51
|
Carey LA, Loirat D, Punie K, Bardia A, Diéras V, Dalenc F, Diamond JR, Fontaine C, Wang G, Rugo HS, Hurvitz SA, Kalinsky K, O'Shaughnessy J, Loibl S, Gianni L, Piccart M, Zhu Y, Delaney R, Phan S, Cortés J. Sacituzumab govitecan as second-line treatment for metastatic triple-negative breast cancer-phase 3 ASCENT study subanalysis. NPJ Breast Cancer 2022; 8:72. [PMID: 35680967 PMCID: PMC9184615 DOI: 10.1038/s41523-022-00439-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 05/04/2022] [Indexed: 11/08/2022] Open
Abstract
Patients with triple-negative breast cancer (TNBC) who relapse early after (neo)adjuvant chemotherapy have more aggressive disease. In the ASCENT trial, sacituzumab govitecan (SG), an antibody-drug conjugate composed of an anti-Trop-2 antibody coupled to SN-38 via a hydrolyzable linker, improved outcomes over single-agent chemotherapy of physician's choice (TPC) in metastatic TNBC (mTNBC). Of 468 patients without known baseline brain metastases, 33/235 vs 32/233 patients (both 14%) in the SG vs TPC arms, respectively, received one line of therapy in the metastatic setting and experienced disease recurrence ≤12 months after (neo)adjuvant chemotherapy. SG prolonged progression-free survival (median 5.7 vs 1.5 months [HR, 0.41; 95% CI, 0.22-0.76]) and overall survival (median 10.9 vs 4.9 months [HR, 0.51; 95% CI, 0.28-0.91]) vs TPC, with a manageable safety profile in this subgroup consistent with the overall population. In this second-line setting, as with later-line therapy, SG improved survival over conventional chemotherapy for patients with mTNBC.
Collapse
Affiliation(s)
- Lisa A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA.
| | - Delphine Loirat
- Medical Oncology Department and D3i, Institut Curie, Paris, France
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Aditya Bardia
- Department of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Véronique Diéras
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | | | - Jennifer R Diamond
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christel Fontaine
- Medical Oncology Department, Oncologisch Centrum, UZ Brussel, Brussels, Belgium
| | | | - Hope S Rugo
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Sara A Hurvitz
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Kevin Kalinsky
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology, US Oncology, Dallas, TX, USA
| | - Sibylle Loibl
- Department of Medicine and Research, Hämatologisch-Onkologische Gemeinschaftspraxis am Bethanien-Krankenhaus, Frankfurt, Germany
| | - Luca Gianni
- Medical Oncology, Gianni Bonadonna Foundation, Milan, Italy
| | - Martine Piccart
- Medical Oncology Department, Institut Jules Bordet and l'Université Libre de Bruxelles, Brussels, Belgium
| | - Yanni Zhu
- Department of Biostatistics, Gilead Sciences, Inc, Foster City, CA, USA
| | - Rosemary Delaney
- Department of Clinical Research, Gilead Sciences, Inc, Morris Plains, NJ, USA
| | - See Phan
- Department of Clinical Development, Gilead Sciences Inc, Foster City, CA, USA
| | - Javier Cortés
- International Breast Cancer Center, Quirón Group, Barcelona, Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
52
|
Fan Y, Li Q, Shen Q, Liu Z, Zhang Z, Hu S, Yu W, He Z, He Q, Zhang Q. Head-to-Head Comparison of the Expression Differences of NECTIN-4, TROP-2, and HER2 in Urothelial Carcinoma and Its Histologic Variants. Front Oncol 2022; 12:858865. [PMID: 35515131 PMCID: PMC9063095 DOI: 10.3389/fonc.2022.858865] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/21/2022] [Indexed: 11/15/2022] Open
Abstract
Background Antibody–drug conjugates (ADC), such as enfortumab vedotin (EV), sacituzumab govitecan (SG), and RC-48, have shown outstanding response rates to local advanced or metastatic urothelial carcinoma (UC). However, their corresponding target expression characteristics in UC and its histologic variants were unknown. Methods We detected the expression of NECTIN-4, TROP-2, and HER2, which are the corresponding targets of ADCs EV, SG, and RC-48 in muscle-invasive UC through immunohistochemistry. Results 161 consecutive samples from 2017 to 2021 of muscle-invasive UC and its histologic variants were obtained in Peking University First Hospital. Variant histology types included 72UC, 10 squamous carcinomas, 23 glandular carcinomas, 19 small cell carcinomas, 19 micropapillary variants, and 18 nested variants. NECTIN-4 expression was found to be 57/72 (79.2%), 10/10 (100%), 15/23 (65.2%), 4/19 (21.1%), 15/19 (78.9%), and 16/18 (88.9%) in conventional UC, squamous carcinoma, glandular carcinoma, small cell carcinoma, micropapillary, and nested variant, respectively, compared with 65/72 (90.3%), 8/10 (80.0%), 13/23 (56.5%), 3/19 (15.8%), 16/19 (84.2%), and 15/18 (83.3%) of TROP-2, and 26/72 (36.1%), 0, 5/23 (21.7%), 6/19 (31.6%), 5/19 (26.3%), and 7/18 (38.9%) of HER2.
Collapse
Affiliation(s)
- Yu Fan
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Qinhan Li
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Qi Shen
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Zhifu Liu
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Zhenan Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Shuai Hu
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Wei Yu
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Zhisong He
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Qun He
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Qian Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| |
Collapse
|
53
|
Yang R, Li Y, Wang H, Qin T, Yin X, Ma X. Therapeutic progress and challenges for triple negative breast cancer: targeted therapy and immunotherapy. MOLECULAR BIOMEDICINE 2022; 3:8. [PMID: 35243562 PMCID: PMC8894518 DOI: 10.1186/s43556-022-00071-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/09/2022] [Indexed: 02/08/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer, with estrogen receptor, human epidermal growth factor receptor 2 and progesterone receptor negative. TNBC is characterized by high heterogeneity, high rates of metastasis, poor prognosis, and lack of therapeutic targets. Now the treatment of TNBC is still based on surgery and chemotherapy, which is effective only in initial stage but almost useless in advanced stage. And due to the lack of hormone target, hormonal therapies have little beneficial effects. In recent years, signaling pathways and receptor-specific targets have been reported to be effective in TNBC patients under specific clinical conditions. Now targeted therapies have been approved for many other cancers and even other subtypes of breast cancer, but treatment options for TNBC are still limited. Most of TNBC patients showed no response, which may be related to the heterogeneity of TNBC, therefore more effective treatments and predictive biomarkers are needed. In the present review, we summarize potential treatment opinions for TNBC based on the dysregulated receptors and signaling pathways, which play a significant role in multiple stages of TNBC development. We also focus on the application of immunotherapy in TNBC, and summarize the preclinical and clinical trials of therapy for patients with TNBC. We hope to accelerate the research and development of new drugs for TNBC by understanding the relevant mechanisms, and to improve survival.
