201a treatment.
- a model derived from a non-small cell lung cancer line NCI-H1975 cells when given a single dose of 5 mg/kg, DS-8201a was unable to suppress tumor growth (Fig.6).
- BB-1701 greatly suppressed tumor growth, with a TGI of over 90%, and caused 40% tumor regression (2/5 CR) (Fig. 6).
- BB-1701 induced ICD and the activation of the immune system
- Certain chemotherapy drugs, oncolytic viruses, and radiotherapy can stimulate the production of neoantigens and promote immunogenic cell death (ICD) in cancer cells, which is characterized by the release of molecules called damage-associated molecular patterns (DAMPs) that can stimulate the immune system (15).
- DAMPs damage-associated molecular patterns
- BT-474 cells were exposed to increasing concentrations of either BB-1701 or eribulin.
- Expression of calreticulin, a type of DAMP was found increased in a dose-dependent manner in BT-474 cells treated with either BB-1701 or eribulin (Fig.8A).
- EC50 values the concentration at which half-maximum calreticulin was induced
- cytotoxic IC 50 values the concentration at which 50% of the cells were killed
- BB-1701 was found to increase the number of immune cells called macrophages in the tumor compared to treatment with a control substance (Fig. 8D).
- Pharmacokinetics and toxicity studies of BB-1701 in cynomolgus monkey To evaluate the pharmacokinetics and potential toxic effects of BB-1701 when given repeatedly at different doses, five male and five female cynomolgus monkeys (Aujun Biological Technology Co., Ltd., Guangzhou, China) were divided into groups and received four repeated doses (intravenous, every 3 weeks ⁇ 4) of BB-1701 at doses of 0, 2, 4, and 8 mg/kg.
- TK Toxicokinetic studies were incorporated into the repeated dose toxicity study. Intensive blood sampling was performed in the first and third dose cycles, while sparse sampling was performed in the second and fourth dose cycles. In addition, cardiovascular and respiratory function were evaluated as part of the safety pharmacology assessment during the repeated dose toxicity study. The results showed that BB-1701 was stable in circulation and that its concentration in the blood was roughly proportional to the dose given (Fig.9, Table 1).
- HNSTD non- severely toxic dose
- T-DM1 has had low incidences of ILD in human patients and a preclinical toxicity study showed that T- DM1 treatment caused infiltration of mononuclear cells into the interstitium of the lung in cynomolgus monkeys at 10 mg/kg (given every 3 weeks for 4 doses).
- T-DM1 treatment caused infiltration of mononuclear cells into the interstitium of the lung in cynomolgus monkeys at 10 mg/kg (given every 3 weeks for 4 doses).
- BB-1701 in monkeys, there were no toxicities suggestive of ILD even at the highest dose groups.
- T-DM1 may be toxin-specific and may be mediated by HER2 expression in the lung (18).
- the main adverse events of T-DM1 are thrombocytopenia and peripheral neuropathy, which are typical toxicities of tubulin inhibitor- conjugated ADCs (22).
- the EMILIA study of T-DM1 in late stage HER2 positive breast cancer patients found an overall response rate of 43.6% and a progression-free survival of 9.6 months, indicating more than 50% primary resistance and an additional 50% acquired resistance within 10 months (20).
- the DESTINY-Breast003 studies of DS-8201a in the third line treatment of late stage HER2 positive breast cancer patients found impressive overall response rates of 69%, respectively, and progression-free survivals of 18.5 months, suggesting that primary resistance to DS-8201 may be relatively infrequent, but around 30% of DS-8201a-treated patients acquired resistance within one year and more than 50% acquired resistance within two years (10).
- DS-8201 has had lower response rates and shorter progression-free survival in other indications, presenting more severe resistance challenges (8).
- HER2-targeting ADCs There are dozens of potential mechanisms of resistance to HER2-targeting ADCs, and as long as HER2 expression persists, ADCs with different mechanisms of action and/or different drug metabolism mechanisms may offer the potential to overcome resistance challenges (21).
- BB-1701 treatment showed significant anti-tumor activity in DS-8201a-resistant model.
- eribulin uses a completely different set of ATP-binding Cassette (ABC) transporters from those used by topotecan and diflomotecan (of the same drug family as DXd) for cytoplasmic membrane trafficking (22).
- ABSC ATP-binding Cassette
- BB-1701 may use nonoverlapping mechanisms to overcome drug resistance and may be effective against tumors with acquired resistance to other HER2-targeting ADCs.
- FIGS. 10A and 10B are waterfall and swimmer plots of HER2 positive breast cancer patients.
- the study showed a best overall response rate of 66.7% (12/18 patients) and disease control rate of 100% (18/18 patients).
- the longest treatment duration was 60 weeks and 40.7% patients were on treatment for over 6 months. All patients received BB-1701 after they had failed treatment of standard therapies or experimental treatments, with prior therapy lines of treatment ranging from 1 to 6 lines. All patients had progressive diseases from their previous treatments when they were enrolled in the study with BB-1701.
- Figures 11A and 11B are waterfall and swimmer plots of HER2-low breast cancer patients.
- Figures 12A and 12B are waterfall and swimmer plots of HER2 expressing lung cancer patients.
- Q3W gastric/esophagogastric junction cancer
- the target lesions of the only one urothelial carcinoma patient had 65.7% reduction; the target lesions of the only one parotid cancer patient had 48.6% reduction; the target lesions of 5/6 gastric/esophagogastric junction cancer patients remained largely unchanged ( ⁇ 30% changes in size), only 1/6 patients with the target lesions grew >30% changes in size.
- the longest treatment duration of gastric/esophagogastric junction cancer patients was 63 weeks. All patients had progressive diseases from their previous treatments when they were enrolled in the study with BB-1701.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Cell Biology (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Immunology (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Oncology (AREA)
- Organic Chemistry (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Pulmonology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
Methods of using an antibody-drug conjugate (ADC) Ab-(L-D)p or its compositions to treat various cancers are provided, where Ab is an anti-HER2 antibody moiety (e.g., an antibody having the same structure as trastuzumab), L is a cleavable linker comprising Mal-(PEG)2-Val-Cit-pAB, D is eribulin, and p is an integer from 1 to 8 inclusive.
