Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2021

Open Access 01-12-2021 | NSCLC | Review

Antibody–drug conjugates in solid tumors: a look into novel targets

Authors: Carmen Criscitiello, Stefania Morganti, Giuseppe Curigliano

Published in: Journal of Hematology & Oncology | Issue 1/2021

Login to get access

Abstract

Antibody–drug conjugates (ADCs) are a relatively new class of anticancer agents designed to merge the selectivity of monoclonal antibodies with cell killing properties of chemotherapy. They are commonly described as the “Trojan Horses” of therapeutic armamentarium, because of their capability of directly conveying cytotoxic drug (payloads) into the tumor space, thus transforming chemotherapy into a targeted agent. Three novel ADCs have been recently approved, i.e., trastuzumab deruxtecan, sacituzumab govitecan and enfortumab vedotin, respectively, targeting HER2, Trop2 and Nectin4. Thanks to progressive advances in engineering technologies these drugs rely on, the spectrum of diseases sensitive to these drugs as well as their indications are in continuous expansion. Several novel ADCs are under evaluation, exploring new potential targets along with innovative payloads. This review aims at providing a summary of the technology behind these compounds and at presenting the latest ADCs approved in solid tumors, as well as at describing novel targets for ADCs under investigation and new strategies to optimize their efficacy in solid tumors.
Literature
1.
go back to reference Staudacher AH, Brown MP. Antibody drug conjugates and bystander killing: is antigen-dependent internalisation required? Br J Cancer. 2017;117:1736–42.PubMedPubMedCentralCrossRef Staudacher AH, Brown MP. Antibody drug conjugates and bystander killing: is antigen-dependent internalisation required? Br J Cancer. 2017;117:1736–42.PubMedPubMedCentralCrossRef
2.
go back to reference Beck A, Goetsch L, Dumontet C, Corvaïa N. Strategies and challenges for the next generation of antibody–drug conjugates. Nat Rev Drug Discov. 2017;16:315–37.PubMedCrossRef Beck A, Goetsch L, Dumontet C, Corvaïa N. Strategies and challenges for the next generation of antibody–drug conjugates. Nat Rev Drug Discov. 2017;16:315–37.PubMedCrossRef
3.
4.
go back to reference Boni V, Sharma MR, Patnaik A. The resurgence of antibody drug conjugates in cancer therapeutics: novel targets and payloads. Am Soc Clin Oncol Educ B. 2020;40:e58–74.CrossRef Boni V, Sharma MR, Patnaik A. The resurgence of antibody drug conjugates in cancer therapeutics: novel targets and payloads. Am Soc Clin Oncol Educ B. 2020;40:e58–74.CrossRef
5.
go back to reference Zhang A, Fang J, Chou RY-T, Bondarenko PV, Zhang Z. Conformational difference in human IgG2 disulfide isoforms revealed by hydrogen/deuterium exchange mass spectrometry. Biochemistry. 2015;54:1956–62.PubMedCrossRef Zhang A, Fang J, Chou RY-T, Bondarenko PV, Zhang Z. Conformational difference in human IgG2 disulfide isoforms revealed by hydrogen/deuterium exchange mass spectrometry. Biochemistry. 2015;54:1956–62.PubMedCrossRef
6.
go back to reference Tsuchikama K, An Z. Antibody–drug conjugates: recent advances in conjugation and linker chemistries. Protein Cell. 2018;9:33–46.PubMedCrossRef Tsuchikama K, An Z. Antibody–drug conjugates: recent advances in conjugation and linker chemistries. Protein Cell. 2018;9:33–46.PubMedCrossRef
8.
go back to reference Bargh JD, Isidro-Llobet A, Parker JS, Spring DR. Cleavable linkers in antibody–drug conjugates. Chem Soc Rev. 2019;48:4361–74.PubMedCrossRef Bargh JD, Isidro-Llobet A, Parker JS, Spring DR. Cleavable linkers in antibody–drug conjugates. Chem Soc Rev. 2019;48:4361–74.PubMedCrossRef
10.
go back to reference Shor B, Gerber HP, Sapra P. Preclinical and clinical development of inotuzumab-ozogamicin in hematological malignancies. Mol Immunol. 2015;67:107–16.PubMedCrossRef Shor B, Gerber HP, Sapra P. Preclinical and clinical development of inotuzumab-ozogamicin in hematological malignancies. Mol Immunol. 2015;67:107–16.PubMedCrossRef
11.
go back to reference Doronina SO, Toki BE, Torgov MY, et al. Development of potent monoclonal antibody auristatin conjugates for cancer therapy. Nat Biotechnol. 2003;21:778–84.PubMedCrossRef Doronina SO, Toki BE, Torgov MY, et al. Development of potent monoclonal antibody auristatin conjugates for cancer therapy. Nat Biotechnol. 2003;21:778–84.PubMedCrossRef
12.
go back to reference Oroudjev E, Lopus M, Wilson L, Audette C, Provenzano C, Erickson H, Kovtun Y, Chari R, Jordan MA. Maytansinoid-antibody conjugates induce mitotic arrest by suppressing microtubule dynamic instability. Mol Cancer Ther. 2010;9:2700–13.PubMedPubMedCentralCrossRef Oroudjev E, Lopus M, Wilson L, Audette C, Provenzano C, Erickson H, Kovtun Y, Chari R, Jordan MA. Maytansinoid-antibody conjugates induce mitotic arrest by suppressing microtubule dynamic instability. Mol Cancer Ther. 2010;9:2700–13.PubMedPubMedCentralCrossRef
14.
go back to reference Hynes NE, Stern DF. The biology of erbB-2/nue/HER-2 and its role in cancer. BBA Rev Cancer. 1994;1198:165–84. Hynes NE, Stern DF. The biology of erbB-2/nue/HER-2 and its role in cancer. BBA Rev Cancer. 1994;1198:165–84.
15.
go back to reference Alroy I, Yarden Y. The ErbB signaling network in embryogenesis and oncogenesis: signal diversification through combinatorial ligand-receptor interactions. FEBS Lett. 1997;410:83–6.PubMedCrossRef Alroy I, Yarden Y. The ErbB signaling network in embryogenesis and oncogenesis: signal diversification through combinatorial ligand-receptor interactions. FEBS Lett. 1997;410:83–6.PubMedCrossRef
16.