Collapse
Affiliation(s)
- Ruoning Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China.,Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yueyi Li
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Hang Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Taolin Qin
- West China Hospital, West China Medical School Sichuan University, Chengdu, PR, China
| | - Xiaomeng Yin
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China.
| |
Collapse
|
54
|
Goldenberg DM, Sharkey RM, Govindan SV, Cardillo TM. Novel Peptide Camptothecin Drug-linkers for Potent ADCs-Letter. Mol Cancer Ther 2022; 21:237. [PMID: 34996865 DOI: 10.1158/1535-7163.mct-21-0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/22/2021] [Accepted: 04/07/2021] [Indexed: 11/16/2022]
Affiliation(s)
| | - Robert M Sharkey
- Center for Molecular Medicine and Immunology, Mendham, New Jersey.
| | - Serengulam V Govindan
- Immunomedics, Inc., a subsidiary of Gilead Sciences, Inc., Morris Plains, New Jersey
| | - Thomas M Cardillo
- Immunomedics, Inc., a subsidiary of Gilead Sciences, Inc., Morris Plains, New Jersey
| |
Collapse
|
55
|
Ceci C, Lacal PM, Graziani G. Antibody-drug conjugates: Resurgent anticancer agents with multi-targeted therapeutic potential. Pharmacol Ther 2022; 236:108106. [PMID: 34990642 DOI: 10.1016/j.pharmthera.2021.108106] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Antibody-drug conjugates (ADCs) constitute a relatively new group of anticancer agents, whose first appearance took place about two decades ago, but a renewed interest occurred in recent years, following the success of anti-cancer immunotherapy with monoclonal antibodies. Indeed, an ADC combines the selectivity of a monoclonal antibody with the cell killing properties of a chemotherapeutic agent (payload), joined together through an appropriate linker. The antibody moiety targets a specific cell surface antigen expressed by tumor cells and/or cells of the tumor microenvironment and acts as a carrier that delivers the cytotoxic payload within the tumor mass. Despite advantages in terms of selectivity and potency, the development of ADCs is not devoid of challenges, due to: i) low tumor selectivity when the target antigens are not exclusively expressed by cancer cells; ii) premature release of the cytotoxic drug into the bloodstream as a consequence of linker instability; iii) development of tumor resistance mechanisms to the payload. All these factors may result in lack of efficacy and/or in no safety improvement compared to unconjugated cytotoxic agents. Nevertheless, the development of antibodies engineered to remain inert until activated in the tumor (e.g., antibodies activated proteolytically after internalization or by the acidic conditions of the tumor microenvironment) together with the discovery of innovative targets and cytotoxic or immunomodulatory payloads, have allowed the design of next-generation ADCs that are expected to possess improved therapeutic properties. This review provides an overview of approved ADCs, with related advantages and limitations, and of novel targets exploited by ADCs that are presently under clinical investigation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy.
| |
Collapse
|
56
|
Zacharias N, Podust VN, Kajihara KK, Leipold D, Del Rosario G, Thayer D, Dong E, Paluch M, Fischer D, Zheng K, Lei C, He J, Ng C, Su D, Liu L, Masih S, Sawyer W, Tinianow J, Marik J, Yip V, Li G, Chuh J, Morisaki JH, Park S, Zheng B, Hernandez-Barry H, Loyet KM, Xu M, Kozak KR, Phillips GL, Shen BQ, Wu C, Xu K, Yu SF, Kamath A, Rowntree RK, Reilly D, Pillow T, Polson A, Schellenberger V, Hazenbos WLW, Sadowsky J. A homogeneous high-DAR antibody-drug conjugate platform combining THIOMAB antibodies and XTEN polypeptides. Chem Sci 2022; 13:3147-3160. [PMID: 35414872 PMCID: PMC8926172 DOI: 10.1039/d1sc05243h] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/27/2022] [Indexed: 11/21/2022] Open
Abstract
The antibody-drug conjugate (ADC) is a well-validated modality for the cell-specific delivery of small molecules with impact expanding rapidly beyond their originally-intended purpose of treating cancer. However, antibody-mediated delivery (AMD)...
Collapse
Affiliation(s)
| | - Vladimir N Podust
- Amunix Pharmaceuticals, Inc. 2 Tower Place South San Francisco CA 94080 USA
| | | | | | | | - Desiree Thayer
- Amunix Pharmaceuticals, Inc. 2 Tower Place South San Francisco CA 94080 USA
| | - Emily Dong
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Maciej Paluch
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - David Fischer
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Kai Zheng
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Corinna Lei
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Jintang He
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Carl Ng
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Dian Su
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Luna Liu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | - William Sawyer
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Jeff Tinianow
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Jan Marik
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Victor Yip
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Guangmin Li
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Josefa Chuh
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | - Summer Park
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Bing Zheng
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | - Kelly M Loyet
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Min Xu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | | | - Ben-Quan Shen
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Cong Wu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Keyang Xu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Shang-Fan Yu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Amrita Kamath
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | | | - Thomas Pillow
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Andrew Polson
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | | | - Jack Sadowsky
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| |
Collapse
|
57
|
O'Shaughnessy J, Brufsky A, Rugo HS, Tolaney SM, Punie K, Sardesai S, Hamilton E, Loirat D, Traina T, Leon-Ferre R, Hurvitz SA, Kalinsky K, Bardia A, Henry S, Mayer I, Zhu Y, Phan S, Cortés J. Analysis of patients without and with an initial triple-negative breast cancer diagnosis in the phase 3 randomized ASCENT study of sacituzumab govitecan in metastatic triple-negative breast cancer. Breast Cancer Res Treat 2022; 195:127-139. [PMID: 35545724 PMCID: PMC9374646 DOI: 10.1007/s10549-022-06602-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/06/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE Sacituzumab govitecan (SG) is an antibody-drug conjugate composed of an anti-Trop-2 antibody coupled to SN-38 via a proprietary hydrolyzable linker. In the ASCENT study, SG improved survival versus single-agent treatment of physician's choice (TPC) in pre-treated metastatic triple-negative breast cancer (mTNBC). Hormone/HER2 receptor changes are common, particularly at relapse/metastasis. This subanalysis assessed outcomes in patients who did/did not have TNBC at initial diagnosis, before enrollment. METHODS TNBC diagnosis was only required at study entry. Patients with mTNBC refractory/relapsing after ≥ 2 prior chemotherapies were randomized 1:1 to receive SG or TPC. Primary endpoint was progression-free survival (PFS) in patients without brain metastases. RESULTS Overall, 70/235 (30%) and 76/233 (33%) patients who received SG and TPC, respectively, did not have TNBC at initial diagnosis. Clinical benefit with SG versus TPC was observed in this subset. Median PFS was 4.6 versus 2.3 months (HR 0.48; 95% CI 0.32-0.72), median overall survival was 12.4 versus 6.7 months (HR 0.44; 95% CI 0.30-0.64), and objective response rate (ORR) was 31% versus 4%; those who also received prior CDK4/6 inhibitors had ORRs of 21% versus 5%. Efficacy and safety for patients with TNBC at initial diagnosis were generally similar to those who did not present with TNBC at initial diagnosis. CONCLUSION Patients without TNBC at initial diagnosis had improved clinical outcomes and a manageable safety profile with SG, supporting SG as a treatment option for mTNBC regardless of subtype at initial diagnosis. Subtype reassessment in advanced breast cancer allows for optimal treatment. Clinical trial registration number NCT02574455, registered October 12, 2015.