Description
ANTIBODY-DRUG CONJUGATE FOR CANCER TREATMENT Background HER2, a member of the epidermal growth factor receptor (EGFR/ErbB) family encoded by the ERBB2 gene, is frequently amplified and overexpressed in various types of cancer, including approximately 20-25% of breast cancers, 20-30% of bladder, 10-30% of gastric esophageal, and 20% of lung cancers(1-3). This gene amplification and overexpression is associated with a poor prognosis of the relevant malignancies. Enhanced or constitutive HER2 activity is believed to play a driving role in the malignancy of HER2-expressing human tumor cells. As a clinically validated therapeutic target, a number of therapeutic agents, including antibodies, small molecules, and antibody-drug conjugates (ADCs), have been approved for the treatment of patients with HER2-expressing tumors(4-6). Unlike anti-HER2 antibodies and small molecule tyrosine kinase inhibitors (TKIs) which inhibit tumor growth through interference with HER2 downstream signaling pathways, HER2- targeting ADCs utilize the high specificity of anti-HER2 antibodies to deliver potent cytotoxic agents to HER2-expressing cells and directly kill the target cancer cells(4-6). Despite the possibility of cancer cells developing resistance to anti-HER2 antibodies or TKI treatment due to mutations in the HER2 gene, downstream signaling molecules, or other genes that may compensate for HER2 signaling pathways, the unique mechanism of action of ADCs may overcome this resistance(7). Kadcyla (T-DM1), which consists of trastuzumab and a tubulin polymerization inhibitor, was the first HER2-targeting ADC approved for the treatment of breast cancer patients whose tumors had become resistant to other HER2-targeting therapies. Recently, ADCs containing new anti-HER2 antibodies or new toxin payloads such as Dxd or MMAE, Enhertu (Trastuzumab deruxtecan, DS-8201a) and AIDIXI® (RC48), have demonstrated promising responses in HER2-positive breast and gastric cancers, as well as in patient populations that do not typically respond well to other HER2-targeting therapies, such as low HER2-expressing breast cancers and urothelial carcinoma(8-11). These newer generations of ADCs significantly expand HER2-targeting indications, but still face challenges of resistance and/or severe side effects associated with their specific payload toxins. Enhertu has reported an overall response rate of 80% in HER2-positive breast cancer, though the response rate in other indications (gastric, lung, and other cancers) was significantly lower, indicating the presence of
primary resistance in these patient populations. Enhertu also reported approximately 12-18% of interstitial lung disease (ILD) in the breast cancer clinical studies (DESTINY-Breast03 & 04) and 7% patients who presented with Grade 3- 4 ILD in DESTINY-Breast03 study and 0.5% patients who presented with Grade 5 ILD in DESTINY-Breast04 study (10-11). Therefore, ADCs containing toxins with different mechanisms of action and desired safety profiles may have an advantage in overcoming resistance to current treatments. Eribulin is a synthetic analog of the marine macrolide halichondrin B that has been approved for the treatment of metastatic breast cancer and liposarcoma(12). Its unique mechanism of action involves binding to the interdimer interface or the β-tubulin subunit alone, inhibiting only the growth of microtubules with no effect on shortening. This distinguishes it from taxanes and vinca alkaloids, which suppress both the growth and shortening phases of microtubule dynamic instability. The unique tubulin-based mechanism of eribulin may contribute to its ability to overcome taxane resistance and display a wider range of anti-cancer activity(12,13). Summary of the Invention In one aspect of the present disclosure, therapeutic uses of an antibody-drug conjugate (ADC) is provided. In some embodiments, the ADC takes the form of Ab-(L-D)p, where Ab is an anti-HER2 antibody moiety (e.g., an antibody having the same structure as trastuzumab), L is a cleavable linker comprising maleimido-PEG2-valine-citrulline-p-aminobenzylcarbamyl (or Mal- (PEG)2-Val-Cit-pAB), D is eribulin, and p is an integer from 1 to 8 inclusive. Also provided herein are therapeutic uses of compositions comprising multiple copies of the described ADCs, wherein the average drug loading (average p) of the ADCs in the composition is between about 2 and 8, or between 3 and 5, or about 3.5 to about 4.5, or about 4. Further provided herein are therapeutic uses of pharmaceutical compositions comprising the ADC described herein and a pharmaceutically acceptable carrier(s). The therapeutic uses include treating a proliferative disease, disorder, or condition in a subject or patient (a mammal, or a human being). For example, the present disclosure provides methods of treating a cancer that expresses an antigen targeted by the antibody moiety of the ADC, such as HER2. In various embodiments, methods are provided of killing or inhibiting the proliferation of tumor cells or cancer cells by administering a therapeutically effective amount and/or regimen of any one of the described ADCs or their compositions. In some embodiments,
the cancer is HER2-low expressing breast cancer, HER2-expressing or HER2 mutation non-small cell lung cancer, parotid cancer, urothelial carcinoma, gastric/esophagogastric junction cancer. In some embodiments, the cancer can be HER2 breast cancer, bladder cancer, urothelial carcinoma salivary gland cancer, breast cancer, biliary tract (or bile duct) cancer, ovarian cancer, urologic cancer, endometrial cancer, cervical cancer, lung cancer, stomach cancer, pancreatic cancer, colorectal cancer, bladder cancer, prostate cancer, tonsil cancer, head and neck tumors, squamous cell carcinoma of the head and neck, liver cancer, mucoepidermoid carcinoma, rectal cancer, oral cavity tumors (or mouth cancer), cholangiocarcinoma, laryngeal tumors, colorectal cancer, urinary system tumors, lip tumors, larynx cancer, palate cancer, mucoepidermoid carcinoma, primary mucinous adenocarcinoma, recurrent cholangiocarcinoma, peritoneum malignant tumors, gastric cancer, tongue tumors, skin tumors, renal cell carcinoma, gastroesophageal junction adenocarcinoma, non-small cell lung cancer, uterine cancer, endometrioid cancer, HER2-mutant non-small cell lung cancer, HER2-positive gastric cancer, HER2-low expression breast cancer, or HER2-positive breast cancer. In some embodiments, the patient is one whose cancer has demonstrated resistance to other HER2-targeting therapeutics including other anti-HER2 antibody drug conjugate treatment. Brief Description of the Drawings Figure 1 shows structure and characterization of BB-1701, an ADC of the present disclosure. Figure 1A shows basic structure of BB-1701 and predicted eribulin release upon cathepsins digestion. Figures 1B & 1C shows plasma stability study of BB-1701: Tab (Trastuzumab, ♦), ADC (■) and free eribulin (▲) measurement in human (1B) and monkey (1C) plasma samples where BB-1701 was incubated for up to 21 days. Figure 2 shows BB-1701 binding to HER2. A: Binding to HER2 (BIAcore). B: Binding to HER2 (ELISA). Figure 3 shows BB-1701 binding to endogenous HER2 expressed on A: NCI-N87; B: BT- 474 or C: JIMT-1 cells. Figure 4 shows internalization of BB-1701 in BT-474 cancer cells. Figures 5 shows in vitro cytotoxicity. Cytotoxicity curves of cell lines of different HER2 expression levels treated with BB-1701 (T-eribulin) and DS-8201a (T-Dxd). Relative HER2