17.
go back to reference Mimura K, Kono K, Hanawa M, Mitsui F, Sugai H, Miyagawa N, Ooi A, Fujii H. Frequencies of HER-2/neu expression and gene amplification in patients with oesophageal squamous cell carcinoma. Br J Cancer. 2005;92:1253–60.PubMedPubMedCentralCrossRef Mimura K, Kono K, Hanawa M, Mitsui F, Sugai H, Miyagawa N, Ooi A, Fujii H. Frequencies of HER-2/neu expression and gene amplification in patients with oesophageal squamous cell carcinoma. Br J Cancer. 2005;92:1253–60.PubMedPubMedCentralCrossRef
18.
go back to reference Yano T, Ochiai A, Doi T, Hashizume K, Nakanishi M, Ouchi K, Tanaka Y, Ohtsu A. Expression of HER2 in gastric cancer: comparison between protein expression and gene amplification using a new commercial kit. J Clin Oncol. 2004;22:4053–4053.CrossRef Yano T, Ochiai A, Doi T, Hashizume K, Nakanishi M, Ouchi K, Tanaka Y, Ohtsu A. Expression of HER2 in gastric cancer: comparison between protein expression and gene amplification using a new commercial kit. J Clin Oncol. 2004;22:4053–4053.CrossRef
20.
go back to reference Meric-Bernstam F, Johnson AM, Ileana Dumbrava EE, Raghav K, Balaji K, Bhatt M, Murthy RK, Rodon J, Piha-Paul SA. Advances in HER2-targeted therapy: novel agents and opportunities beyond breast and gastric cancer. Clin Cancer Res. 2019;25:2033–41.PubMedCrossRef Meric-Bernstam F, Johnson AM, Ileana Dumbrava EE, Raghav K, Balaji K, Bhatt M, Murthy RK, Rodon J, Piha-Paul SA. Advances in HER2-targeted therapy: novel agents and opportunities beyond breast and gastric cancer. Clin Cancer Res. 2019;25:2033–41.PubMedCrossRef
22.
go back to reference Krop IE, Kim S-B, González-Martín A, LoRusso PM, Ferrero J-M, Smitt M, Yu R, Leung ACF, Wildiers H. Trastuzumab emtansine versus treatment of physician’s choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15:689–99.PubMedCrossRef Krop IE, Kim S-B, González-Martín A, LoRusso PM, Ferrero J-M, Smitt M, Yu R, Leung ACF, Wildiers H. Trastuzumab emtansine versus treatment of physician’s choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15:689–99.PubMedCrossRef
23.
go back to reference von Minckwitz G, Huang C-S, Mano MS, et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 2019;380:617–28.CrossRef von Minckwitz G, Huang C-S, Mano MS, et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 2019;380:617–28.CrossRef
24.
go back to reference Ogitani Y, Aida T, Hagihara K, et al. DS-8201a, a novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising antitumor efficacy with differentiation from T-DM1. Clin Cancer Res. 2016;22:5097–108.PubMedCrossRef Ogitani Y, Aida T, Hagihara K, et al. DS-8201a, a novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising antitumor efficacy with differentiation from T-DM1. Clin Cancer Res. 2016;22:5097–108.PubMedCrossRef
25.
go back to reference Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody–drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci. 2016;107:1039–46.PubMedPubMedCentralCrossRef Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody–drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci. 2016;107:1039–46.PubMedPubMedCentralCrossRef
26.
go back to reference Modi S, Saura C, Yamashita T, et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N Engl J Med. 2020;382:610–21.PubMedCrossRef Modi S, Saura C, Yamashita T, et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N Engl J Med. 2020;382:610–21.PubMedCrossRef
27.
go back to reference Jerusalem G, Park YH, Yamashita T, Hurvitz SA, Chen S, Cathcart J, Lee C, Perrin C. 138O CNS metastases in HER2-positive metastatic breast cancer treated with trastuzumab deruxtecan: DESTINY-Breast01 subgroup analyses. Ann Oncol. 2020;31:S63–4.CrossRef Jerusalem G, Park YH, Yamashita T, Hurvitz SA, Chen S, Cathcart J, Lee C, Perrin C. 138O CNS metastases in HER2-positive metastatic breast cancer treated with trastuzumab deruxtecan: DESTINY-Breast01 subgroup analyses. Ann Oncol. 2020;31:S63–4.CrossRef
28.
go back to reference Bang Y-J, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687–97.PubMedCrossRef Bang Y-J, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687–97.PubMedCrossRef
29.
go back to reference Pietrantonio F, Fuca G, Morano F, et al. Biomarkers of primary resistance to trastuzumab in HER2-positive metastatic gastric cancer patients: the AMNESIA case-control study. Clin Cancer Res. 2018;24:1082–9.PubMedCrossRef Pietrantonio F, Fuca G, Morano F, et al. Biomarkers of primary resistance to trastuzumab in HER2-positive metastatic gastric cancer patients: the AMNESIA case-control study. Clin Cancer Res. 2018;24:1082–9.PubMedCrossRef
30.
go back to reference Shitara K, Bang Y-J, Iwasa S, et al. Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer. N Engl J Med. 2020;382:2419–30.PubMedCrossRef Shitara K, Bang Y-J, Iwasa S, et al. Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer. N Engl J Med. 2020;382:2419–30.PubMedCrossRef
31.
go back to reference Smit EF, Nakagawa K, Nagasaka M, et al. Trastuzumab deruxtecan (T-DXd; DS-8201) in patients with HER2-mutated metastatic non-small cell lung cancer (NSCLC): interim results of DESTINY-Lung01. J Clin Oncol. 2020;38:9504–9504.CrossRef Smit EF, Nakagawa K, Nagasaka M, et al. Trastuzumab deruxtecan (T-DXd; DS-8201) in patients with HER2-mutated metastatic non-small cell lung cancer (NSCLC): interim results of DESTINY-Lung01. J Clin Oncol. 2020;38:9504–9504.CrossRef
32.
go back to reference Siena S, Di Bartolomeo M, Raghav KPS, et al. A phase II, multicenter, open-label study of trastuzumab deruxtecan (T-DXd; DS-8201) in patients (pts) with HER2-expressing metastatic colorectal cancer (mCRC): DESTINY-CRC01. J Clin Oncol. 2020;38:4000–4000.CrossRef Siena S, Di Bartolomeo M, Raghav KPS, et al. A phase II, multicenter, open-label study of trastuzumab deruxtecan (T-DXd; DS-8201) in patients (pts) with HER2-expressing metastatic colorectal cancer (mCRC): DESTINY-CRC01. J Clin Oncol. 2020;38:4000–4000.CrossRef
33.
go back to reference Banerji U, van Herpen CML, Saura C, et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 2019;20:1124–35.PubMedCrossRef Banerji U, van Herpen CML, Saura C, et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 2019;20:1124–35.PubMedCrossRef
34.
go back to reference Takai Y, Miyoshi J, Ikeda W, Ogita H. Nectins and nectin-like molecules: roles in contact inhibition of cell movement and proliferation. Nat Rev Mol Cell Biol. 2008;9:603–15.PubMedCrossRef Takai Y, Miyoshi J, Ikeda W, Ogita H. Nectins and nectin-like molecules: roles in contact inhibition of cell movement and proliferation. Nat Rev Mol Cell Biol. 2008;9:603–15.PubMedCrossRef
35.
go back to reference Nishiwada S, Sho M, Yasuda S, Shimada K, Yamato I, Akahori T, Kinoshita S, Nagai M, Konishi N, Nakajima Y. Nectin-4 expression contributes to tumor proliferation, angiogenesis and patient prognosis in human pancreatic cancer. J Exp Clin Cancer Res. 2015;34:30.PubMedPubMedCentralCrossRef Nishiwada S, Sho M, Yasuda S, Shimada K, Yamato I, Akahori T, Kinoshita S, Nagai M, Konishi N, Nakajima Y. Nectin-4 expression contributes to tumor proliferation, angiogenesis and patient prognosis in human pancreatic cancer. J Exp Clin Cancer Res. 2015;34:30.PubMedPubMedCentralCrossRef
36.