Collapse
Affiliation(s)
- Joyce O'Shaughnessy
- Medical Oncology, Texas Oncology-Baylor Charles A. Sammons Cancer Center, 3410 Worth St., Suite 400, Dallas, TX, 75246, USA.
| | - Adam Brufsky
- Magee-Womens Hospital and the Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Hope S Rugo
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Sagar Sardesai
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Erika Hamilton
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | - Delphine Loirat
- Medical Oncology Department and D3i, Institut Curie, Paris, France
| | - Tiffany Traina
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Sara A Hurvitz
- Medical Oncology, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Kevin Kalinsky
- Columbia University Irving Medical Center, New York, NY, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Aditya Bardia
- Department of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Stephanie Henry
- Department of Oncology-Hematology, Radiotherapy, and Nuclear Medicine, CHU UCL Namur, Namur, Belgium
| | - Ingrid Mayer
- Breast Cancer Program, Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Yanni Zhu
- Department of Biostatistics, Gilead Sciences, Inc., Foster City, CA, USA
| | - See Phan
- Department of Clinical Development, Gilead Sciences, Inc., Foster City, CA, USA
| | - Javier Cortés
- International Breast Cancer Center, Quironsalud Group, Barcelona, Spain
| |
Collapse
|
58
|
Marks S, Naidoo J. Antibody drug conjugates in non-small cell lung cancer: An emerging therapeutic approach. Lung Cancer 2021; 163:59-68. [PMID: 34923203 DOI: 10.1016/j.lungcan.2021.11.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 10/19/2022]
Abstract
The current standard-of-care for the treatment of advanced non-small cell lung cancer (NSCLC) incorporates targeted therapies, immune-checkpoint inhibitors (ICI) and systemic chemotherapy. Antibody-drug conjugates (ADC) are a class of anti-cancer therapy capable of transporting cytotoxic drugs directly to tumour cells, thus harnessing the strengths of both cytotoxic chemotherapy and targeted therapy. In this review we provide a comprehensive review the design, mode of action, and mechanisms of resistance to ADCs in NSCLC. We also summarize the clinical development of several promising ADCs in early phase clinical trials for the treatment NSCLC. including ADCs against well-established targets (e.g.HER2 in breast cancer, Nectin4 in urothelial cancer), novel antigenic targets (e.g. HER3, TROP2, PTK7, CEACAM5), as well as promising combinations with agents known to be active in NSCLC such as tyrosine kinase inhibitors and ICI therapy, as a strategy to overcome mechanisms of resistance to ADC therapy.
Collapse
Affiliation(s)
- S Marks
- Beaumont RCSI Cancer Centre, Dublin 9, Republic of Ireland.
| | - J Naidoo
- Beaumont RCSI Cancer Centre, Dublin 9, Republic of Ireland; Upper Aerodigestive Division, Sidney Kimmel Comprehensive Cancer Centre at Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
59
|
António JPM, Carvalho JI, André AS, Dias JNR, Aguiar SI, Faustino H, Lopes RMRM, Veiros LF, Bernardes GJL, Silva FA, Gois PMP. Diazaborines Are a Versatile Platform to Develop ROS‐Responsive Antibody Drug Conjugates**. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202109835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- João P. M. António
- Research Institute for Medicines (iMed.ULisboa) Faculdade de Farmácia Universidade de Lisboa Av. Prof. Gama Pinto 1649-003 Lisboa Portugal
| | - Joana Inês Carvalho
- Research Institute for Medicines (iMed.ULisboa) Faculdade de Farmácia Universidade de Lisboa Av. Prof. Gama Pinto 1649-003 Lisboa Portugal
| | - Ana S. André
- Centro de Investigação Interdisciplinar em Sanidade Animal Faculdade de Medicina Veterinária Universidade de Lisboa Av. Universidade Técnica 1300-477 Lisboa Portugal
| | - Joana N. R. Dias
- Centro de Investigação Interdisciplinar em Sanidade Animal Faculdade de Medicina Veterinária Universidade de Lisboa Av. Universidade Técnica 1300-477 Lisboa Portugal
| | - Sandra I. Aguiar
- Centro de Investigação Interdisciplinar em Sanidade Animal Faculdade de Medicina Veterinária Universidade de Lisboa Av. Universidade Técnica 1300-477 Lisboa Portugal
| | - Hélio Faustino
- Research Institute for Medicines (iMed.ULisboa) Faculdade de Farmácia Universidade de Lisboa Av. Prof. Gama Pinto 1649-003 Lisboa Portugal
| | - Ricardo M. R. M. Lopes
- Research Institute for Medicines (iMed.ULisboa) Faculdade de Farmácia Universidade de Lisboa Av. Prof. Gama Pinto 1649-003 Lisboa Portugal
| | - Luis F. Veiros
- Centro de Química Estrutural and Departamento de Engenharia Química Instituto Superior Técnico Universidade de Lisboa Av. Rovisco Pais 1 1049-001 Lisboa Portugal
| | - Gonçalo J. L. Bernardes
- Instituto de Medicina Molecular João Lobo Antunes Faculdade de Medicina Universidade de Lisboa Avenida Professor Egas Moniz 1649-028 Lisboa Portugal
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road CB2 1EW Cambridge United Kingdom
| | - Frederico A. Silva
- Centro de Investigação Interdisciplinar em Sanidade Animal Faculdade de Medicina Veterinária Universidade de Lisboa Av. Universidade Técnica 1300-477 Lisboa Portugal
| | - Pedro M. P. Gois
- Research Institute for Medicines (iMed.ULisboa) Faculdade de Farmácia Universidade de Lisboa Av. Prof. Gama Pinto 1649-003 Lisboa Portugal
| |
Collapse
|
60
|
António JPM, Carvalho JI, André AS, Dias JNR, Aguiar SI, Faustino H, Lopes RMRM, Veiros LF, Bernardes GJL, da Silva FA, Gois PMP. Diazaborines Are a Versatile Platform to Develop ROS-Responsive Antibody Drug Conjugates*. Angew Chem Int Ed Engl 2021; 60:25914-25921. [PMID: 34741376 DOI: 10.1002/anie.202109835] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Indexed: 12/11/2022]
Abstract
Antibody-drug conjugates (ADCs) are a new class of therapeutics that combine the lethality of potent cytotoxic drugs with the targeting ability of antibodies to selectively deliver drugs to cancer cells. In this study we show for the first time the synthesis of a reactive-oxygen-species (ROS)-responsive ADC (VL-DAB31-SN-38) that is highly selective and cytotoxic to B-cell lymphoma (CLBL-1 cell line, IC50 value of 54.1 nM). The synthesis of this ADC was possible due to the discovery that diazaborines (DABs) are a very effective ROS-responsive unit that are also very stable in buffer and in plasma. DFT calculations performed on this system revealed a favorable energetic profile (ΔGR=-74.3 kcal mol-1 ) similar to the oxidation mechanism of aromatic boronic acids. DABs' very fast formation rate and modularity enabled the construction of different ROS-responsive linkers featuring self-immolative modules, bioorthogonal functions, and bioconjugation handles. These structures were used in the site-selective functionalization of a VL antibody domain and in the construction of the homogeneous ADC.