expression of each cell line was evaluate by flow cytometry, left peak and right peak represented vehicle and anti-HER2 antibody-stained cell populations, respectively. Figure 6 shows in vivo tumor suppression in xenograft models that were insensitive to DS- 8201a treatment: Each point represents the mean and standard error. Figures 7A-7C show the bystander effect of BB-1701. in vivo bystander effect with co- inoculation of N87 (HER2+++) and U87-RFP(HER2-null) cells. (A) tumor volume of the mixed tumor; (B) live imaging of the tumor bearing mice from dosing day (day0) to day 9; (C) Average luminescence in (B). Figures 8D-8F shows U87-mCherry only tumors volume and luminescence. D: live imaging of tumor bearing mice in PBS and BB-1701 treatment groups on day 30; E: statistical analysis of mCherry luminescence in D; F: tumor volume of PBS or BB-1701 treated U87-mCherry model. Each point represents the mean and standard error. N.S., not significant. Figures 8A-8F show immunogenic cell death induced by BB-1701. (A) Calreticulin expression on the cell surface of BT-474 cells treated with eribulin or BB-1701, analyzed with flow cytometry. (B) ATP level was quantified in the culture medium of 2nM eribulin or 0.167nM BB-1701 treated BT-474 cells at indicated time points. (C) IHC analysis of HMGB1 expression in PBS (left box) and BB-1701 (5 mg/kg, right box) treated BT-474 tumors in vivo, black arrow heads indicate the tumor cells experiencing ICD where HMGB1 translocate to cytosol from nuclear area. (D) IHC analysis of macrophage infiltration (F4/80 staining) in PBS (left box) and BB-1701 (5 mg/kg, right box) treated BT-474 tumors in vivo. Figure 9 shows mean serum concentration-time curve of BB-1701 ADC and Tab following single i.v. administration at 2, 4, 8 mg/kg in cynomolgus monkeys. Figures 10A is the Waterfall of target lesion changes in patients of HER2 positive breast cancer treated with BB-1701. Figure 10B is the Swimmer plot treatment duration of BB-1701 in patients with HER2 positive breast cancer. Figure 11A: Waterfall plot of target lesion changes in patients of HER2 low expressing breast cancer treated with BB-1701. Figure 11B: Swimmer plot treatment duration of BB-1701 in patients with HER2 low expressing breast cancer. Figure 12A: Waterfall plot of target lesion changes in NSCLC patients with Her2 amplification or mutations treated with BB-1701 (data cutoff date Sep, 30th 2023). Figure 12B: Swimmer plot treatment duration of BB-1701 in NSCLC patients with HER2 amplification or mutations (data cutoff date Sep. 30th, 2023)
Detailed Description In one aspect of the present disclosure, an antibody-drug conjugate (ADC) is provided. In some embodiments, the ADC takes the form of Ab-(L-D)p, where Ab is an anti-HER2 antibody moiety (e.g., an antibody having the same structure as trastuzumab), L is a cleavable linker comprising maleimido-PEG2-valine-citrulline-p-aminobenzylcarbamyl (or Mal-(PEG)2-Val-Cit- pAB), D is eribulin, and p is an integer from 1 to 8 inclusive. Also provided herein are compositions comprising multiple copies of the described ADCs, wherein the average drug loading (average p) of the ADCs in the composition is between about 2 and 8, or 3 and 5, or about 3.5 to about 4.5, or about 4. Further provided herein are pharmaceutical compositions comprising an ADC and a pharmaceutically acceptable carrier. Another aspect of the present disclosure includes therapeutic uses for the described ADC compounds and compositions, e.g., in treating a cancer that expresses an antigen targeted by the antibody moiety of the ADC, such as HER2. In various embodiments, methods are provided of killing or inhibiting the proliferation of tumor cells or cancer cells by administering a therapeutically effective amount and/or regimen of any one of the described ADCs. In some embodiments, the cancer is HER2 breast cancer, HER2-low expressing breast cancer, HER2- expressing or HER2 mutation non-small cell lung cancer, bladder cancer, urothelial carcinoma, or gastric/esophagogastric junction cancer. In some embodiments, the cancer includes salivary gland cancer, breast cancer, biliary tract (or bile duct) cancer, ovarian cancer, urologic cancer, endometrial cancer, cervical cancer, lung cancer, stomach cancer, pancreatic cancer, colorectal cancer, bladder cancer, prostate cancer, tonsil cancer, head and neck tumors, squamous cell carcinoma of the head and neck, liver cancer, mucoepidermoid carcinoma, rectal cancer, oral cavity tumors (or mouth cancer), cholangiocarcinoma, laryngeal tumors, colorectal cancer, urinary system tumors, lip tumors, larynx cancer, palate cancer, mucoepidermoid carcinoma, primary mucinous adenocarcinoma, recurrent cholangiocarcinoma, peritoneum malignant tumors, gastric cancer, tongue tumors, skin tumors, renal cell carcinoma, gastroesophageal junction adenocarcinoma, non-small cell lung cancer, uterine cancer, endometrioid cancer, HER2-mutant non-small cell lung cancer, HER2-positive gastric cancer, HER2-low expression breast cancer, or HER2-positive breast cancer.
In some embodiments, the patient is one whose cancer has demonstrated resistance to other HER2-targeting therapeutics including other anti-HER2 antibody drug conjugate treatment. Trastuzumab is a recombinant IgG1 kappa, humanized monoclonal antibody that selectively binds with high affinity to the extracellular domain of the human epidermal growth factor receptor protein. Under the brand name Herceptin, it was first FDA approved in 1998 to treat HER2+ metastatic breast cancer, and in 2006, it was approved by the FDA to treat Her2+ early breast cancer, and in 2010, it was approved by the FDA to treat HER2+ metastatic stomach cancer. In some embodiments of the ADC, the drug-linker can be attached to the antibody moiety through cysteine coupling, amine coupling, terminal coupling, enzymatic coupling, or carbohydrate coupling. In the present disclosure also provide pharmaceutically acceptable salts of the ADCs or compositions described herein, which include acid addition salts of inorganic acids, carboxylic acids and sulfonic acids, for example, salts of the following acids: hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, naphthalene disulfonic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid. The pharmaceutically acceptable salts of the antibody-drug conjugates of the present disclosure also include salts of conventional bases, for example alkali metal salts (e.g., sodium salts and potassium salts), alkaline earth metal salts (e.g., calcium salts and magnesium salts) and ammonium salts derived from ammonia or organic amines containing from 1 to 16 carbon atoms, in which the organic amines are, for example, ethylamine, diethylamine, triethylamine, ethyl diisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzamide, N-methylpiperidine, N-methylmorpholine, arginine, lysine and 1,2-ethylenediamine. In the present disclosure, “subject” refers to mammal, such as cattle, cats, dogs, and horses, primates, mice and rats. In certain embodiments, the mammal refers to a human. The term “about” as used herein with reference to a certain given value or quantity herein means a range of up to 25% deviation from (greater or smaller than) the given value.