go back to reference Athanassiadou AM, Patsouris E, Tsipis A, Gonidi M, Athanassiadou P. The significance of survivin and nectin-4 expression in the prognosis of breast carcinoma. Folia Histochem Cytobiol. 2011;49:26–33.PubMedCrossRef Athanassiadou AM, Patsouris E, Tsipis A, Gonidi M, Athanassiadou P. The significance of survivin and nectin-4 expression in the prognosis of breast carcinoma. Folia Histochem Cytobiol. 2011;49:26–33.PubMedCrossRef
37.
go back to reference Takano A, Ishikawa N, Nishino R, et al. Identification of Nectin-4 oncoprotein as a diagnostic and therapeutic target for lung cancer. Cancer Res. 2009;69:6694–703.PubMedCrossRef Takano A, Ishikawa N, Nishino R, et al. Identification of Nectin-4 oncoprotein as a diagnostic and therapeutic target for lung cancer. Cancer Res. 2009;69:6694–703.PubMedCrossRef
38.
go back to reference DeRycke MS, Pambuccian SE, Gilks CB, et al. Nectin 4 overexpression in ovarian cancer tissues and serum: Potential role as a serum biomarker. Am J Clin Pathol. 2010;134:835–45.PubMedCrossRef DeRycke MS, Pambuccian SE, Gilks CB, et al. Nectin 4 overexpression in ovarian cancer tissues and serum: Potential role as a serum biomarker. Am J Clin Pathol. 2010;134:835–45.PubMedCrossRef
39.
go back to reference Rosenberg JE, O’Donnell PH, Balar AV, et al. Pivotal trial of enfortumab vedotin in urothelial carcinoma after platinum and anti-programmed death 1/programmed death ligand 1 therapy. J Clin Oncol. 2019;37:2592–600.PubMedPubMedCentralCrossRef Rosenberg JE, O’Donnell PH, Balar AV, et al. Pivotal trial of enfortumab vedotin in urothelial carcinoma after platinum and anti-programmed death 1/programmed death ligand 1 therapy. J Clin Oncol. 2019;37:2592–600.PubMedPubMedCentralCrossRef
40.
go back to reference Rosenberg J, Sridhar SS, Zhang J, et al. EV-101: a phase I study of single-agent enfortumab vedotin in patients with nectin-4-positive solid tumors, including metastatic urothelial carcinoma. J Clin Oncol. 2020;38:1041–9.PubMedPubMedCentralCrossRef Rosenberg J, Sridhar SS, Zhang J, et al. EV-101: a phase I study of single-agent enfortumab vedotin in patients with nectin-4-positive solid tumors, including metastatic urothelial carcinoma. J Clin Oncol. 2020;38:1041–9.PubMedPubMedCentralCrossRef
41.
go back to reference Drakaki A, Kirby CJ, Van Der Heijden MS, et al. Docetaxel with or without ramucirumab after immune checkpoint inhibition in platinum-refractory metastatic urothelial carcinoma (mUC): prespecified subgroup analysis from the phase 3 RANGE trial. J Clin Oncol. 2018;36:434–434.CrossRef Drakaki A, Kirby CJ, Van Der Heijden MS, et al. Docetaxel with or without ramucirumab after immune checkpoint inhibition in platinum-refractory metastatic urothelial carcinoma (mUC): prespecified subgroup analysis from the phase 3 RANGE trial. J Clin Oncol. 2018;36:434–434.CrossRef
42.
go back to reference Bellmunt J, Théodore C, Demkov T, et al. Phase III trial of vinflunine plus best supportive care compared with best supportive care alone after a platinum-containing regimen in patients with advanced transitional cell carcinoma of the urothelial tract. J Clin Oncol. 2009;27:4454–61.PubMedCrossRef Bellmunt J, Théodore C, Demkov T, et al. Phase III trial of vinflunine plus best supportive care compared with best supportive care alone after a platinum-containing regimen in patients with advanced transitional cell carcinoma of the urothelial tract. J Clin Oncol. 2009;27:4454–61.PubMedCrossRef
44.
go back to reference Masters JC, Nickens DJ, Xuan D, Shazer RL, Amantea M. Clinical toxicity of antibody drug conjugates: a meta-analysis of payloads. Investig New Drugs. 2018;36:121–35.CrossRef Masters JC, Nickens DJ, Xuan D, Shazer RL, Amantea M. Clinical toxicity of antibody drug conjugates: a meta-analysis of payloads. Investig New Drugs. 2018;36:121–35.CrossRef
45.
46.
go back to reference Lipinski M, Parks DR, Rouse RV, Herzenberg LA. Human trophoblast cell-surface antigens defined by monoclonal antibodies. Proc Natl Acad Sci USA. 1981;78:5147–50.PubMedCrossRefPubMedCentral Lipinski M, Parks DR, Rouse RV, Herzenberg LA. Human trophoblast cell-surface antigens defined by monoclonal antibodies. Proc Natl Acad Sci USA. 1981;78:5147–50.PubMedCrossRefPubMedCentral
47.
go back to reference Stepan LP, Trueblood ES, Hale K, Babcook J, Borges L, Sutherland CL. Expression of Trop2 cell surface glycoprotein in normal and tumor tissues: potential implications as a cancer therapeutic target. J Histochem Cytochem. 2011;59:701–10.PubMedPubMedCentralCrossRef Stepan LP, Trueblood ES, Hale K, Babcook J, Borges L, Sutherland CL. Expression of Trop2 cell surface glycoprotein in normal and tumor tissues: potential implications as a cancer therapeutic target. J Histochem Cytochem. 2011;59:701–10.PubMedPubMedCentralCrossRef
48.
go back to reference Guerra E, Trerotola M, Aloisi AL, Tripaldi R, Vacca G, La Sorda R, Lattanzio R, Piantelli M, Alberti S. The Trop-2 signalling network in cancer growth. Oncogene. 2013;32:1594–600.PubMedCrossRef Guerra E, Trerotola M, Aloisi AL, Tripaldi R, Vacca G, La Sorda R, Lattanzio R, Piantelli M, Alberti S. The Trop-2 signalling network in cancer growth. Oncogene. 2013;32:1594–600.PubMedCrossRef
49.
go back to reference Goldenberg DM, Cardillo TM, Govindan SV, Rossi EA, Sharkey RM. Trop-2 is a novel target for solid cancer therapy with sacituzumab govitecan (IMMU-132), an antibody–drug conjugate (ADC). Oncotarget. 2015;6:22496–512.PubMedPubMedCentralCrossRef Goldenberg DM, Cardillo TM, Govindan SV, Rossi EA, Sharkey RM. Trop-2 is a novel target for solid cancer therapy with sacituzumab govitecan (IMMU-132), an antibody–drug conjugate (ADC). Oncotarget. 2015;6:22496–512.PubMedPubMedCentralCrossRef
50.