Collapse
Affiliation(s)
- João P M António
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Joana Inês Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Ana S André
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Joana N R Dias
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Sandra I Aguiar
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Hélio Faustino
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Ricardo M R M Lopes
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Luis F Veiros
- Centro de Química Estrutural and Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisboa, Portugal
| | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal.,Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, United Kingdom
| | - Frederico A da Silva
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Pedro M P Gois
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| |
Collapse
|
61
|
Pavone G, Motta L, Martorana F, Motta G, Vigneri P. A New Kid on the Block: Sacituzumab Govitecan for the Treatment of Breast Cancer and Other Solid Tumors. Molecules 2021; 26:molecules26237294. [PMID: 34885875 PMCID: PMC8659286 DOI: 10.3390/molecules26237294] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Human trophoblast cell-surface antigen-2 (Trop-2) is a membrane glycoprotein involved in cell proliferation and motility, frequently overexpressed in epithelial tumors. Thus, it represents an attractive target for anticancer therapies. Sacituzumab govitecan (SG) is a third-generation antibody-drug conjugate, consisting of an anti-Trop-2 monoclonal antibody (hRS7), a hydrolyzable linker, and a cytotoxin (SN38), which inhibits topoisomerase 1. Specific pharmacological features, such as the high antibody to payload ratio, the ultra-toxic nature of SN38, and the capacity to kill surrounding tumor cells (the bystander effect), make SG a very promising drug for cancer treatment. Indeed, unprecedented results have been observed with SG in patients with heavily pretreated advanced triple-negative breast cancer and urothelial carcinomas, and the drug has already received approval for these indications. These results are coupled with a manageable toxicity profile, with neutropenia and diarrhea as the most frequent adverse events, mainly of grades 1-2. While several trials are exploring SG activity in different tumor types and settings, potential biomarkers of response are under investigation. Among these, Trop-2 overexpression and the presence of BRCA1/2 mutations seem to be the most promising. We review the available literature concerning SG, with a focus on its toxicity spectrum and possible biomarkers of its response.
Collapse
Affiliation(s)
- Giuliana Pavone
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Medical Oncology Unit, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy
| | - Lucia Motta
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Medical Oncology Unit, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy
- Correspondence: ; Tel.: +39-095-3781959
| | - Federica Martorana
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Gianmarco Motta
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Medical Oncology Unit, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy
| | - Paolo Vigneri
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy; (G.P.); (F.M.); (G.M.); (P.V.)
- Medical Oncology Unit, A.O.U. Policlinico “G.Rodolico-S.Marco”, 95123 Catania, Italy
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
62
|
Liao S, Wang B, Zeng R, Bao H, Chen X, Dixit R, Xing X. Recent advances in trophoblast cell-surface antigen 2 targeted therapy for solid tumors. Drug Dev Res 2021; 82:1096-1110. [PMID: 34462935 DOI: 10.1002/ddr.21870] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/27/2021] [Accepted: 05/02/2021] [Indexed: 12/27/2022]
Abstract
Trophoblast cell-surface antigen 2 (Trop 2) is a transmembrane glycoprotein that is highly expressed in various cancer types with relatively low or no baseline expression in most normal tissues. Its overexpression is associated with tumor growth and poor prognosis; Trop 2 is, therefore, an ideal therapeutic target for epithelial cancers. Several Trop 2 targeted therapeutics have recently been developed for the treatment of cancers, such as anti-Trop 2 antibodies and antibody-drug conjugates (ADCs), as well as Trop 2-specific cell therapy. In particular, the safety and clinical benefit of Trop 2-based ADCs have been demonstrated in clinical trials across multiple tumor types, including those with limited treatment options, such as triple-negative breast cancer, platinum-resistant urothelial cancer, and heavily pretreated non-small cell lung cancer. In this review, we elaborate on recent advances in Trop 2 targeted modalities and provide an overview of novel insights for future developments in this field.
Collapse
Affiliation(s)
- Shutan Liao
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Bing Wang
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Rong Zeng
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Haifeng Bao
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Xiaomin Chen
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Rakesh Dixit
- Department of Consultation, Bionavigen LLC, Gaithersburg, Maryland, USA
| | - Xiaoyan Xing
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| |
Collapse
|
63
|
Santi DV, Cabel L, Bidard FC. Does sacituzumab-govitecan act as a conventional antibody drug conjugate (ADC), a prodrug of SN-38 or both? ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1113. [PMID: 34430554 PMCID: PMC8350658 DOI: 10.21037/atm-21-1103] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/03/2021] [Indexed: 01/06/2023]
Affiliation(s)
| | - Luc Cabel
- Medical Oncology, Institut Curie, Paris, France
| | - François-Clément Bidard
- Medical Oncology, Institut Curie, Paris, France.,UVSQ/Paris-Saclay University, Paris, France
| |
Collapse
|
64
|
Ahmed Y, Berenguer-Pina JJ, Mahgoub T. The Rise of the TROP2-Targeting Agents in NSCLC: New Options on the Horizon. Oncology 2021; 99:673-680. [PMID: 34280931 DOI: 10.1159/000517438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Lung cancer is the most common thoracic malignancy, representing the leading cause of cancer-related deaths worldwide with a 5-year survival rate of <10%. SUMMARY The emergence of targeted therapy and immunotherapy has changed the treatment paradigm of advanced non-small cell lung cancer (NSCLC). However, for those who are not eligible for such therapy or currently have no available standard treatment options, new precision treatment approaches are needed. Human trophoblast cell-surface antigen 2 (TROP2) is a transmembrane glycoprotein that is highly expressed on several epithelial tumours including NSCLC. TROP2 is recognized as a promising molecular target for therapeutic development in various types of TROP2-expressing malignancies. As a result, several TROP2-targeted therapeutics have recently been developed for clinical use, such as anti-TROP2 antibodies and TROP2-targeted antibody-drug conjugates. Key Message: This review explores the literature data on the role of TROP2 in cancer development and the potential use of emerging TROP2 antibody-drug conjugates in NSCLC treatment.