The term “isolated antibody” as used herein refers to an antibody that is substantially free of other antibodies having different antigenic specificities. An isolated antibody that specifically binds to an antigen is substantially free of antibodies that do not bind to that antigen. The term “monoclonal antibody” as used herein refer to a preparation of a population of antibody molecules of substantially homogeneous molecular composition, wherein the individual antibodies in the population of the antibody molecules are identical except for possible naturally occurring mutations that may be present in miniscule amounts. In further aspect, the present disclosure provides a pharmaceutical composition comprising the ADCs herein or the pharmaceutically acceptable salts thereof, together with a pharmaceutically acceptable carrier for the treatment of various HER2-expressing cancers described herein. As used herein, “pharmaceutically acceptable carrier” includes pharmaceutically acceptable diluents, or stabilizers, or other excipients. These include but are not limited solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, surface active agents, thickening or emulsifying agents, solid binders, dispersion or suspension aids, solubilizers, colorants, flavoring agents, coatings, disintegrating agents, lubricants, sweeteners, preservatives, isotonic agents, and the like that are physiologically compatible. The selection of a suitable carrier is within the knowledge of an artisan skilled in the art. The composition may comprise one or more additional pharmaceutically active ingredients, such as another antibody, a drug, e.g., a cytotoxic or anti-tumor agent. Pharmaceutical composition can be suitable for intravenous, intramuscular, subcutaneous, parenteral, epidermal, and other routes of administration. Depending on the route of administration, the active ingredient can be coated with a material or otherwise loaded in a material or structure to protect it from the action of acids and other natural conditions that may inactivate it. The phrase “parenteral administration” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. Alternatively, the composition of the invention can be administered via a non-parenteral route, such as a topical,
epidermal or mucosal route of administration, e.g., intranasally, orally, vaginally, rectally, sublingually or topically. In a further aspect, the present invention provides a method of treating cancer in a human subject, comprising administering an effective amount of the ADC (such as BB-1701) or the pharmaceutical composition herein containing the ADC. The cancer can be a cancer associated with expression of HER2. Her2 positive breast cancer, HER2-low expressing breast cancer, HER2- expressing or HER mutation non-small cell lung cancer, bladder cancer, urothelial carcinoma, gastric/esophagogastric junction cancer. biliary tract cancer, ovarian cancer, endometrial cancer, cervical cancer. In some embodiments, the patient is one whose cancer has demonstrated resistance to other HER2-targeting therapeutics including other anti-HER2 antibody drug conjugate treatment. The route of administration can be intravenous, intramuscular, subcutaneous, parenteral, epidermal, and other routes of administration. In the administration of the composition to the subject, dosage regimens can be adjusted to provide the optimum desired response (e.g., a therapeutic response). Single bolus or divided doses can be administered based on the subject, the disease to be treated, etc. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated. Each unit contains a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Sustained release formulation can be used in which case less frequent administration is required. For administration of an antibody or ADC pharmaceutical salts thereof of the present disclosure, the dosage may range from about 0.0001 to 100 mg/kg, and more usually 0.01 to 10 mg/kg, of the body weight of the subject. For example, dosages can be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg. A suitable treatment regime can be once per week, once every two weeks, once every three weeks, once every four weeks, once a month, etc. Example dosage regimens for an anti-HER2 antibody of the invention can include 1 mg/kg body weight or 10 mg/kg body weight via intravenous administration. A “therapeutically effective amount” or “effective amount” of the ADCs or compositions herein preferably results in a decrease in severity of disease symptoms, an increase in frequency and/or duration of disease symptom-free periods, prevention or reduction of likelihood of impairment or disability due to the disease affliction, or inhibition or delaying of the progression
of disease. For example, for the treatment of tumor-bearing subjects, a “therapeutically effective amount” of the ADC or compositions herein may inhibit tumor growth by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects. Examples Part A: Pre-clinical Evaluation Generation and characterization of an example of the anti-HER2 eribulin containing ADC, BB-1701 As shown in Fig.1A, BB-1701 is composed of an anti-HER2 antibody linked to a cytotoxic payload of eribulin, a synthetic analog of the marine macrolide halichondrin B. The sequence of the anti-HER2 antibody used in BB-1701 is identical to that of the well-known trastuzumab which was produced by mammalian cell culture in CHO cells and purified with a process that includes three chromatography steps and a dedicated viral clearance step. The conjugation of the eribulin payload to the antibody is achieved through a maleimido-PEG2-valine-citrulline-p- aminobenzylcarbamyl linker. Each of BB-1701 molecule can include 2, 4, 6, 8 linked drug portion, and in this study the drug distribution as determined by hydrophobic interaction chromatography (HIC) had an average Drug to Antibody Ratio (DAR) of about 4. The linker peptide (valine-citrulline, VC) can be decomposed by lysosomal cathepsins which are highly expressed in tumor cells. After binding to HER2 molecules on the cell membrane and internalized into tumor cells, BB-1701 is cleaved by lysosomal enzymes (14), the p- aminobenzycarbamyl (pAB) spacer undergoes self-immolation upon cleavage of VC linker, and free eribulin, not the eribulin-linker construct, is the primary form of free toxin after being released from the ADC (Fig. 1A). Study of in vitro plasma stability where BB-1701 was incubated in human or cynomolgus monkey plasma for up to 21 days revealed that both ADC and TAb appeared to be stable during the 21-day incubation (Fig. 1B and 1C,). There was a trend of time-dependent increase of free eribulin released in plasma, however, there was no more than 0.2% of conjugated eribulin in BB-1701 that was released even at the end of incubation time (day 21), (Fig. 1B, and 1C), suggesting that BB-1701 would be stable in circulation.