go back to reference Bardia A, Mayer IA, Vahdat LT, et al. Sacituzumab govitecan-hziy in refractory metastatic triple-negative breast cancer. N Engl J Med. 2019;380:741–51.PubMedCrossRef Bardia A, Mayer IA, Vahdat LT, et al. Sacituzumab govitecan-hziy in refractory metastatic triple-negative breast cancer. N Engl J Med. 2019;380:741–51.PubMedCrossRef
51.
go back to reference Bardia A, Tolaney SM, Loirat D, et al. LBA17 ASCENT: a randomized phase III study of sacituzumab govitecan (SG) vs treatment of physician’s choice (TPC) in patients (pts) with previously treated metastatic triple-negative breast cancer (mTNBC). Ann Oncol. 2020;31:S1149–50.CrossRef Bardia A, Tolaney SM, Loirat D, et al. LBA17 ASCENT: a randomized phase III study of sacituzumab govitecan (SG) vs treatment of physician’s choice (TPC) in patients (pts) with previously treated metastatic triple-negative breast cancer (mTNBC). Ann Oncol. 2020;31:S1149–50.CrossRef
52.
go back to reference Bardia A, Mayer IA, Diamond JR, et al. Efficacy & safety of anti-Trop-2 antibody drug conjugate sacituzumab govitecan (IMMU-132) in heavily pretreated patients with metastatic triple-negative breast cancer. J Clin Oncol. 2017;35:2141–8.PubMedPubMedCentralCrossRef Bardia A, Mayer IA, Diamond JR, et al. Efficacy & safety of anti-Trop-2 antibody drug conjugate sacituzumab govitecan (IMMU-132) in heavily pretreated patients with metastatic triple-negative breast cancer. J Clin Oncol. 2017;35:2141–8.PubMedPubMedCentralCrossRef
54.
go back to reference Tagawa ST, Faltas BM, Lam ET, et al. Sacituzumab govitecan (IMMU-132) in patients with previously treated metastatic urothelial cancer (mUC): results from a phase I/II study. J Clin Oncol. 2019;37:354–354.CrossRef Tagawa ST, Faltas BM, Lam ET, et al. Sacituzumab govitecan (IMMU-132) in patients with previously treated metastatic urothelial cancer (mUC): results from a phase I/II study. J Clin Oncol. 2019;37:354–354.CrossRef
55.
go back to reference Loriot Y, Balar AV, Petrylak DP, et al. LBA24 TROPHY-U-01 cohort 1 final results: a phase II study of sacituzumab govitecan (SG) in metastatic urothelial cancer (mUC) that has progressed after platinum (PLT) and checkpoint inhibitors (CPI). Ann Oncol. 2020;31:S1156.CrossRef Loriot Y, Balar AV, Petrylak DP, et al. LBA24 TROPHY-U-01 cohort 1 final results: a phase II study of sacituzumab govitecan (SG) in metastatic urothelial cancer (mUC) that has progressed after platinum (PLT) and checkpoint inhibitors (CPI). Ann Oncol. 2020;31:S1156.CrossRef
56.
go back to reference Santin A, Komiya T, Goldenberg DM, Sharkey RM, Hong Q, Wegener WA, Goswami T, Bardia A. Sacituzumab govitecan (SG) in patients (pts) with previously treated metastatic endometrial cancer (mEC): results from a phase I/II study. J Clin Oncol. 2020;38:6081–6081.CrossRef Santin A, Komiya T, Goldenberg DM, Sharkey RM, Hong Q, Wegener WA, Goswami T, Bardia A. Sacituzumab govitecan (SG) in patients (pts) with previously treated metastatic endometrial cancer (mEC): results from a phase I/II study. J Clin Oncol. 2020;38:6081–6081.CrossRef
57.
go back to reference Heist RS, Guarino MJ, Masters G, et al. Therapy of advanced non-small-cell lung cancer with an SN-38-Anti-Trop-2 drug conjugate, sacituzumab govitecan. J Clin Oncol. 2017;35:2790–7.PubMedCrossRef Heist RS, Guarino MJ, Masters G, et al. Therapy of advanced non-small-cell lung cancer with an SN-38-Anti-Trop-2 drug conjugate, sacituzumab govitecan. J Clin Oncol. 2017;35:2790–7.PubMedCrossRef
58.
go back to reference Taylor KM, Morgan HE, Smart K, Zahari NM, Pumford S, Ellis IO, Robertson JFR, Nicholson RI. The emerging role of the LIV-1 subfamily of zinc transporters in breast cancer. Mol Med. 2007;13:396–406.PubMedPubMedCentralCrossRef Taylor KM, Morgan HE, Smart K, Zahari NM, Pumford S, Ellis IO, Robertson JFR, Nicholson RI. The emerging role of the LIV-1 subfamily of zinc transporters in breast cancer. Mol Med. 2007;13:396–406.PubMedPubMedCentralCrossRef
59.
go back to reference Manning DL, Robertson JFR, Ellis IO, et al. Oestrogen-regulated genes in breast cancer: Association of pLIV1 with lymph node involvement. Eur J Cancer. 1994;30:675–8.CrossRef Manning DL, Robertson JFR, Ellis IO, et al. Oestrogen-regulated genes in breast cancer: Association of pLIV1 with lymph node involvement. Eur J Cancer. 1994;30:675–8.CrossRef
60.
go back to reference Manning DL, Daly RJ, Lord PG, Kelly KF, Green CD. Effects of oestrogen on the expression of a 4.4 kb mRNA in the ZR-75-1 human breast cancer cell line. Mol Cell Endocrinol. 1988;59:205–12.PubMedCrossRef Manning DL, Daly RJ, Lord PG, Kelly KF, Green CD. Effects of oestrogen on the expression of a 4.4 kb mRNA in the ZR-75-1 human breast cancer cell line. Mol Cell Endocrinol. 1988;59:205–12.PubMedCrossRef
61.
go back to reference Sussman D, Smith LM, Anderson ME, et al. SGN-LIV1A: a novel antibody-drug conjugate targeting LIV-1 for the treatment of metastatic breast cancer. Mol Cancer Ther. 2014;13:2991–3000.PubMedCrossRef Sussman D, Smith LM, Anderson ME, et al. SGN-LIV1A: a novel antibody-drug conjugate targeting LIV-1 for the treatment of metastatic breast cancer. Mol Cancer Ther. 2014;13:2991–3000.PubMedCrossRef
62.
go back to reference Modi S, Pusztai L, Forero A, et al. Abstract PD3–14: phase 1 study of the antibody-drug conjugate SGN-LIV1A in patients with heavily pretreated triple-negative metastatic breast cancer. Cancer Res. 2018;78:PD3-14-PD3-14.CrossRef Modi S, Pusztai L, Forero A, et al. Abstract PD3–14: phase 1 study of the antibody-drug conjugate SGN-LIV1A in patients with heavily pretreated triple-negative metastatic breast cancer. Cancer Res. 2018;78:PD3-14-PD3-14.CrossRef
63.