Collapse
Affiliation(s)
- Yasar Ahmed
- Medical Oncology Department, St. Vincent's University Hospital, Dublin, Ireland
| | | | - Thamir Mahgoub
- Medical Oncology Department, Mid-Western Cancer Centre, University Hospital Limerick, Limerick, Ireland
| |
Collapse
|
65
|
Khaiwa N, Maarouf NR, Darwish MH, Alhamad DWM, Sebastian A, Hamad M, Omar HA, Orive G, Al-Tel TH. Camptothecin's journey from discovery to WHO Essential Medicine: Fifty years of promise. Eur J Med Chem 2021; 223:113639. [PMID: 34175539 DOI: 10.1016/j.ejmech.2021.113639] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/24/2021] [Accepted: 06/06/2021] [Indexed: 12/16/2022]
Abstract
Nature represents a rich source of compounds used for the treatment of many diseases. Camptothecin (CPT), isolated from the bark of Camptotheca acuminata, is a cytotoxic alkaloid that attenuates cancer cell replication by inhibiting DNA topoisomerase 1. Despite its promising and wide spectrum antiproliferative activity, its use is limited due to low solubility, instability, acquired tumour cell resistance, and remarkable toxicity. This has led to the development of numerous CPT analogues with improved pharmacodynamic and pharmacokinetic profiles. Three natural product-inspired drugs, namely, topotecan, irinotecan, and belotecan, are clinically approved and prescribed drugs for the treatment of several types of cancer, whereas other derivatives are in clinical trials. In this review, which covers literature from 2015 to 2020, we aim to provide a comprehensive overview and describe efforts that led to the development of a variety of CPT analogues. These efforts have led to the discovery of potent, first-in-class chemotherapeutic agents inspired by CPT. In addition, the mechanism of action, SAR studies, and recent advances of novel CPT drug delivery systems and antibody drug conjugates are discussed.
Collapse
Affiliation(s)
- Noura Khaiwa
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Noor R Maarouf
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Mhd H Darwish
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Dima W M Alhamad
- Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates
| | - Anusha Sebastian
- Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates
| | - Mohamad Hamad
- Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates; College of Health Sciences, 27272, Sharjah, United Arab Emirates
| | - Hany A Omar
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates; Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Taleb H Al-Tel
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates; Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
66
|
Biomarker analyses in the phase III ASCENT study of sacituzumab govitecan versus chemotherapy in patients with metastatic triple-negative breast cancer. Ann Oncol 2021; 32:1148-1156. [PMID: 34116144 DOI: 10.1016/j.annonc.2021.06.002] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/20/2021] [Accepted: 06/02/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The pivotal phase III ASCENT trial demonstrated improved survival outcomes associated with sacituzumab govitecan (SG), an anti-trophoblast cell-surface antigen 2 (anti-Trop-2) antibody-drug conjugate linked with the topoisomerase-inhibitor SN-38, over single-agent chemotherapy treatment of physician's choice (TPC) in previously treated metastatic triple-negative breast cancer (mTNBC). This prespecified, exploratory biomarker analysis from the ASCENT trial evaluates the association between tumor Trop-2 expression and germline BRCA1/2 mutation status with clinical outcomes. PATIENTS AND METHODS Patients with mTNBC refractory to or progressing after two or more prior chemotherapies, with one or more in the metastatic setting, were randomized to receive SG (10 mg/kg intravenously days 1 and 8, every 21 days) or TPC (capecitabine, eribulin, vinorelbine, or gemcitabine) until disease progression/unacceptable toxicity. Biopsy or surgical specimens were collected at study entry to determine Trop-2 expression level using a validated immunohistochemistry assay and histochemical scoring. Germline BRCA1/2 mutation status was collected at baseline. RESULTS Of 468 assessable patients, 290 had Trop-2 expression data [64% (n = 151 SG) versus 60% (n = 139 TPC)] and 292 had known BRCA1/2 mutation status [63% (n = 149 SG) versus 61% (n = 143 TPC)]. Median progression-free survival in SG- versus TPC-treated patients was 6.9, 5.6, and 2.7 months versus 2.5, 2.2, and 1.6 months for high, medium, and low Trop-2 expression, respectively. Median overall survival (14.2, 14.9, and 9.3 months versus 6.9, 6.9, and 7.6 months) and objective response rates (44%, 38%, and 22% versus 1%, 11%, and 6%) were numerically higher with SG versus TPC in patients with high, medium, and low Trop-2 expression, respectively. Efficacy outcomes were numerically higher with SG versus TPC in patients with and without germline BRCA1/2 mutations. CONCLUSIONS SG benefits patients with previously treated mTNBC expressing high/medium Trop-2 compared with standard-of-care chemotherapy and regardless of germline BRCA1/2 mutation status. The small number of patients with low Trop-2 expression precludes definitive conclusions on the benefit of SG in this subgroup.
Collapse
|
67
|
Bogen JP, Grzeschik J, Jakobsen J, Bähre A, Hock B, Kolmar H. Treating Bladder Cancer: Engineering of Current and Next Generation Antibody-, Fusion Protein-, mRNA-, Cell- and Viral-Based Therapeutics. Front Oncol 2021; 11:672262. [PMID: 34123841 PMCID: PMC8191463 DOI: 10.3389/fonc.2021.672262] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/11/2021] [Indexed: 01/02/2023] Open
Abstract
Bladder cancer is a frequent malignancy and has a clinical need for new therapeutic approaches. Antibody and protein technologies came a long way in recent years and new engineering approaches were applied to generate innovative therapeutic entities with novel mechanisms of action. Furthermore, mRNA-based pharmaceuticals recently reached the market and CAR-T cells and viral-based gene therapy remain a major focus of biomedical research. This review focuses on the engineering of biologics, particularly therapeutic antibodies and their application in preclinical development and clinical trials, as well as approved monoclonal antibodies for the treatment of bladder cancer. Besides, newly emerging entities in the realm of bladder cancer like mRNA, gene therapy or cell-based therapeutics are discussed and evaluated. As many discussed molecules exhibit unique mechanisms of action based on innovative protein engineering, they reflect the next generation of cancer drugs. This review will shed light on the engineering strategies applied to develop these next generation treatments and provides deeper insights into their preclinical profiles, clinical stages, and ongoing trials. Furthermore, the distribution and expression of the targeted antigens and the intended mechanisms of action are elucidated.
Collapse
Affiliation(s)
- Jan P Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany.,Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Joern Jakobsen
- Ferring Pharmaceuticals, International PharmaScience Center, Copenhagen, Denmark
| | - Alexandra Bähre
- Ferring Pharmaceuticals, International PharmaScience Center, Copenhagen, Denmark
| | - Björn Hock
- Global Pharmaceutical Research and Development, Ferring International Center S.A., Saint-Prex, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
68
|
Furuuchi K, Rybinski K, Fulmer J, Moriyama T, Drozdowski B, Soto A, Fernando S, Wilson K, Milinichik A, Dula ML, Tanaka K, Cheng X, Albone E, Uenaka T. Antibody-drug conjugate MORAb-202 exhibits long-lasting antitumor efficacy in TNBC PDx models. Cancer Sci 2021; 112:2467-2480. [PMID: 33756060 PMCID: PMC8177789 DOI: 10.1111/cas.14898] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
The antibody‐drug conjugate (ADC) MORAb‐202, consisting of farletuzumab paired with a cathepsin B–cleavable linker and eribulin, targets folate receptor alpha (FRA), which is frequently overexpressed in various tumor types. MORAb‐202 was highly cytotoxic to FRA‐positive cells in vitro, with limited off‐target killing of FRA‐negative cells. Furthermore, MORAb‐202 showed a clear in vitro bystander cytotoxic effect in coculture with FRA‐positive/negative cells. In vivo antitumor efficacy studies of MORAb‐202 were conducted with a single administration of MORAb‐202 in triple‐negative breast cancer (TNBC) patient–derived xenograft (PDx) models expressing low and high levels of FRA. MORAb‐202 exhibited durable efficacy proportional to tumor FRA expression. Toxicology studies (Q3Wx2) in nonhuman primates suggested that the major observed toxicity of MORAb‐202 is hematologic toxicity. Overall, these findings support the concept that MORAb‐202 represents a promising investigational ADC for the treatment of TNBC patients.