To determine the potential impact of the eribulin payload on the binding and internalization properties of the anti-HER2 antibody, in vitro studies were conducted comparing BB-1701 with the naked antibody. The results of these studies, which included binding to recombinant human HER2, binding to HER2 expressed on the cell membrane, and internalization into cancer cells, indicated that there were no significant differences between BB-1701 and the naked antibody in any of these measured properties (Figs. 2-4). This suggests that the conjugation of the eribulin payload did not negatively affect the ability of the anti-HER2 antibody to engage and internalize within target cancer cells. Cytotoxicity activity of BB-1701 in comparison with DS-8201a The cytotoxic activity of BB-1701 was evaluated and compared with DS-8201a which had the same anti-HER2 antibody linked to a different toxin payload Dxd (exatecan derivative for ADC) in four cell lines: human breast cancer cell line BT-474, gastric cancer cell line NCI-N87, lung cancer cell line NCI-H1975 and A549, where HER2 expression of these cell lines were analyzed by flow cytometry and histogram graphs were shown in the corresponding kill curves graphs in Fig. 5. BT474 and NCI-N87 have previously been found to be sensitive to DS-8201a. Results showed that BB-1701 had higher potencies against NCI-N87 and BT-474 cells than DS- 8201a, with IC50 values that were 5-10-fold lower than the IC50 values of DS-8201a-treated cells (Fig. 5 upper panels). Under the same assay conditions, DS-8201a was unable to completely suppress cell growth, as indicated by the higher low-plateau in the kill curves compared with BB- 1701. In contrast, while BB-1701 retained some cytotoxicity against NCI-1975 and A549 cells, these two cell lines were largely resistant to DS-8201a treatment. These findings suggest that BB- 1701 may be a more effective treatment option for certain cancer types compared with DS-8201a (Fig. 5 lower panels). Potent anti-tumor activity in tumors insensitive to DS-8201a The in vivo antitumor activity of BB-1701 was also evaluated in several animal models that had developed resistance to DS-8201a treatment. In a model derived from a non-small cell lung cancer line NCI-H1975 cells, when given a single dose of 5 mg/kg, DS-8201a was unable to suppress tumor growth (Fig.6). In contrast, BB-1701 greatly suppressed tumor growth, with a TGI of over 90%, and caused 40% tumor regression (2/5 CR) (Fig. 6). No gross toxicity (bodyweight
loss, food consumption or any other behavior changes) was noted with the treatment of BB-1701 in the tested model (data not shown). Overall, these results suggest that BB-1701 may be a potentially effective treatment option for tumors that are resistant to the best known HER2- targeting ADC on the market. Bystander effect of BB-1701 The bystander effect of BB-1701 was investigated. In an in vivo study, Her2-null U87-RFP cells (which have stable expression of the red fluorescent protein mCherry) were co-inoculated with NCI-N87 (HER2+++) cells into mice. When given a single low-dose (3 mg/kg) treatment, BB-1701 caused significant tumor growth suppression compared with PBS treatment (Fig. 7A). Additionally, while the mean fluorescence intensity in the PBS-treated group increased steadily over the course of the study, the mean fluorescence intensity in the BB-1701-treated group remained largely unchanged (Fig.7B-C). This suggests that the bystander effect of BB-1701 may have contributed to the suppression of HER2- null U87-RFP tumor cell growth in mix-cell tumors. In a separate experiment with mice bearing tumors of U87-RFP cells only, BB-1701 treatment at the same dose did not slow down tumor growth or reduce the increase in mean fluorescence intensity (Fig. 7D-F). The strong bystander effect observed in these results suggests that BB-1701 may be a promising treatment option for HER-low breast cancer and other indications with heterogeneous HER2 expression. BB-1701 induced ICD and the activation of the immune system Certain chemotherapy drugs, oncolytic viruses, and radiotherapy can stimulate the production of neoantigens and promote immunogenic cell death (ICD) in cancer cells, which is characterized by the release of molecules called damage-associated molecular patterns (DAMPs) that can stimulate the immune system (15). To determine if the treatment BB-1701 can induce ICD, BT-474 cells were exposed to increasing concentrations of either BB-1701 or eribulin. Expression of calreticulin, a type of DAMP, was found increased in a dose-dependent manner in BT-474 cells treated with either BB-1701 or eribulin (Fig.8A). The EC50 values (the concentration at which half-maximum calreticulin was induced) for BB-1701 and eribulin were similar to their cytotoxic IC50 values (the concentration at which 50% of the cells were killed) for this cell line (Fig. 8A). Additionally, the release of ATP, another type of DAMP, was transiently increased in
BT-474 cells treated with either BB-1701 or eribulin (Fig. 8B). HMGB1 (yet another type of DAMP) were also observed in a BT-474 xenograft model by analyzing formalin-fixed paraffin- embedded tissue sections with immunohistochemistry (Fig.8C). Finally, treatment with BB-1701 was found to increase the number of immune cells called macrophages in the tumor compared to treatment with a control substance (Fig. 8D). Pharmacokinetics and toxicity studies of BB-1701 in cynomolgus monkey To evaluate the pharmacokinetics and potential toxic effects of BB-1701 when given repeatedly at different doses, five male and five female cynomolgus monkeys (Aujun Biological Technology Co., Ltd., Guangzhou, China) were divided into groups and received four repeated doses (intravenous, every 3 weeks × 4) of BB-1701 at doses of 0, 2, 4, and 8 mg/kg. The animals were observed for clinical signs, and various laboratory tests were conducted to evaluate hematology, clinical chemistry, coagulation, and immunophenotype. They were euthanized one week after the last dose or at the end of the 6-week recovery period, after which gross pathology and histopathology evaluations were performed. Toxicokinetic (TK) studies were incorporated into the repeated dose toxicity study. Intensive blood sampling was performed in the first and third dose cycles, while sparse sampling was performed in the second and fourth dose cycles. In addition, cardiovascular and respiratory function were evaluated as part of the safety pharmacology assessment during the repeated dose toxicity study. The results showed that BB-1701 was stable in circulation and that its concentration in the blood was roughly proportional to the dose given (Fig.9, Table 1). System exposure of BB-1701 was achieved at all three dose levels. The half-life of BB-1701 was around 111-192h (Table 1), and there was no accumulation of the drug or development of neutralizing antibodies at any of the doses tested. Table 1. Serum pharmacokinetic parameters of ADC and Tab after administration of BB-1701 in cynomolgus monkeys.
asons due to a systemic bacterial infection on the 11th day of treatment, and one male monkey was found dead unexpectedly on the 13th day of treatment. The infection and death were likely caused by a weakened immune system and were accompanied by significant decreases in certain types of white blood cells, including neutrophils (NEUT) and lymphocytes (LYMP). Remarkable hematological toxicity was observed in all animals in this dose group, with WBC, NEUT, LYMP, and RET significantly reduced on day 7 after administration and then rebounded in each dosing cycle. Microscopic examination of the affected organs revealed immune organ toxicity, lymphoid depletion in the thymus, spleen, and lymph nodes (mandibular, mesenteric, inguinal), as well as Peyer's patches. The changes in the immune system were also accompanied by toxicity in the bone marrow, including decreases in the degree of cell proliferation and reduced numbers of myeloid cells. There was also an imbalance in the ratio of myeloid to erythroid cells (M:E ratio) in the bone marrow. These effects tended to recover or improve by the end of a six-week recovery period. At lower doses of 4 and 2 mg/kg, the monkeys experienced similar, but less severe, changes. These changes were considered not toxicologically important because they were within the normal range for the test facility and did not produce significant harmful effects. Based on the mortality and severity of the effects seen at the different doses, the highest non- severely toxic dose (HNSTD) for monkeys was determined to be 4 mg/kg. This means that BB- 1701 was well-tolerated at doses up to 4 mg/kg in monkeys when given repeatedly, and the nonclinical safety profile was acceptable for testing in humans.