go back to reference Cao AT, Higgins S, Stevens N, Gardai SJ, Sussman D. Abstract 2742: additional mechanisms of action of ladiratuzumab vedotin contribute to increased immune cell activation within the tumor. Cancer Res. 2018;78:2742–2742.CrossRef Cao AT, Higgins S, Stevens N, Gardai SJ, Sussman D. Abstract 2742: additional mechanisms of action of ladiratuzumab vedotin contribute to increased immune cell activation within the tumor. Cancer Res. 2018;78:2742–2742.CrossRef
64.
go back to reference Han HS, Alemany CA, Brown-Glaberman UA, et al. SGNLVA-002: single-arm, open label phase Ib/II study of ladiratuzumab vedotin (LV) in combination with pembrolizumab for first-line treatment of patients with unresectable locally advanced or metastatic triple-negative breast cancer. J Clin Oncol. 2019;37:TPS1110–TPS1110.CrossRef Han HS, Alemany CA, Brown-Glaberman UA, et al. SGNLVA-002: single-arm, open label phase Ib/II study of ladiratuzumab vedotin (LV) in combination with pembrolizumab for first-line treatment of patients with unresectable locally advanced or metastatic triple-negative breast cancer. J Clin Oncol. 2019;37:TPS1110–TPS1110.CrossRef
66.
go back to reference Tchoupa AK, Schuhmacher T, Hauck CR. Signaling by epithelial members of the CEACAM family—mucosal docking sites for pathogenic bacteria. Cell Commun Signal. 2014;12:27.PubMedPubMedCentralCrossRef Tchoupa AK, Schuhmacher T, Hauck CR. Signaling by epithelial members of the CEACAM family—mucosal docking sites for pathogenic bacteria. Cell Commun Signal. 2014;12:27.PubMedPubMedCentralCrossRef
67.
go back to reference Beauchemin N, Arabzadeh A. Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression and metastasis. Cancer Metastasis Rev. 2013;32:643–71.PubMedCrossRef Beauchemin N, Arabzadeh A. Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression and metastasis. Cancer Metastasis Rev. 2013;32:643–71.PubMedCrossRef
68.
go back to reference Decary S, Berne P-F, Nicolazzi C, et al. Abstract 1688: a novel anti-CEACAM5 maytansinoid-antibody-drug conjugate for the treatment of colorectal, lung and gastric tumors. Cancer Res. 2015;75:1688–1688.CrossRef Decary S, Berne P-F, Nicolazzi C, et al. Abstract 1688: a novel anti-CEACAM5 maytansinoid-antibody-drug conjugate for the treatment of colorectal, lung and gastric tumors. Cancer Res. 2015;75:1688–1688.CrossRef
69.
go back to reference Gazzah A, Stjepanovic N, Ryu MH, et al. First-in-human phase I trial of the anti-CEACAM5 antibody-drug conjugate SAR408701 in patients with advanced solid tumors (NCT02187848). Eur J Cancer. 2016;69:S14–5.CrossRef Gazzah A, Stjepanovic N, Ryu MH, et al. First-in-human phase I trial of the anti-CEACAM5 antibody-drug conjugate SAR408701 in patients with advanced solid tumors (NCT02187848). Eur J Cancer. 2016;69:S14–5.CrossRef
70.
go back to reference Gazzah A, Ricordel C, Cousin S, et al. Efficacy and safety of the antibody-drug conjugate (ADC) SAR408701 in patients (pts) with non-squamous non-small cell lung cancer (NSQ NSCLC) expressing carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5). J Clin Oncol. 2020;38:9505–9505.CrossRef Gazzah A, Ricordel C, Cousin S, et al. Efficacy and safety of the antibody-drug conjugate (ADC) SAR408701 in patients (pts) with non-squamous non-small cell lung cancer (NSQ NSCLC) expressing carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5). J Clin Oncol. 2020;38:9505–9505.CrossRef
71.
go back to reference Segal NH, Dotan E, Berlin JD, et al. Abstract CT211: IMMU-130, an SN-38 antibody-drug conjugate (ADC) targeting CEACAM5, is therapeutically active in metastatic colorectal cancer (mCRC): Initial clinical results of two phase I studies. Cancer Res. 2014;74:CT211–CT211.CrossRef Segal NH, Dotan E, Berlin JD, et al. Abstract CT211: IMMU-130, an SN-38 antibody-drug conjugate (ADC) targeting CEACAM5, is therapeutically active in metastatic colorectal cancer (mCRC): Initial clinical results of two phase I studies. Cancer Res. 2014;74:CT211–CT211.CrossRef
72.
go back to reference Dotan E, Cohen SJ, Starodub AN, et al. Phase I/II trial of labetuzumab govitecan (Anti-CEACAM5/SN-38 antibody-drug conjugate) in patients with refractory or relapsing metastatic colorectal cancer. J Clin Oncol. 2017;35:3338–46.CrossRefPubMedPubMedCentral Dotan E, Cohen SJ, Starodub AN, et al. Phase I/II trial of labetuzumab govitecan (Anti-CEACAM5/SN-38 antibody-drug conjugate) in patients with refractory or relapsing metastatic colorectal cancer. J Clin Oncol. 2017;35:3338–46.CrossRefPubMedPubMedCentral
73.
go back to reference Schulze WX, Deng L, Mann M. Phosphotyrosine interactome of the ErbB-receptor kinase family. Mol Syst Biol. 2005;1(2005):0008.PubMed Schulze WX, Deng L, Mann M. Phosphotyrosine interactome of the ErbB-receptor kinase family. Mol Syst Biol. 2005;1(2005):0008.PubMed
75.
go back to reference Mishra R, Patel H, Alanazi S, Yuan L, Garrett JT. HER3 signaling and targeted therapy in cancer. Oncol Rev. 2018;12:355.PubMedPubMedCentral Mishra R, Patel H, Alanazi S, Yuan L, Garrett JT. HER3 signaling and targeted therapy in cancer. Oncol Rev. 2018;12:355.PubMedPubMedCentral
76.
go back to reference Kiavue N, Cabel L, Melaabi S, Bataillon G, Callens C, Lerebours F, Pierga J-Y, Bidard F-C. ERBB3 mutations in cancer: biological aspects, prevalence and therapeutics. Oncogene. 2020;39:487–502.PubMedCrossRef Kiavue N, Cabel L, Melaabi S, Bataillon G, Callens C, Lerebours F, Pierga J-Y, Bidard F-C. ERBB3 mutations in cancer: biological aspects, prevalence and therapeutics. Oncogene. 2020;39:487–502.PubMedCrossRef
77.
go back to reference Yonemori K, Masuda N, Takahashi S, Kogawa T, Nakayama T, Yamamoto Y, Takahashi M, Toyama T, Saeki T, Iwata H. Single agent activity of U3-1402, a HER3-targeting antibody-drug conjugate, in HER3-overexpressing metastatic breast cancer: updated results from a phase I/II trial. Ann Oncol. 2019;30:iii48.CrossRef Yonemori K, Masuda N, Takahashi S, Kogawa T, Nakayama T, Yamamoto Y, Takahashi M, Toyama T, Saeki T, Iwata H. Single agent activity of U3-1402, a HER3-targeting antibody-drug conjugate, in HER3-overexpressing metastatic breast cancer: updated results from a phase I/II trial. Ann Oncol. 2019;30:iii48.CrossRef
78.