Collapse
Affiliation(s)
- Keiji Furuuchi
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - Katherine Rybinski
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - James Fulmer
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | | | - Brian Drozdowski
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - Allis Soto
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - Shawn Fernando
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - Kerrianne Wilson
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - Andrew Milinichik
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - Mary Lou Dula
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - Keigo Tanaka
- Tsukuba Research Laboratory, Eisai, Co. Ltd, Tsukuba, Japan
| | - Xin Cheng
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - Earl Albone
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| | - Toshimitsu Uenaka
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Eisai Inc, Exton, PA, USA
| |
Collapse
|
69
|
Bardia A, Messersmith WA, Kio EA, Berlin JD, Vahdat L, Masters GA, Moroose R, Santin AD, Kalinsky K, Picozzi V, O'Shaughnessy J, Gray JE, Komiya T, Lang JM, Chang JC, Starodub A, Goldenberg DM, Sharkey RM, Maliakal P, Hong Q, Wegener WA, Goswami T, Ocean AJ. Sacituzumab govitecan, a Trop-2-directed antibody-drug conjugate, for patients with epithelial cancer: final safety and efficacy results from the phase I/II IMMU-132-01 basket trial. Ann Oncol 2021; 32:746-756. [PMID: 33741442 DOI: 10.1016/j.annonc.2021.03.005] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Sacituzumab govitecan (SG), a trophoblast cell surface antigen-2 (Trop-2)-directed antibody-drug conjugate, has demonstrated antitumor efficacy and acceptable tolerability in a phase I/II multicenter trial (NCT01631552) in patients with advanced epithelial cancers. This report summarizes the safety data from the overall safety population (OSP) and efficacy data, including additional disease cohorts not published previously. PATIENTS AND METHODS Patients with refractory metastatic epithelial cancers received intravenous SG (8, 10, 12, or 18 mg/kg) on days 1 and 8 of 21-day cycles until disease progression or unacceptable toxicity. Endpoints for the OSP included safety and pharmacokinetic parameters with investigator-evaluated objective response rate (ORR per RECIST 1.1), duration of response, clinical benefit rate, progression-free survival, and overall survival evaluated for cohorts (n > 10 patients) of small-cell lung, colorectal, esophageal, endometrial, pancreatic ductal adenocarcinoma, and castrate-resistant prostate cancer. RESULTS In the OSP (n = 495, median age 61 years, 68% female; UGT1A1∗28 homozygous, n = 46; 9.3%), 41 (8.3%) permanently discontinued treatment due to adverse events (AEs). Most common treatment-related AEs were nausea (62.6%), diarrhea (56.2%), fatigue (48.3%), alopecia (40.4%), and neutropenia (57.8%). Most common treatment-related serious AEs (n = 75; 15.2%) were febrile neutropenia (4.0%) and diarrhea (2.8%). Grade ≥3 neutropenia and febrile neutropenia occurred in 42.4% and 5.3% of patients, respectively. Neutropenia (all grades) was numerically more frequent in UGT1A1∗28 homozygotes (28/46; 60.9%) than heterozygotes (69/180; 38.3%) or UGT1A1∗1 wild type (59/177; 33.3%). There was one treatment-related death due to an AE of aspiration pneumonia. Partial responses were seen in endometrial cancer (4/18, 22.2% ORR) and small-cell lung cancer (11/62, 17.7% ORR), and one castrate-resistant prostate cancer patient had a complete response (n = 1/11; 9.1% ORR). CONCLUSIONS SG demonstrated a toxicity profile consistent with previous published reports. Efficacy was seen in several cancer cohorts, which validates Trop-2 as a broad target in solid tumors.
Collapse
Affiliation(s)
- A Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | | | - E A Kio
- Goshen Center for Cancer Care, Goshen, USA
| | - J D Berlin
- Vanderbilt-Ingram Cancer Center, Nashville, USA
| | - L Vahdat
- Weill Cornell Medicine, New York, USA
| | - G A Masters
- Helen F Graham Cancer Center and Research Institute, Newark, USA
| | - R Moroose
- Orlando Health UF Health Cancer Center, Orlando, USA
| | - A D Santin
- Yale University School of Medicine, New Haven, USA
| | - K Kalinsky
- Columbia University Irving Medical Center-Herbert Irving Comprehensive Cancer Center, New York, USA
| | - V Picozzi
- Virginia Mason Cancer Center, Seattle, USA
| | - J O'Shaughnessy
- Texas Oncology, Baylor University Medical Center, US Oncology, Dallas, USA
| | - J E Gray
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, USA
| | - T Komiya
- Parkview Cancer Institute, Fort Wayne, USA
| | - J M Lang
- University of Wisconsin Carbone Cancer Center, Madison, USA
| | - J C Chang
- Houston Methodist Cancer Center, Houston, USA
| | - A Starodub
- Riverside Peninsula Cancer Institute, Newport News, USA
| | - D M Goldenberg
- Immunomedics, Inc., a Subsidiary of Gilead Sciences, Inc., Morris Plains, USA
| | - R M Sharkey
- Immunomedics, Inc., a Subsidiary of Gilead Sciences, Inc., Morris Plains, USA
| | - P Maliakal
- Immunomedics, Inc., a Subsidiary of Gilead Sciences, Inc., Morris Plains, USA
| | - Q Hong
- Immunomedics, Inc., a Subsidiary of Gilead Sciences, Inc., Morris Plains, USA
| | - W A Wegener
- Immunomedics, Inc., a Subsidiary of Gilead Sciences, Inc., Morris Plains, USA
| | - T Goswami
- Immunomedics, Inc., a Subsidiary of Gilead Sciences, Inc., Morris Plains, USA
| | - A J Ocean
- Weill Cornell Medicine, New York, USA.