The safety profile of ADCs is largely determined by their toxin payload. DS-8201a has been associated with approximately 10% interstitial lung disease (ILD) in breast cancer clinical studies, with 2.2% of deaths resulting from ILD (8, 10, 17). A repeated dose of DS-8201a to monkeys caused pulmonary toxicity at 30 mg/kg (given every 3 weeks for 3 doses) (16). T-DM1 has had low incidences of ILD in human patients and a preclinical toxicity study showed that T- DM1 treatment caused infiltration of mononuclear cells into the interstitium of the lung in cynomolgus monkeys at 10 mg/kg (given every 3 weeks for 4 doses). In the repeated dose toxicity study of BB-1701 in monkeys, there were no toxicities suggestive of ILD even at the highest dose groups. Therefore, despite the expression of HER2 on the cell membranes of epithelial cells in the human lung, it seems that the pulmonary effects of DS-8201a and T-DM1 may be toxin-specific and may be mediated by HER2 expression in the lung (18). The main adverse events of T-DM1 are thrombocytopenia and peripheral neuropathy, which are typical toxicities of tubulin inhibitor- conjugated ADCs (22). In the repeat-dose toxicology study of BB-1701 in cynomolgus monkeys, the main toxicities included histopathological changes in the thymus, spleen, lymph nodes, bone marrow, and male reproductive system at high doses, including two unscheduled deaths at the 8 mg/kg dose level due to immune system deficiency. This toxicity profile is consistent with that of eribulin in human patients and similar to other tubulin inhibitor-conjugated ADCs, such as MMAE-containing ADCs. Both primary and acquired resistance to current cancer therapies present challenges and opportunities for a new drug in development. The EMILIA study of T-DM1 in late stage HER2 positive breast cancer patients found an overall response rate of 43.6% and a progression-free survival of 9.6 months, indicating more than 50% primary resistance and an additional 50% acquired resistance within 10 months (20). The DESTINY-Breast003 studies of DS-8201a in the third line treatment of late stage HER2 positive breast cancer patients found impressive overall response rates of 69%, respectively, and progression-free survivals of 18.5 months, suggesting that primary resistance to DS-8201 may be relatively infrequent, but around 30% of DS-8201a-treated patients acquired resistance within one year and more than 50% acquired resistance within two years (10). DS-8201 has had lower response rates and shorter progression-free survival in other indications, presenting more severe resistance challenges (8). There are dozens of potential mechanisms of resistance to HER2-targeting ADCs, and as long as HER2 expression persists, ADCs with different mechanisms of action and/or different drug metabolism mechanisms may
offer the potential to overcome resistance challenges (21). BB-1701 treatment showed significant anti-tumor activity in DS-8201a-resistant model. In addition to its unique antimitotic and non- antimitotic mode of action, eribulin uses a completely different set of ATP-binding Cassette (ABC) transporters from those used by topotecan and diflomotecan (of the same drug family as DXd) for cytoplasmic membrane trafficking (22). Therefore, it is hopeful that BB-1701 may use nonoverlapping mechanisms to overcome drug resistance and may be effective against tumors with acquired resistance to other HER2-targeting ADCs. Part B: Clinical Evaluation Example 1: BB-1701 is effective in suppressing tumor growth in HER2 positive breast cancer patients HER2 positive breast cancer patients who had positive HER2 diagnosis (HER23+ by IHC, or 2+ by IHC plus FISH positive ), and had progressed on, or were intolerant to, or developed resistance to prior standard treatment, received BB-1701 treatment once every three weeks (Q3W) at 0.8 (n=3), 1.2 (n=2), 1.6 (n=6), 2.0 (n=8) mg/kg. At the tumor evaluation after the treatment, the target lesions of 66.7% patients showed ≥30% reduction, of 33.3% patients largely remain unchanged (<30% changes in size); no patient with target lesion grew >30% changes in size. Figures 10A and 10B are waterfall and swimmer plots of HER2 positive breast cancer patients. The study showed a best overall response rate of 66.7% (12/18 patients) and disease control rate of 100% (18/18 patients). The longest treatment duration was 60 weeks and 40.7% patients were on treatment for over 6 months. All patients received BB-1701 after they had failed treatment of standard therapies or experimental treatments, with prior therapy lines of treatment ranging from 1 to 6 lines. All patients had progressive diseases from their previous treatments when they were enrolled in the study with BB-1701. The tumor shrinkage and tumor growth suppression observed with the study treatment of BB-1701, was likely the results of the anti-tumor activity of BB-1701 in these HER2 positive breast cancer patients. Example 2: BB-1701 is effective in suppressing tumor growth in patients bearing HER2 low- expressing breast cancer
HER2 low-expressing breast cancer patients who had diagnosis of HER21+ by IHC, or 2+ by IHC plus FISH negative, and had progressed on at least two lines of prior standard therapies, received BB-1701 treatment intravenously once every three weeks (Q3W) at 1.0 (n=5), 1.2 (n=18), 1.4 (n=5), 1.6 (n=10) mg/kg. Figures 11A and 11B are waterfall and swimmer plots of HER2-low breast cancer patients. Tumor evaluation after the treatment showed that the target lesions of 34.2% patients showed ≥30% reduction, of 55.3% patients largely remain unchanged (<30% changes in size); in contrast the target lesions of only 10.5% patients grew >30% changes in size. The study showed a best overall response rate of 34.2% (13/38 patients) and disease control rate of 89.5% (34/38 patients). The longest treatment duration was 64 weeks and 31.6% patients were on treatment for over 6 months at the cutoff date of Sep 30th 2023 (study ongoing). Among these patients, some had failed other anti-HER2 ADC treatment. At 1.2 mg/kg dose level, one patient who achieved SD and the treatment duration is 62 weeks and is still ongoing was previously progressed on Disitamab Vedotin®. At 1.4 mg/kg dose level, one patient who achieved PR was previously progressed on TRODELVY®. At 1.6 mg/kg dose level, one patient who achieved CR was previously progressed on Disitamab Vedotin®, and one patient who achieved PR was previously progressed on ENHERTU®. All patients had failed standard therapies with prior lines of therapy ranging from 2 to 9 lines. All patients had progressive diseases from their previous treatments when they were enrolled in the study with BB-1701. The tumor shrinkage and tumor growth suppression observed with the study treatment of BB-1701, was likely the result of the anti-tumor activity of BB-1701 in these HER2 low expressing breast cancer patients. Example 3: BB-1701 is effective in suppressing tumor growth in patients bearing HER2 expressing Lung cancer HER2 expressing lung cancer patients who had HER2 amplification or mutation by NGS, and had progressed on at least two lines of prior standard therapies, received BB-1701 treatment intravenously once every three weeks (Q3W) at 1.0 (n=2), 1.2 (n=9), 1.4 (n=3), 1.6 (n=1) mg/kg. Figures 12A and 12B are waterfall and swimmer plots of HER2 expressing lung cancer patients. Tumor evaluation after the treatment has shown that the target lesions of 33.3% patients showed ≥30% reduction, 60.6% patients largely remain unchanged (<30% changes in size); in contrast the target lesions of only 6.7% patients grew >30% changes in size. At 1.2 mg/kg, the