go back to reference Scharpenseel H, Hanssen A, Loges S, et al. EGFR and HER3 expression in circulating tumor cells and tumor tissue from non-small cell lung cancer patients. Sci Rep. 2019;9:7406.PubMedPubMedCentralCrossRef Scharpenseel H, Hanssen A, Loges S, et al. EGFR and HER3 expression in circulating tumor cells and tumor tissue from non-small cell lung cancer patients. Sci Rep. 2019;9:7406.PubMedPubMedCentralCrossRef
79.
go back to reference Hashimoto Y, Koyama K, Kamai Y, et al. A novel HER3-Targeting antibody-drug conjugate, U3-1402, exhibits potent therapeutic efficacy through the delivery of cytotoxic payload by efficient internalization. Clin Cancer Res. 2019;25:7151 LP – 7161.CrossRef Hashimoto Y, Koyama K, Kamai Y, et al. A novel HER3-Targeting antibody-drug conjugate, U3-1402, exhibits potent therapeutic efficacy through the delivery of cytotoxic payload by efficient internalization. Clin Cancer Res. 2019;25:7151 LP – 7161.CrossRef
80.
go back to reference Yu HA, Baik CS, Gold K, et al. Efficacy and safety of patritumab deruxtecan (U3–1402), a novel HER3 directed antibody drug conjugate, in patients (pts) with EGFR-mutated (EGFRm) NSCLC. Ann Oncol. 2020;31:S1189–90.CrossRef Yu HA, Baik CS, Gold K, et al. Efficacy and safety of patritumab deruxtecan (U3–1402), a novel HER3 directed antibody drug conjugate, in patients (pts) with EGFR-mutated (EGFRm) NSCLC. Ann Oncol. 2020;31:S1189–90.CrossRef
81.
go back to reference Koganemaru S, Kuboki Y, Koga Y, et al. U3-1402, a novel HER3-targeting antibody–drug conjugate, for the treatment of colorectal cancer. Mol Cancer Ther. 2019;18:2043–50.PubMedCrossRef Koganemaru S, Kuboki Y, Koga Y, et al. U3-1402, a novel HER3-targeting antibody–drug conjugate, for the treatment of colorectal cancer. Mol Cancer Ther. 2019;18:2043–50.PubMedCrossRef
82.
go back to reference Rump A, Morikawa Y, Tanaka M, Minami S, Umesaki N, Takeuchi M, Miyajima A. Binding of ovarian cancer antigen CA125/MUC61 to mesothelin mediates cell adhesion. J Biol Chem. 2004;279:9190–8.PubMedCrossRef Rump A, Morikawa Y, Tanaka M, Minami S, Umesaki N, Takeuchi M, Miyajima A. Binding of ovarian cancer antigen CA125/MUC61 to mesothelin mediates cell adhesion. J Biol Chem. 2004;279:9190–8.PubMedCrossRef
83.
84.
go back to reference Golfier S, Kopitz C, Kahnert A, et al. Anetumab ravtansine: a novel mesothelin-targeting antibody-drug conjugate cures tumors with heterogeneous target expression favored by bystander effect. Mol Cancer Ther. 2014;13:1537–48.PubMedCrossRef Golfier S, Kopitz C, Kahnert A, et al. Anetumab ravtansine: a novel mesothelin-targeting antibody-drug conjugate cures tumors with heterogeneous target expression favored by bystander effect. Mol Cancer Ther. 2014;13:1537–48.PubMedCrossRef
85.
go back to reference Hassan R, Blumenschein GR, Moore KN, et al. First-in-human, multicenter, phase I dose-escalation and expansion study of anti-mesothelin antibody-drug conjugate anetumab ravtansine in advanced or metastatic solid tumors. J Clin Oncol. 2020;38:1824–35.PubMedCrossRefPubMedCentral Hassan R, Blumenschein GR, Moore KN, et al. First-in-human, multicenter, phase I dose-escalation and expansion study of anti-mesothelin antibody-drug conjugate anetumab ravtansine in advanced or metastatic solid tumors. J Clin Oncol. 2020;38:1824–35.PubMedCrossRefPubMedCentral
86.
go back to reference Kindler HL, Novello S, Fennell D, et al. OA 02.01 randomized phase II study of anetumab ravtansine or vinorelbine in patients with metastatic pleural mesothelioma. J Thorac Oncol. 2017;12:S1746.CrossRef Kindler HL, Novello S, Fennell D, et al. OA 02.01 randomized phase II study of anetumab ravtansine or vinorelbine in patients with metastatic pleural mesothelioma. J Thorac Oncol. 2017;12:S1746.CrossRef
87.
go back to reference Clarke J, Chu S-C, Siu LL, et al. Abstract B057: BMS-986148, an anti-mesothelin antibody-drug conjugate (ADC), alone or in combination with nivolumab demonstrates clinical activity in patients with select advanced solid tumors. Mol Cancer Ther. 2019;18:B057–B057.CrossRef Clarke J, Chu S-C, Siu LL, et al. Abstract B057: BMS-986148, an anti-mesothelin antibody-drug conjugate (ADC), alone or in combination with nivolumab demonstrates clinical activity in patients with select advanced solid tumors. Mol Cancer Ther. 2019;18:B057–B057.CrossRef
88.
go back to reference Zhang Y, Xia M, Jin K, et al. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol Cancer. 2018;17:1–14.PubMedPubMedCentralCrossRef Zhang Y, Xia M, Jin K, et al. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol Cancer. 2018;17:1–14.PubMedPubMedCentralCrossRef
90.
go back to reference Strickler JH, Weekes CD, Nemunaitis J, et al. First-in-human phase I, dose-escalation and -expansion study of telisotuzumab vedotin, an antibody–drug conjugate targeting c-Met, in patients with advanced solid tumors. J Clin Oncol. 2018;36:3298–306.PubMedCrossRef Strickler JH, Weekes CD, Nemunaitis J, et al. First-in-human phase I, dose-escalation and -expansion study of telisotuzumab vedotin, an antibody–drug conjugate targeting c-Met, in patients with advanced solid tumors. J Clin Oncol. 2018;36:3298–306.PubMedCrossRef
91.
go back to reference Camidge DR, Barlesi F, Goldman JW, et al. Results of the phase 1b study of ABBV-399 (telisotuzumab vedotin; teliso-v) in combination with erlotinib in patients with c-Met+ non-small cell lung cancer by EGFR mutation status. J Clin Oncol. 2019;37:3011–3011.CrossRef Camidge DR, Barlesi F, Goldman JW, et al. Results of the phase 1b study of ABBV-399 (telisotuzumab vedotin; teliso-v) in combination with erlotinib in patients with c-Met+ non-small cell lung cancer by EGFR mutation status. J Clin Oncol. 2019;37:3011–3011.CrossRef
92.