| |
Collapse
|
70
|
Dourado MR, Machado RA, Paranaíba LMR, González-Arriagada WA, da Silva SD, Sawazaki-Calone Í, Graner E, Salo T, Coletta RD. Trophoblast cell surface antigen 2 expression predicts outcome in oral squamous cell carcinomas. Oral Dis 2021; 28:1085-1093. [PMID: 33615627 DOI: 10.1111/odi.13809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/28/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Trophoblast cell surface antigen 2 (TROP2) has unclear clinical role in oral squamous cell carcinomas (OSCC). Here, we investigated the association of TROP2 immunoexpression with clinicopathological parameters and survival of OSCC patients. SUBJECTS AND METHODS Cancer-specific survival (CSS) and disease-free survival (DFS) were assessed in a cohort composed of 266 OSCC. An independent cohort with 88 OSCC samples matched with the normal oral tissue, as well as 17 metastatic lymph nodes, was used for validation. RESULTS Multivariate analysis showed TROP2 as an independent marker of favorable prognosis for both CSS (HR: 0.60, 95% CI: 0.40-0.90, p = .01) and DFS (HR: 0.57, 95% CI: 0.36-0.89, p = .01). Furthermore, TROP2 protein expression was significantly higher in morphologically normal tissues compared to primary tumors (p < .0001) and lymph node metastases (p = .001), and it was significantly associated with CSS (HR: 0.26, 95% CI: 0.09-0.74, p = .008) in the validation cohort. A pooled mRNA analysis performed on the Oncomine™ database confirmed the underexpression in OSCC compared with normal tissues (p = .014). CONCLUSIONS In summary, our results point to a favorable prognostic significance of TROP2 overexpression in a large cohort of oral cancer patients, suggesting it as an attractive clinical marker.
Collapse
Affiliation(s)
- Mauricio Rocha Dourado
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Renato Assis Machado
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil.,Hospital for Rehabilitation of Craniofacial Anomalies, University of São Paulo (HRAC/USP), Bauru, Brazil
| | - Lívia Máris Ribeiro Paranaíba
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | | | - Sabrina Daniela da Silva
- Lady Davis Institute for Medical Research, Segal Cancer Center, Jewish General Hospital, Montreal, QC, Canada.,Department of Otolaryngology-Head and Neck Surgery, McGill University, Montreal, QC, Canada
| | - Íris Sawazaki-Calone
- Department of Oral Pathology and Oral Medicine, Dental School, Western Paraná State University, Cascavel, Brazil
| | - Edgard Graner
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine and Medical Research Centre Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland.,Department of Pathology, Institute of Oral and Maxillofacial Disease, Helsinki University Hospital, University of Helsinki and HUSLAB, Helsinki, Finland
| | - Ricardo D Coletta
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| |
Collapse
|
71
|
Matsuda Y, Mendelsohn BA. An overview of process development for antibody-drug conjugates produced by chemical conjugation technology. Expert Opin Biol Ther 2020; 21:963-975. [PMID: 33141625 DOI: 10.1080/14712598.2021.1846714] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: We discuss chemical conjugation strategies for antibody-drug conjugates (ADCs) from an industrial perspective and compare three promising chemical conjugation technologies to produce site-specific ADCs.Areas covered: Currently, nine ADCs are commercially approved and all are produced by chemical conjugation technology. However, seven of these ADCs contain a relatively broad drug distribution, potentially limiting their therapeutic indices. In 2019, the first site-specific ADC was launched on the market by Daiichi-Sankyo. This achievement, and an analysis of clinical trials over the last decade, indicates that current industrial interest in the ADC field is shifting toward site-specific conjugation technologies. From an industrial point of view, we aim to provide guidance regarding established conjugation methodologies that have already been applied to scale-up stages. With an emphasis on highly productive, scalable, and synthetic process robustness, conjugation methodologies for ADC production is discussed herein.Expert opinion: All three chemical conjugation technologies described in this review have various advantages and disadvantages, therefore drug developers can utilize these depending on their biological and/or protein targets. The future landscape of the ADC field is also discussed.
Collapse
Affiliation(s)
- Yutaka Matsuda
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Brian A Mendelsohn
- Process Development & Tech Transfer, Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, CA 92121, United States
| |
Collapse
|
72
|
Lenárt S, Lenárt P, Šmarda J, Remšík J, Souček K, Beneš P. Trop2: Jack of All Trades, Master of None. Cancers (Basel) 2020; 12:E3328. [PMID: 33187148 PMCID: PMC7696911 DOI: 10.3390/cancers12113328] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Trophoblast cell surface antigen 2 (Trop2) is a widely expressed glycoprotein and an epithelial cell adhesion molecule (EpCAM) family member. Although initially identified as a transmembrane protein, other subcellular localizations and processed forms were described. Its congenital mutations cause a gelatinous drop-like corneal dystrophy, a disease characterized by loss of barrier function in corneal epithelial cells. Trop2 is considered a stem cell marker and its expression associates with regenerative capacity in various tissues. Trop2 overexpression was described in tumors of different origins; however, functional studies revealed both oncogenic and tumor suppressor roles. Nevertheless, therapeutic potential of Trop2 was recognized and clinical studies with drug-antibody conjugates have been initiated in various cancer types. One of these agents, sacituzumab govitecan, has been recently granted an accelerated approval for therapy of metastatic triple-negative breast cancer. In this article, we review the current knowledge about the yet controversial function of Trop2 in homeostasis and pathology.
Collapse
Affiliation(s)
- Sára Lenárt
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
| | - Peter Lenárt
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Jan Šmarda
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
| | - Ján Remšík
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Karel Souček
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 612 65 Brno, Czech Republic
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Petr Beneš
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| |
Collapse
|
73
|
Weddell J, Chiney MS, Bhatnagar S, Gibbs JP, Shebley M. Mechanistic Modeling of Intra-Tumor Spatial Distribution of Antibody-Drug Conjugates: Insights into Dosing Strategies in Oncology. Clin Transl Sci 2020; 14:395-404. [PMID: 33073529 PMCID: PMC7877868 DOI: 10.1111/cts.12892] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Antibody drug conjugates (ADCs) provide targeted delivery of cytotoxic agents directly inside tumor cells. However, many ADCs targeting solid tumors have exhibited limited clinical efficacy, in part, due to insufficient penetration within tumors. To better understand the relationship between ADC tumor penetration and efficacy, previously applied Krogh cylinder models that explore tumor growth dynamics following ADC administration in preclinical species were expanded to a clinical framework by integrating clinical pharmacokinetics, tumor penetration, and tumor growth inhibition. The objective of this framework is to link ADC tumor penetration and distribution to clinical efficacy. The model was validated by comparing virtual patient population simulations to observed overall response rates from trastuzumab‐DM1 treated patients with metastatic breast cancer. To capture clinical outcomes, we expanded upon previous Krogh cylinder models to include the additional mechanism of heterogeneous tumor growth inhibition spatially across the tumor. This expansion mechanistically captures clinical response rates by describing heterogeneous ADC binding and tumor cell killing; high binding and tumor cell death close to capillaries vs. low binding, and high tumor cell proliferation far from capillaries. Sensitivity analyses suggest that clinical efficacy could be optimized through dose fractionation, and that clinical efficacy is primarily dependent on the ADC‐target affinity, payload potency, and tumor growth rate. This work offers a mechanistic basis to predict and optimize ADC clinical efficacy for solid tumors, allowing dosing strategy optimization to improve patient outcomes.
Collapse
Affiliation(s)
- Jared Weddell
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois, USA
| | - Manoj S Chiney
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois, USA
| | - Sumit Bhatnagar
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois, USA
| | - John P Gibbs
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois, USA
| | - Mohamad Shebley
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois, USA
| |
Collapse
|
74
|
Kalinsky K, Diamond JR, Vahdat LT, Tolaney SM, Juric D, O'Shaughnessy J, Moroose RL, Mayer IA, Abramson VG, Goldenberg DM, Sharkey RM, Maliakal P, Hong Q, Goswami T, Wegener WA, Bardia A. Sacituzumab govitecan in previously treated hormone receptor-positive/HER2-negative metastatic breast cancer: final results from a phase I/II, single-arm, basket trial. Ann Oncol 2020; 31:1709-1718. [PMID: 32946924 DOI: 10.1016/j.annonc.2020.09.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/01/2020] [Accepted: 09/06/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Trophoblast cell-surface antigen-2 (Trop-2) is expressed in epithelial cancers, including hormone receptor-positive (HR+) metastatic breast cancer (mBC). Sacituzumab govitecan (SG; Trodelvy®) is an antibody-drug conjugate composed of a humanized anti-Trop-2 monoclonal antibody coupled to SN-38 at a high drug-to-antibody ratio via a unique hydrolyzable linker that delivers SN-38 intracellularly and in the tumor microenvironment. SG was granted accelerated FDA approval for metastatic triple-negative BC treatment in April 2020. PATIENTS AND METHODS We analyzed a prespecified subpopulation of patients with HR+/human epidermal growth factor receptor 2-negative (HER2-) HR+/HER2- mBC from the phase I/II, single-arm trial (NCT01631552), who received intravenous SG (10 mg/kg) and whose disease progressed on endocrine-based therapy and at least one prior chemotherapy for mBC. End points included objective response rate (ORR; RECIST version 1.1) assessed locally, duration of response (DOR), clinical benefit rate, progression-free survival (PFS), overall survival (OS), and safety. RESULTS Fifty-four women were enrolled between 13 February 2015 and 1 June 2017. Median (range) age was 54 (33-79) years and all received at least two prior lines of therapy for mBC. At data cut-off (1 March 2019), 12 patients were still alive. Key grade ≥3 treatment-related toxicities included neutropenia (50.0%), anemia (11.1%), and diarrhea (7.4%). Two patients discontinued treatment due to treatment-related adverse events. No treatment-related deaths occurred. At a median follow-up of 11.5 months, the ORR was 31.5% [95% confidence interval (CI), 19.5%-45.6%; 17 partial responses]; median DOR was 8.7 months (95% CI 3.7-12.7), median PFS was 5.5 months (95% CI 3.6-7.6), and median OS was 12 months (95% CI 9.0-18.2). CONCLUSIONS SG shows encouraging activity in patients with pretreated HR+/HER2- mBC and a predictable, manageable safety profile. Further evaluation in a randomized phase III trial (TROPiCS-02) is ongoing (NCT03901339). TRIAL REGISTRATION ClinicalTrials.gov NCT01631552; https://clinicaltrials.gov/ct2/show/NCT01631552.
Collapse
Affiliation(s)
- K Kalinsky
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center-Herbert Irving Comprehensive Cancer Center, New York, USA.
| | - J R Diamond
- Department of Medicine, Medical Oncology, University of Colorado Cancer Center, Aurora, USA
| | - L T Vahdat
- Department of Medicine, Weill Cornell Medical College, New York, USA
| | - S M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - D Juric
- Department of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - J O'Shaughnessy
- Department of Medical Oncology, Texas Oncology, Baylor University Medical Center, US Oncology, Dallas, USA
| | - R L Moroose
- Department of Hematology/Oncology, Orlando Health UF Health Cancer Center, Orlando, USA
| | - I A Mayer
- Department of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Nashville, USA
| | - V G Abramson
- Department of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Nashville, USA
| | - D M Goldenberg
- Clinical Development, Immunomedics, Inc., Morris Plains, USA
| | - R M Sharkey
- Clinical Development, Immunomedics, Inc., Morris Plains, USA
| | - P Maliakal
- Clinical Development, Immunomedics, Inc., Morris Plains, USA
| | - Q Hong
- Clinical Development, Immunomedics, Inc., Morris Plains, USA
| | - T Goswami
- Clinical Development, Immunomedics, Inc., Morris Plains, USA
| | - W A Wegener
- Clinical Development, Immunomedics, Inc., Morris Plains, USA
| | - A Bardia
- Department of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| |
Collapse
|
75
|
Manzano A, Ocaña A. Antibody-Drug Conjugates: A Promising Novel Therapy for the Treatment of Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12082223. [PMID: 32784819 PMCID: PMC7464539 DOI: 10.3390/cancers12082223] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Antibody-drug conjugates (ADCs) represent a novel and promising therapeutic strategy for the treatment of cancer patients. ADCs target antigens highly expressed on the membrane surface of tumor cells to selectively deliver a cytotoxic drug. Ovarian tumors differentially express tumor-specific antigens, which can be used to guide ADCs. This strategy allows for optimizing tumor targeting while minimizing systemic toxicity compared to classical chemotherapeutic agents. ADCs can be improved by using a cleavable linker allowing the delivery of the toxic payload in surrounding cells not expressing the target protein, therefore acting on heterogeneous tumors with different cell populations. Currently, more than 15 ADCs are under preclinical investigation in ovarian cancer, and some of them have already been tested in early-phase clinical trials with promising results. In this review, we summarize the mechanism of action and the toxicity profile of ADCs and discuss the latest preclinical discoveries and forthcoming applications in ovarian cancer.
Collapse
|
76
|
Antibody-Drug Conjugates and Targeted Treatment Strategies for Hepatocellular Carcinoma: A Drug-Delivery Perspective. Molecules 2020; 25:molecules25122861. [PMID: 32575828 PMCID: PMC7356544 DOI: 10.3390/molecules25122861] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
Increased understanding of cancer biology, pharmacology and drug delivery has provided a new framework for drug discovery and product development that relies on the unique expression of specific macromolecules (i.e., antigens) on the surface of tumour cells. This has enabled the development of anti-cancer treatments that combine the selectivity of antibodies with the efficacy of highly potent chemotherapeutic small molecules, called antibody-drug conjugates (ADCs). ADCs are composed of a cytotoxic drug covalently linked to an antibody which then selectively binds to a highly expressed antigen on a cancer cell; the conjugate is then internalized by the cell where it releases the potent cytotoxic drug and efficiently kills the tumour cell. There are, however, many challenges in the development of ADCs, mainly around optimizing the therapeutic/safety benefits. These challenges are discussed in this review; they include issues with the plasma stability and half-life of the ADC, its transport from blood into and distribution throughout the tumour compartment, cancer cell antigen expression and the ADC binding affinity to the target antigen, the cell internalization process, cleaving of the cytotoxic drug from the ADC, and the cytotoxic effect of the drug on the target cells. Finally, we present a summary of some of the experimental ADC strategies used in the treatment of hepatocellular carcinoma, from the recent literature.
Collapse
|