study showed a best overall response rate of 66.7% (4/6 patients) and disease control rate of 100% (6/6 patients) for HER2 mutation NSCLC. The longest treatment duration was 52 weeks and 46.7% patients were on treatment for over 6 months at the cut-off date of Sep 30th 2023 (study ongoing). All patients had progressive diseases from their previous treatments when they were enrolled in the study with BB-1701. The tumor shrinkage and tumor growth suppression observed with the study treatment of BB-1701, was likely the result of the anti-tumor activity of BB-1701 in NSCLC patients with Her2 amplification or mutations. Example 4: BB-1701 is effective in suppressing tumor growth in patients with other HER2 expressing cancer types HER2 expressing cancer patients of gastric/esophagogastric junction cancer (n=6), colorectal cancer (n=4), urothelial carcinoma (n=1) who had HER2 expression status (HER23+ by IHC, or 2+ by IHC plus FISH positive, or HER2 amplification by NGS), and had progressed on at least two lines of prior standard therapies, received BB-1701 treatment intravenously once every three weeks (Q3W) at 0.4 (n=1), 1.2 (n=6), 1.6 (n=1), 2.0 (n=1), 2.6 (n=2) mg/kg. Tumor evaluation after the treatment has shown that, the target lesions of the only one urothelial carcinoma patient had 65.7% reduction; the target lesions of the only one parotid cancer patient had 48.6% reduction; the target lesions of 5/6 gastric/esophagogastric junction cancer patients remained largely unchanged (<30% changes in size), only 1/6 patients with the target lesions grew >30% changes in size. The longest treatment duration of gastric/esophagogastric junction cancer patients was 63 weeks. All patients had progressive diseases from their previous treatments when they were enrolled in the study with BB-1701. The tumor shrinkage and tumor growth suppression observed with the study treatment of BB-1701, was likely the result of the anti-tumor activity of BB-1701 in the patients with different Her2 positive cancers. References 1. Oh, D. Y., & Bang, Y. J. (2020). HER2-targeted therapies - a role beyond breast cancer. Nature reviews. Clinical oncology, 17(1), 33–48. https://doi.org/10.1038/s41571-019- 0268-3
2. Yu, S., Liu, Q., Han, X., Qin, S., Zhao, W., Li, A., & Wu, K. (2017). Development and clinical application of anti-HER2 monoclonal and bispecific antibodies for cancer treatment. Experimental hematology & oncology, 6, 31. https://doi.org/10.1186/s40164-017-0091-4. 3. Barbier, L., Declerck, P., Simoens, S., Neven, P., Vulto, A. G., & Huys, I. (2019). The arrival of biosimilar monoclonal antibodies in oncology: clinical studies for trastuzumab biosimilars. British journal of cancer, 121(3), 199–210. https://doi.org/10.1038/s41416-019- 0480-z. 4. Zhang, X., Huang, A. C., Chen, F., Chen, H., Li, L., Kong, N., Luo, W., & Fang, J. (2022). Novel development strategies and challenges for anti-Her2 antibody-drug conjugates. Antibody therapeutics, 5(1), 18–29. https://doi.org/10.1093/abt/tbac001. 5. Wynn, C. S., & Tang, S. C. (2022). Anti-HER2 therapy in metastatic breast cancer: many choices and future directions. Cancer metastasis reviews, 41(1), 193–209. https://doi.org/10.1007/s10555-022-10021-x. 6. Ferraro, E., Drago, J. Z., & Modi, S. (2021). Implementing antibody-drug conjugates (ADCs) in HER2-positive breast cancer: state of the art and future directions. Breast cancer research : BCR, 23(1), 84. https://doi.org/10.1186/s13058-021-01459-y. 7. Rinnerthaler, G., Gampenrieder, S. P., & Greil, R. (2019). HER2 Directed Antibody- Drug-Conjugates beyond T-DM1 in Breast Cancer. International journal of molecular sciences, 20(5), 1115. https://doi.org/10.3390/ijms20051115. 8. Li, B. T., Smit, E. F., Goto, Y., Nakagawa, K., Udagawa, H., Mazières, J., Nagasaka, M., Bazhenova, L., Saltos, A. N., Felip, E., Pacheco, J. M., Pérol, M., Paz-Ares, L., Saxena, K., Shiga, R., Cheng, Y., Acharyya, S., Vitazka, P., Shahidi, J., Planchard, D., … DESTINY- Lung01 Trial Investigators (2022). Trastuzumab Deruxtecan in HER2-Mutant Non-Small- Cell Lung Cancer. The New England journal of medicine, 386(3), 241–251. https://doi.org/10.1056/NEJMoa2112431.
9. Sheng, X., Yan, X., Wang, L., Shi, Y., Yao, X., Luo, H., Shi, B., Liu, J., He, Z., Yu, G., Ying, J., Han, W., Hu, C., Ling, Y., Chi, Z., Cui, C., Si, L., Fang, J., Zhou, A., & Guo, J. (2021). Open-label, Multicenter, Phase II Study of RC48-ADC, a HER2-Targeting Antibody- Drug Conjugate, in Patients with Locally Advanced or Metastatic Urothelial Carcinoma. Clinical cancer research : an official journal of the American Association for Cancer Research, 27(1), 43–51. https://doi.org/10.1158/1078-0432.CCR-20-2488. 10. Cortés, J., Kim, S. B., Chung, W. P., Im, S. A., Park, Y. H., Hegg, R., Kim, M. H., Tseng, L. M., Petry, V., Chung, C. F., Iwata, H., Hamilton, E., Curigliano, G., Xu, B., Huang, C. S., Kim, J. H., Chiu, J. W. Y., Pedrini, J. L., Lee, C., Liu, Y., … DESTINY-Breast03 Trial Investigators (2022). Trastuzumab Deruxtecan versus Trastuzumab Emtansine for Breast Cancer. The New England journal of medicine, 386(12), 1143–1154. https://doi.org/10.1056/NEJMoa2115022. 11. Modi, S., Jacot, W., Yamashita, T., Sohn, J., Vidal, M., Tokunaga, E., Tsurutani, J., Ueno, N. T., Prat, A., Chae, Y. S., Lee, K. S., Niikura, N., Park, Y. H., Xu, B., Wang, X., Gil- Gil, M., Li, W., Pierga, J. Y., Im, S. A., Moore, H. C. F., … DESTINY-Breast04 Trial Investigators (2022). Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer. The New England journal of medicine, 387(1), 9 – 20. https://doi.org/10.1056/NEJMoa2203690。 12. Jain, S., & Cigler, T. (2012). Eribulin mesylate in the treatment of metastatic breast cancer. Biologics : targets & therapy, 6, 21–29. https://doi.org/10.2147/BTT.S19811. 13. Cortes, J., Schöffski, P., & Littlefield, B. A. (2018). Multiple modes of action of eribulin mesylate: Emerging data and clinical implications. Cancer treatment reviews, 70, 190–198. https://doi.org/10.1016/j.ctrv.2018.08.008. 14. Gikanga, B., Adeniji, N. S., Patapoff, T. W., Chih, H. W., & Yi, L. (2016). Cathepsin B Cleavage of vcMMAE-Based Antibody-Drug Conjugate Is Not Drug Location or Monoclonal Antibody Carrier Specific. Bioconjugate chemistry, 27(4), 1040–1049. https://doi.org/10.1021/acs.bioconjchem.6b00055.
15. Bauzon, M., Drake, P. M., Barfield, R. M., Cornali, B. M., Rupniewski, I., & Rabuka, D. (2019). Maytansine-bearing antibody-drug conjugates induce in vitro hallmarks of immunogenic cell death selectively in antigen-positive target cells. Oncoimmunology, 8(4), e1565859. https://doi.org/10.1080/2162402X.2019.1565859. 16. Ogitani, Y., Aida, T., Hagihara, K., Yamaguchi, J., Ishii, C., Harada, N., Soma, M., Okamoto, H., Oitate, M., Arakawa, S., Hirai, T., Atsumi, R., Nakada, T., Hayakawa, I., Abe, Y., & Agatsuma, T. (2016). DS-8201a, A Novel HER2-Targeting ADC with a Novel DNA Topoisomerase I Inhibitor, Demonstrates a Promising Antitumor Efficacy with Differentiation from T-DM1. Clinical cancer research : an official journal of the American Association for Cancer Research, 22(20), 5097–5108. https://doi.org/10.1158/1078- 0432.CCR-15-2822.
Leighton, J. K. (2015). An FDA oncology analysis of antibody-drug conjugates. Regulatory toxicology and pharmacology: RTP, 71(3), 444–452. https://doi.org/10.1016/j.yrtph.2015.01.014.
Slamon, D. J. (1990). Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissues. Oncogene, 5(7), 953–962. 19. Hackshaw, M. D., Danysh, H. E., Singh, J., Ritchey, M. E., Ladner, A., Taitt, C., Camidge, D. R., Iwata, H., & Powell, C. A. (2020). Incidence of pneumonitis/interstitial lung disease induced by HER2-targeting therapy for HER2-positive metastatic breast cancer. Breast cancer research and treatment, 183(1), 23–39. https://doi.org/10.1007/s10549-020- 05754-8. 20. Diéras, V., Miles, D., Verma, S., Pegram, M., Welslau, M., Baselga, J., Krop, I. E., Blackwell, K., Hoersch, S., Xu, J., Green, M., & Gianni, L. (2017). Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final overall survival results from a 20 andomized, open-label, phase 3 trial. The Lancet. Oncology, 18(6), 732–742. https://doi.org/10.1016/S1470-2045(17)30312-1.
21. Schlam, I., Tarantino, P., & Tolaney, S. M. (2022). Overcoming Resistance to HER2- Directed Therapies in Breast Cancer. Cancers, 14(16), 3996. https://doi.org/10.3390/cancers14163996. 22. Morisaki, K., Robey, R. W., Ozvegy-Laczka, C., Honjo, Y., Polgar, O., Steadman, K., Sarkadi, B., & Bates, S. E. (2005). Single nucleotide polymorphisms modify the transporter activity of ABCG2. Cancer chemotherapy and pharmacology, 56(2), 161–172. https://doi.org/10.1007/s00280-004-0931-x. The disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated herein by reference in their entirety. While the invention has been described above in connection with one or more embodiments, it should be understood that the invention is not limited to those embodiments, and the description is intended to cover all alternatives, modifications, and equivalents, as may be included within the spirit and scope of the appended claims.
Claims
CLAIMS 1. A method of treating a patient bearing a HER2-expressing caner, comprising: administering to a patient a therapeutically effective amount of an antibody-drug conjugate (ADC) having the Formula I: Ab-(L-D)p (I) wherein Ab is an anti-HER2 antibody moiety having the same structure as trastuzumab, L is a cleavable linker comprising Mal-(PEG)2-Val-Cit-pAB, D is eribulin, and p is an integer from 1 to 8 inclusive, wherein the HER2-expressing cancer is selected from the group consisting of non- small cell lung cancer, gastric/esophagogastric junction cancer, urothelial carcinoma, parotid cancer, and HER2-low expressing breast cancer. 2. The method of claim 1, the patient is one whose cancer has demonstrated resistance to other HER2-targeting therapeutics including other anti-HER2 antibody drug conjugate treatment. 3. The method of claim 1, wherein the cancer is HER2-low expressing breast cancer, and where HER2-expression has been determined using HER21+ by IHC, or 2+ by IHC plus FISH negative diagnosis.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202363484699P | 2023-02-13 | 2023-02-13 | |
US63/484,699 | 2023-02-13 | ||
US202363590731P | 2023-10-16 | 2023-10-16 | |
US63/590,731 | 2023-10-16 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024173281A1 true WO2024173281A1 (en) | 2024-08-22 |
Family
ID=92420633
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2024/015450 WO2024173281A1 (en) | 2023-02-13 | 2024-02-12 | Antibody-drug conjugate for cancer treatment |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024173281A1 (en) |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20170252458A1 (en) * | 2016-03-02 | 2017-09-07 | Eisai Co., Ltd. | Eribulin-based antibody-drug conjugates and methods of use |
-
2024
- 2024-02-12 WO PCT/US2024/015450 patent/WO2024173281A1/en unknown
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20170252458A1 (en) * | 2016-03-02 | 2017-09-07 | Eisai Co., Ltd. | Eribulin-based antibody-drug conjugates and methods of use |
Non-Patent Citations (1)
Title |
---|
GUIDI LORENZO, PELLIZZARI GLORIA, TARANTINO PAOLO, VALENZA CARMINE, CURIGLIANO GIUSEPPE: "Resistance to Antibody-Drug Conjugates Targeting HER2 in Breast Cancer: Molecular Landscape and Future Challenges", CANCERS, vol. 15, no. 4, 1 January 2023 (2023-01-01), CH , pages 1 - 14, XP093205405, ISSN: 2072-6694, DOI: 10.3390/cancers15041130 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7458981B2 (en) | Combination of antibody-drug conjugate and tubulin inhibitor | |
CN108601828B (en) | Therapeutic combinations comprising anti-FOLR 1 immunoconjugates | |
JP7608164B2 (en) | Combination of antibody-drug conjugates with PARP inhibitors | |
US10792370B2 (en) | Antibody-drug conjugate | |
US10583151B2 (en) | Polymalic acid-based nanobiopolymer compositions | |
TW202011998A (en) | Treatment for metastatic brain tumor comprising administering antibody-drug conjugate | |
Cho et al. | Sustained potentiation of bystander killing via PTEN-loss driven macropinocytosis targeted peptide-drug conjugate therapy in metastatic triple-negative breast cancer | |
TW202216208A (en) | Combination of antibody-drug conjugate and atr inhibitor | |
WO2024173281A1 (en) | Antibody-drug conjugate for cancer treatment | |
Wang et al. | Preclinical studies of BB-1701, a HER2-targeting eribulin-containing ADC with potent bystander effect and ICD activity | |
KR20240130081A (en) | Combination use of anti-TROP-2 antibody-drug conjugates with additional therapeutics | |
TW202440169A (en) | Combination of antibody-drug conjugates and dnmt inhibitors | |
US20170333570A1 (en) | Egfr antibody-based combination therapy | |
Teicher | Antibody drug and radionuclide conjugates for GI cancers | |
KR20240130087A (en) | Combination of antibody-drug conjugates and ATR inhibitors | |
US20250049940A1 (en) | Combined Use of Anti-TROP-2 Antibody-drug Conjugate and other Therapeutic Agents | |
WO2024191826A1 (en) | Methods of treating cancer using anti-her2 antibody-drug conjugates and her2 kinase inhibitors | |
Gajdosik | Depatuxizumab mafodotin. Anti-egfr antibody-drug conjugate, treatment of glioblastoma multiforme | |
EA046932B1 (en) | TREATMENT OF METASTATIC BRAIN TUMOR BY ADMINISTRATION OF ANTIBODY-DRUG CONJUGATE | |
Andrikopoulou et al. | Amisulpride is an antiemetic and antipsychotic medication used at lower doses intravenously to prevent and treat postoperative nausea and vomiting; and at higher doses by mouth to treat schizophrenia and acute psychotic episodes Menu |
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: 24757504 Country of ref document: EP Kind code of ref document: A1 |