go back to reference Camidge DR, Goldman JW, Cole GW, Sun Z, Ocampo CJ, Komarnitsky P, Blot V. Evaluating telisotuzumab vedotin in combination with osimertinib in patients with advanced non-small cell lung cancer: a phase I/Ib study cohort. Ann Oncol. 2020;31:S894.CrossRef Camidge DR, Goldman JW, Cole GW, Sun Z, Ocampo CJ, Komarnitsky P, Blot V. Evaluating telisotuzumab vedotin in combination with osimertinib in patients with advanced non-small cell lung cancer: a phase I/Ib study cohort. Ann Oncol. 2020;31:S894.CrossRef
94.
go back to reference Gymnopoulos M, Betancourt O, Blot V, et al. TR1801-ADC: a highly potent cMet antibody–drug conjugate with high activity in patient-derived xenograft models of solid tumors. Mol Oncol. 2020;14:54–68.PubMedCrossRef Gymnopoulos M, Betancourt O, Blot V, et al. TR1801-ADC: a highly potent cMet antibody–drug conjugate with high activity in patient-derived xenograft models of solid tumors. Mol Oncol. 2020;14:54–68.PubMedCrossRef
95.
go back to reference Yang C, Wang L, Sun X, Tang M, Quan H, Zhang L, Lou L, Gou S. SHR-A1403, a novel c-Met antibody-drug conjugate, exerts encouraging anti-tumor activity in c-Met-overexpressing models. Acta Pharmacol Sin. 2019;40:971–9.PubMedPubMedCentralCrossRef Yang C, Wang L, Sun X, Tang M, Quan H, Zhang L, Lou L, Gou S. SHR-A1403, a novel c-Met antibody-drug conjugate, exerts encouraging anti-tumor activity in c-Met-overexpressing models. Acta Pharmacol Sin. 2019;40:971–9.PubMedPubMedCentralCrossRef
96.
go back to reference Luhrs CA, Slomiany BL. A human membrane-associated folate binding protein is anchored by a glycosyl-phosphatidylinositol tail. J Biol Chem. 1989;264:21446–9.PubMedCrossRef Luhrs CA, Slomiany BL. A human membrane-associated folate binding protein is anchored by a glycosyl-phosphatidylinositol tail. J Biol Chem. 1989;264:21446–9.PubMedCrossRef
97.
go back to reference Parker N, Turk MJ, Westrick E, Lewis JD, Low PS, Leamon CP. Folate receptor expression in carcinomas and normal tissues determined by a quantitative radioligand binding assay. Anal Biochem. 2005;338:284–93.PubMedCrossRef Parker N, Turk MJ, Westrick E, Lewis JD, Low PS, Leamon CP. Folate receptor expression in carcinomas and normal tissues determined by a quantitative radioligand binding assay. Anal Biochem. 2005;338:284–93.PubMedCrossRef
98.
go back to reference Scaranti M, Cojocaru E, Banerjee S, Banerji U. Exploiting the folate receptor α in oncology. Nat Rev Clin Oncol. 2020;17:349–59.PubMedCrossRef Scaranti M, Cojocaru E, Banerjee S, Banerji U. Exploiting the folate receptor α in oncology. Nat Rev Clin Oncol. 2020;17:349–59.PubMedCrossRef
99.
go back to reference Moore KN, Borghaei H, O’Malley DM, et al. Phase 1 dose-escalation study of mirvetuximab soravtansine (IMGN853), a folate receptor α-targeting antibody-drug conjugate, in patients with solid tumors. Cancer. 2017;123:3080–7.PubMedCrossRef Moore KN, Borghaei H, O’Malley DM, et al. Phase 1 dose-escalation study of mirvetuximab soravtansine (IMGN853), a folate receptor α-targeting antibody-drug conjugate, in patients with solid tumors. Cancer. 2017;123:3080–7.PubMedCrossRef
100.
go back to reference Martin L, Konner J, O’Malley D, Bauer T, Matulonis U, Seward S, Oza A, Kirby M, Birrer M, Moore K. Phase I expansion study of mirvetuximab soravtansine, a folate receptor alpha (FRA)-targeting antibody-drug conjugate (ADC), in patients with endometrial cancer. Int J Gynecol Cancer. 2017;27:1975. Martin L, Konner J, O’Malley D, Bauer T, Matulonis U, Seward S, Oza A, Kirby M, Birrer M, Moore K. Phase I expansion study of mirvetuximab soravtansine, a folate receptor alpha (FRA)-targeting antibody-drug conjugate (ADC), in patients with endometrial cancer. Int J Gynecol Cancer. 2017;27:1975.
101.
go back to reference Moore KN, Martin LP, O’Malley DM, Matulonis UA, Konner JA, Perez RP, Bauer TM, Ruiz-Soto R, Birrer MJ. Safety and activity of mirvetuximab soravtansine (IMGN853), a folate receptor alpha-targeting antibody-drug conjugate, in platinum-resistant ovarian, fallopian tube, or primary peritoneal cancer: a phase I expansion study. J Clin Oncol. 2017;35:1112–8.PubMedCrossRef Moore KN, Martin LP, O’Malley DM, Matulonis UA, Konner JA, Perez RP, Bauer TM, Ruiz-Soto R, Birrer MJ. Safety and activity of mirvetuximab soravtansine (IMGN853), a folate receptor alpha-targeting antibody-drug conjugate, in platinum-resistant ovarian, fallopian tube, or primary peritoneal cancer: a phase I expansion study. J Clin Oncol. 2017;35:1112–8.PubMedCrossRef
102.
go back to reference Moore K, Oza A, Colombo N, et al. FORWARD I (GOG 3011): a phase III study of mirvetuximab soravtansine, a folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC), versus chemotherapy in patients (pts) with platinumresistant ovarian cancer (PROC). Ann Oncol. 2019;30:v403.CrossRef Moore K, Oza A, Colombo N, et al. FORWARD I (GOG 3011): a phase III study of mirvetuximab soravtansine, a folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC), versus chemotherapy in patients (pts) with platinumresistant ovarian cancer (PROC). Ann Oncol. 2019;30:v403.CrossRef
104.
go back to reference O’Malley DM, Moore KN, Vergote I, Martin LP, Gilbert L, Martin AG, Nepert DL, Ruiz-Soto R, Birrer MJ, Matulonis UA. Safety findings from FORWARD II: a phase 1b study evaluating the folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC) mirvetuximab soravtansine (IMGN853) in combination with bevacizumab, carboplatin, pegylated liposomal doxorubicin (PLD) or pembrolizumab in patients (pts) with ovarian cancer. J Clin Oncol. 2017;35:5553.CrossRef O’Malley DM, Moore KN, Vergote I, Martin LP, Gilbert L, Martin AG, Nepert DL, Ruiz-Soto R, Birrer MJ, Matulonis UA. Safety findings from FORWARD II: a phase 1b study evaluating the folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC) mirvetuximab soravtansine (IMGN853) in combination with bevacizumab, carboplatin, pegylated liposomal doxorubicin (PLD) or pembrolizumab in patients (pts) with ovarian cancer. J Clin Oncol. 2017;35:5553.CrossRef
106.
go back to reference Versteeg HH, Peppelenbosch MP, Spek CA. The pleiotropic effects of tissue factor: a possible role for factor VIIa-induced intracellular signalling? Thromb Haemost. 2001;86:1353–9.PubMedCrossRef Versteeg HH, Peppelenbosch MP, Spek CA. The pleiotropic effects of tissue factor: a possible role for factor VIIa-induced intracellular signalling? Thromb Haemost. 2001;86:1353–9.PubMedCrossRef
107.
go back to reference Van Den Berg YW, Osanto S, Reitsma PH, Versteeg HH. The relationship between tissue factor and cancer progression: insights from bench and bedside. Blood. 2012;119:924–32.PubMedCrossRef Van Den Berg YW, Osanto S, Reitsma PH, Versteeg HH. The relationship between tissue factor and cancer progression: insights from bench and bedside. Blood. 2012;119:924–32.PubMedCrossRef
108.
go back to reference Breij ECW, De Goeij BECG, Verploegen S, et al. An antibody-drug conjugate that targets tissue factor exhibits potent therapeutic activity against a broad range of solid tumors. Cancer Res. 2014;74:1214–26.PubMedCrossRef Breij ECW, De Goeij BECG, Verploegen S, et al. An antibody-drug conjugate that targets tissue factor exhibits potent therapeutic activity against a broad range of solid tumors. Cancer Res. 2014;74:1214–26.PubMedCrossRef
109.
go back to reference de Bono JS, Concin N, Hong DS, et al. Tisotumab vedotin in patients with advanced or metastatic solid tumours (InnovaTV 201): a first-in-human, multicentre, phase 1–2 trial. Lancet Oncol. 2019;20:383–93.PubMedCrossRef de Bono JS, Concin N, Hong DS, et al. Tisotumab vedotin in patients with advanced or metastatic solid tumours (InnovaTV 201): a first-in-human, multicentre, phase 1–2 trial. Lancet Oncol. 2019;20:383–93.PubMedCrossRef
111.
go back to reference Coleman RL, Lorusso D, Gennigens C, et al. LBA32 Tisotumab vedotin in previously treated recurrent or metastatic cervical cancer: results from the phase II innovaTV 204/GOG-3023/ENGOT-cx6 study. Ann Oncol. 2020;31:S1162–3.CrossRef Coleman RL, Lorusso D, Gennigens C, et al. LBA32 Tisotumab vedotin in previously treated recurrent or metastatic cervical cancer: results from the phase II innovaTV 204/GOG-3023/ENGOT-cx6 study. Ann Oncol. 2020;31:S1162–3.CrossRef
112.
go back to reference Vergote I, Dean E, Lassen U, et al. A phase IIa study of tisotumab vedotin (HuMax® -TF-ADC) in patients with relapsed, recurrent and/or metastatic cervical cancer. Ann Oncol. 2017;28:v330–1.CrossRef Vergote I, Dean E, Lassen U, et al. A phase IIa study of tisotumab vedotin (HuMax® -TF-ADC) in patients with relapsed, recurrent and/or metastatic cervical cancer. Ann Oncol. 2017;28:v330–1.CrossRef
113.
go back to reference Boni V, Burris HA III, Liu JF, et al. CX-2009, a CD166-directed probody drug conjugate (PDC): results from the first-in-human study in patients (Pts) with advanced cancer including breast cancer (BC). J Clin Oncol. 2020;38:526–526.CrossRef Boni V, Burris HA III, Liu JF, et al. CX-2009, a CD166-directed probody drug conjugate (PDC): results from the first-in-human study in patients (Pts) with advanced cancer including breast cancer (BC). J Clin Oncol. 2020;38:526–526.CrossRef
114.
go back to reference Johnson ML, El-Khoueiry AB, Hafez N, et al. CX-2029, a PROBODY drug conjugate targeting CD71 (transferrin receptor): results from a first-in-human study (PROCLAIM-CX-2029) in patients (Pts) with advanced cancer. J Clin Oncol. 2020;38:3502–3502.CrossRef Johnson ML, El-Khoueiry AB, Hafez N, et al. CX-2029, a PROBODY drug conjugate targeting CD71 (transferrin receptor): results from a first-in-human study (PROCLAIM-CX-2029) in patients (Pts) with advanced cancer. J Clin Oncol. 2020;38:3502–3502.CrossRef
115.
go back to reference Goldberg SD, Cardoso RMF, Lin T, Spinka-Doms T, Klein D, Jacobs SA, Dudkin V, Gilliland G, O’Neil KT. Engineering a targeted delivery platform using Centyrins. Protein Eng Des Sel. 2016;29:563–72.PubMed Goldberg SD, Cardoso RMF, Lin T, Spinka-Doms T, Klein D, Jacobs SA, Dudkin V, Gilliland G, O’Neil KT. Engineering a targeted delivery platform using Centyrins. Protein Eng Des Sel. 2016;29:563–72.PubMed
116.
go back to reference Li JY, Perry SR, Muniz-Medina V, et al. A Biparatopic HER2-targeting antibody-drug conjugate induces tumor regression in primary models refractory to or ineligible for HER2-targeted therapy. Cancer Cell. 2016;29:117–29.PubMedCrossRef Li JY, Perry SR, Muniz-Medina V, et al. A Biparatopic HER2-targeting antibody-drug conjugate induces tumor regression in primary models refractory to or ineligible for HER2-targeted therapy. Cancer Cell. 2016;29:117–29.PubMedCrossRef
117.
go back to reference Sellmann C, Doerner A, Knuehl C, et al. Balancing selectivity and efficacy of bispecific Epidermal Growth Factor Receptor (EGFR) x c-MET antibodies and antibody-drug conjugates. J Biol Chem. 2016;291:25106–19.PubMedPubMedCentralCrossRef Sellmann C, Doerner A, Knuehl C, et al. Balancing selectivity and efficacy of bispecific Epidermal Growth Factor Receptor (EGFR) x c-MET antibodies and antibody-drug conjugates. J Biol Chem. 2016;291:25106–19.PubMedPubMedCentralCrossRef
119.
go back to reference Matulonis UA, Moore KN, Martin LP, Vergote IB, Castro C, Gilbert L, Malek K, Birrer MJ, O’Malley DM. Mirvetuximab soravtansine, a folate receptor alpha (FRa)-targeting antibody-drug conjugate (ADC), with pembrolizumab in platinumresistant ovarian cancer (PROC): initial results of an expansion cohort from FORWARD II, a phase Ib study. Ann Oncol. 2018;29:viii339.CrossRef Matulonis UA, Moore KN, Martin LP, Vergote IB, Castro C, Gilbert L, Malek K, Birrer MJ, O’Malley DM. Mirvetuximab soravtansine, a folate receptor alpha (FRa)-targeting antibody-drug conjugate (ADC), with pembrolizumab in platinumresistant ovarian cancer (PROC): initial results of an expansion cohort from FORWARD II, a phase Ib study. Ann Oncol. 2018;29:viii339.CrossRef
Metadata
Title
Antibody–drug conjugates in solid tumors: a look into novel targets
Authors
Carmen Criscitiello
Stefania Morganti
Giuseppe Curigliano
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2021
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-021-01035-z

Other articles of this Issue 1/2021

Journal of Hematology & Oncology 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine