1
|
Mallick S, Duttaroy AK, Dutta S. The PIK3CA gene and its pivotal role in tumor tropism of triple-negative breast cancer. Transl Oncol 2024; 50:102140. [PMID: 39369580 PMCID: PMC11491976 DOI: 10.1016/j.tranon.2024.102140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/04/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
The PIK3CA gene is a linchpin in the intricate molecular network governing triple-negative breast cancer (TNBC) tumor tropism, serving as a focal point for understanding this aggressive disease. Anchored within the PI3K/AKT/mTOR signaling axis, PIK3CA mutations exert substantial influence, driving cellular processes that highlight the unique biology of TNBC. This review meticulously highlights the association between PIK3CA mutations and distinct TNBC subtypes, elucidating the gene's multifaceted contributions to tumor tropism. Molecular dissection reveals how PIK3CA mutations dynamically modulate chemokine responses, growth factor signaling, and extracellular matrix interactions, orchestrating the complex migratory behaviour characteristic of TNBC cells. A detailed exploration of PIK3CA-targeted strategies in the therapeutic arena is presented, outlining the current landscape of clinical trials and precision medicine approaches. As the scientific narrative converges, this review underscores the critical role of PIK3CA in shaping the molecular intricacies of TNBC tumor tropism and illuminates pathways toward tailored interventions, promising a paradigm shift in the clinical management of TNBC.
Collapse
Affiliation(s)
- Sumit Mallick
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Mangalore, Karnataka, India
| | - Asim K Duttaroy
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway.
| | - Suman Dutta
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK; Dorothy Crowfoot Hodgkin Building, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
2
|
Guidi L, Boldrini L, Trapani D, Curigliano G. Antibody-drug conjugates in metastatic breast cancer: sequencing, combinations and resistances. Curr Opin Oncol 2024; 36:487-494. [PMID: 39246172 DOI: 10.1097/cco.0000000000001087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
PURPOSE OF REVIEW Significant advancements have been made in treating metastatic breast cancer (MBC) with antibody drug conjugates (ADCs). However, due to the development of resistance, patients experience disease progression. The aim of this review is to summarize current evidence on ADCs sequencing strategies and combination approaches in the treatment of MBC. RECENT FINDINGS Concerning HER2 positive MBC, current evidence on the optimal ADC-sequencing is primarily about T-DXd, which demonstrated therapeutic value when used post-T-DM1. Conversely, data are limited about the reverse sequence. Similarly, in HER2-negative MBC, recent studies evaluated the sequential use of Sacituzumab Govitecan and T-DXd, which was associated with poor responses. Retrospective analyses have not demonstrated an optimal sequencing strategy for ADCs, and it is still very unclear whether switching the payload or targeting a different antigen may represent the best approach. Combinations may better overcome ADC resistance: interesting data associating immunotherapy or tyrosine kinase inhibitors to ADCs appear promising, albeit data are still immature. SUMMARY In MBC, ADCs have expanded treatment options but their sequential use requires further study. Evidence suggests that sequencing ADCs with similar payloads is ineffective, though current data are inconclusive. More research is needed to optimize treatment strategies, including potential combination therapies.
Collapse
Affiliation(s)
- Lorenzo Guidi
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Laura Boldrini
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
3
|
Sawant S, Naik GG, Sahu AN, Jagtap VA. Understanding the chemistry & pharmacology of antibody-drug conjugates in triple-negative breast cancer with special reference to exatecan derivatives. Med Oncol 2024; 41:301. [PMID: 39460856 DOI: 10.1007/s12032-024-02542-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
In the spectrum of breast malignancies, triple-negative breast cancer is the most widely spreading subtype of breast cancer due to a low availability of therapeutic remedies. Recently, antibody-drug conjugates dramatically resolved the landscape for the treatment of triple-negative breast cancer. This review mainly focuses on the chemistry, structure, mechanism of action, and role of antibody-drug conjugates in triple-negative breast cancer. Datopotecan Deruxtecan (Dato-DXd) is a new-generation ADC showing encouraging results for TNBC. In this review, we have also emphasized TROP-2-directed Datopotamab deruxtecan ADCs to treat triple-negative breast cancer, its synthesis, mechanism of action, pharmacokinetics, pharmacodynamics, adverse events, and their ongoing clinical trials.
Collapse
Affiliation(s)
- Sanjana Sawant
- Department of Pharmaceutical Chemistry, Yashwantrao Bhonsale College of Pharmacy, Affiliated to Mumbai University, Sawantwadi, 416510, India
| | - Gaurav Gopal Naik
- Department of Pharmaceutical Chemistry, Yashwantrao Bhonsale College of Pharmacy, Affiliated to Mumbai University, Sawantwadi, 416510, India.
| | | | - Vijay A Jagtap
- Department of Pharmaceutical Chemistry, Yashwantrao Bhonsale College of Pharmacy, Affiliated to Mumbai University, Sawantwadi, 416510, India
| |
Collapse
|
4
|
Gaudio G, Martino E, Pellizzari G, Cavallone M, Castellano G, Omar A, Katselashvili L, Trapani D, Curigliano G. Developing combination therapies with biologics in triple-negative breast cancer. Expert Opin Biol Ther 2024; 24:1075-1094. [PMID: 39360776 DOI: 10.1080/14712598.2024.2408756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/22/2024] [Indexed: 10/11/2024]
Abstract
INTRODUCTION Novel compounds have entered the triple-negative breast cancer (TNBC) treatment algorithm, namely immune checkpoints inhibitors (ICIs), PARP inhibitors and antibody-drug conjugates (ADCs). The optimization of treatment efficacy can be enhanced with the use of combination treatments, and the incorporation of novel compounds. In this review, we discuss the combination treatments under development for the treatment of TNBC. AREAS COVERED The development of new drugs occurring in recent years has boosted the research for novel combinations to target TNBC heterogeneity and improve outcomes. ICIs, ADCs, tyrosine kinase inhibitors (TKIs), and PARP inhibitors have emerged as leading players in this new landscape, while other compounds like novel intracellular pathways inhibitors or cancer vaccines are drawing more and more interest. The future of TNBC is outlined in combination approaches, and based on new cancer targets, including many chemotherapy-free treatments. EXPERT OPINION A large number of TNBC therapies have either proved clinically ineffective or weighted by unacceptable safety profiles. Others, however, have provided promising results and are currently in late-stage clinical trials, while a few have actually changed clinical practice in recent years. As novel, more and more selective drugs come up, combination strategies focusing the concept of synergy are fully warranted for the future.
Collapse
Affiliation(s)
- Gilda Gaudio
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Enzo Martino
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Gloria Pellizzari
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Matteo Cavallone
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Grazia Castellano
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Abeid Omar
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Nuclear Medicine, Kenyatta University Teaching Referral and Research Hospital, Nairobi, Kenya
| | - Lika Katselashvili
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology, Caucasus Medical Centre, Tbilisi, Georgia
| | - Dario Trapani
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
5
|
Wang J, Tong Z, Tan Y, Shi Y, Wu Y, Zhou Q, Xing X, Chen X, Qiu F, Ma F. Phase 1a study of ESG401, a Trop2 antibody-drug conjugate, in patients with locally advanced/metastatic solid tumors. Cell Rep Med 2024; 5:101707. [PMID: 39216478 PMCID: PMC11524954 DOI: 10.1016/j.xcrm.2024.101707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/14/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
This phase 1a study assesses ESG401 in patients with heavily pretreated locally advanced or metastatic solid tumors, focusing on metastatic breast cancer. Forty patients are enrolled: three experience dose-limiting toxicities, establishing the maximum tolerated dose at 16 mg/kg on days 1, 8, and 15 of a 28-day cycle. The most common grade ≥3 treatment-related adverse events are neutropenia and leukopenia. Among 38 efficacy-evaluable patients, the objective response rate (ORR) is 34.2%, the disease control rate (DCR) is 65.8%, and the clinical benefit rate (CBR) is 50.0% (including stable disease for at least 6 months). The median progression-free survival is 5.1 months, and the median duration of response is 6.3 months. In patients receiving therapeutically relevant doses, the ORR, DCR, and CBR are 40.6%, 75.0%, and 56.3%, respectively. ESG401 demonstrates a favorable safety profile and promising antitumor activity in this heavily treated population. The trial is registered at ClinicalTrials.gov (NCT04892342).
Collapse
Affiliation(s)
- Jiani Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongsheng Tong
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Yinuo Tan
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yehui Shi
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Yun Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Zhou
- Shanghai Escugen Biotechnology Co., Ltd., Shanghai, China
| | - Xiaoyan Xing
- Shanghai Escugen Biotechnology Co., Ltd., Shanghai, China
| | - Xiaomei Chen
- Shanghai Escugen Biotechnology Co., Ltd., Shanghai, China
| | - Fuming Qiu
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Pinto A, Guarini C, Giampaglia M, Sanna V, Melaccio A, Lanotte L, Santoro AN, Pini F, Cusmai A, Giuliani F, Gadaleta-Caldarola G, Fedele P. Synergizing Immunotherapy and Antibody-Drug Conjugates: New Horizons in Breast Cancer Therapy. Pharmaceutics 2024; 16:1146. [PMID: 39339183 PMCID: PMC11435286 DOI: 10.3390/pharmaceutics16091146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
The advent of immunotherapy and antibody-drug conjugates (ADCs) have revolutionized breast cancer treatment, offering new hope to patients. However, challenges, such as resistance and limited efficacy in certain cases, remain. Recently, the combination of these therapies has emerged as a promising approach to address these challenges. ADCs play a crucial role by delivering cytotoxic agents directly to breast cancer cells, minimizing damage to healthy tissue and enhancing the tumor-killing effect. Concurrently, immunotherapies harness the body's immune system to recognize and eliminate cancer cells. This integration offers potential to overcome resistance mechanisms and significantly improve therapeutic outcomes. This review explores the rationale behind combining immunotherapies with ADCs, recent advances in this field, and the potential implications for breast cancer treatment.
Collapse
Affiliation(s)
- Antonello Pinto
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| | - Chiara Guarini
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| | | | - Valeria Sanna
- Oncology Unit, "Ospedale Civile Santissima Annunziata" Hospital, 07100 Sassari, Italy
| | | | - Laura Lanotte
- Oncology Unit, "Mons. Dimiccoli" Hospital, 70051 Barletta, Italy
| | | | - Francesca Pini
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| | - Antonio Cusmai
- "Don Tonino Bello", I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy
| | | | | | - Palma Fedele
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| |
Collapse
|
7
|
Bardia A, Krop IE, Kogawa T, Juric D, Tolcher AW, Hamilton EP, Mukohara T, Lisberg A, Shimizu T, Spira AI, Tsurutani J, Damodaran S, Papadopoulos KP, Greenberg J, Kobayashi F, Zebger-Gong H, Wong R, Kawasaki Y, Nakamura T, Meric-Bernstam F. Datopotamab Deruxtecan in Advanced or Metastatic HR+/HER2- and Triple-Negative Breast Cancer: Results From the Phase I TROPION-PanTumor01 Study. J Clin Oncol 2024; 42:2281-2294. [PMID: 38652877 PMCID: PMC11210948 DOI: 10.1200/jco.23.01909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/09/2024] [Accepted: 02/08/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE Datopotamab deruxtecan (Dato-DXd) is an antibody-drug conjugate consisting of a humanized antitrophoblast cell-surface antigen 2 (TROP2) monoclonal antibody linked to a potent, exatecan-derived topoisomerase I inhibitor payload via a plasma-stable, selectively cleavable linker. PATIENTS AND METHODS TROPION-PanTumor01 (ClinicalTrials.gov identifier: NCT03401385) is a phase I, dose-escalation, and dose-expansion study evaluating Dato-DXd in patients with previously treated solid tumors. The primary study objective was to assess the safety and tolerability of Dato-DXd. Secondary objectives included evaluation of antitumor activity and pharmacokinetics. Results from patients with advanced/metastatic hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) breast cancer (BC) or triple-negative BC (TNBC) are reported. RESULTS At data cutoff (July 22, 2022), 85 patients (HR+/HER2- BC = 41, and TNBC = 44) had received Dato-DXd. The objective response rate by blinded independent central review was 26.8% (95% CI, 14.2 to 42.9) and 31.8% (95% CI, 18.6 to 47.6) for patients with HR+/HER2- BC and TNBC, respectively. The median duration of response was not evaluable in the HR+/HER2- BC cohort and 16.8 months in the TNBC cohort. The median progression-free survival in patients with HR+/HER2- BC and TNBC was 8.3 and 4.4 months, respectively. All-cause treatment-emergent adverse events (TEAEs; any grade, grade ≥3) were observed in 100% and 41.5% of patients with HR+/HER2- BC and 100% and 52.3% of patients with TNBC. Stomatitis was the most common TEAE (any grade, grade ≥3) in both HR+/HER2- BC (82.9%, 9.8%) and TNBC (72.7%, 11.4%) cohorts. CONCLUSION In patients with heavily pretreated advanced HR+/HER2- BC and TNBC, Dato-DXd demonstrated promising clinical activity and a manageable safety profile. Dato-DXd is currently being evaluated in phase III studies.
Collapse
MESH Headings
- Humans
- Female
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/pathology
- Middle Aged
- Aged
- Immunoconjugates/therapeutic use
- Immunoconjugates/adverse effects
- Immunoconjugates/pharmacokinetics
- Adult
- Receptor, ErbB-2/metabolism
- Camptothecin/analogs & derivatives
- Camptothecin/therapeutic use
- Receptors, Estrogen/metabolism
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Receptors, Progesterone/metabolism
- Antigens, Neoplasm
- Cell Adhesion Molecules/metabolism
- Trastuzumab
Collapse
Affiliation(s)
- Aditya Bardia
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Ian E. Krop
- Yale Cancer Center, New Haven, CT
- Dana-Farber Cancer Institute, Boston, MA
| | - Takahiro Kogawa
- Department of Advanced Medical Development, Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Dejan Juric
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Anthony W. Tolcher
- South Texas Accelerated Research Therapeutics, San Antonio, TX
- NEXT Oncology, San Antonio, TX
- Texas Oncology, San Antonio, TX
| | - Erika P. Hamilton
- Sarah Cannon Research Institute, Nashville, TN
- Tennessee Oncology, PLLC, Nashville, TN
| | - Toru Mukohara
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Aaron Lisberg
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Toshio Shimizu
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Pulmonary Medicine and Medical Oncology, Wakayama Medical University Hospital, Wakayama, Japan
| | | | - Junji Tsurutani
- Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan
| | - Senthil Damodaran
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Jonathan Greenberg
- Global Oncology Clinical Development, Daiichi Sankyo, Inc, Basking Ridge, NJ
- Global Oncology Clinical Development, Daiichi Sankyo Europe GmbH, Munich, Germany
| | | | - Hong Zebger-Gong
- Global Oncology Clinical Development, Daiichi Sankyo Europe GmbH, Munich, Germany
| | - Rie Wong
- Global Oncology Clinical Development, Daiichi Sankyo, Co, Ltd, Tokyo, Japan
| | - Yui Kawasaki
- Global Oncology Clinical Development, Daiichi Sankyo, Inc, Basking Ridge, NJ
| | | | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
8
|
Keskinkilic M, Sacks R. Antibody-Drug Conjugates in Triple Negative Breast Cancer. Clin Breast Cancer 2024; 24:163-174. [PMID: 38341370 DOI: 10.1016/j.clbc.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 02/12/2024]
Abstract
Triple negative breast cancer (TNBC) accounts for 15%-20% of all breast cancer. It is a heterogeneous breast cancer subtype with a poor prognosis. Given these negative features, there is a need for new treatment options beyond conventional chemotherapy in both the early stage and palliative setting. Impressive results have been reported with antibody-drug conjugates (ADCs) that link a cytotoxic payload to a monoclonal antibody, such as sacituzumab govitecan and trastuzumab deruxtecan, in the metastatic stage. The focus of this review is to discuss completed and ongoing trials involving ADCs in TNBC.
Collapse
Affiliation(s)
- Merve Keskinkilic
- Department of Medical Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Ruth Sacks
- Winship Cancer Institute, Emory University, Atlanta GA.
| |
Collapse
|
9
|
Anderson TS, McCormick AL, Smith SL, Lowe DB. Modeling antibody drug conjugate potential using a granzyme B antibody fusion protein. BMC Biol 2024; 22:66. [PMID: 38486229 PMCID: PMC10941411 DOI: 10.1186/s12915-024-01860-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Antibody drug conjugates (ADCs) constitute a promising class of targeted anti-tumor therapeutics that harness the selectivity of monoclonal antibodies with the potency of cytotoxic drugs. ADC development is best suited to initially screening antibody candidates for desired properties that potentiate target cell cytotoxicity. However, validating and producing an optimally designed ADC requires expertise and resources not readily available to certain laboratories. RESULTS In this study, we propose a novel approach to help streamline the identification of potential ADC candidates by utilizing a granzyme B (GrB)-based antibody fusion protein (AFP) for preliminary screening. GrB is a non-immunogenic serine protease expressed by immune effector cells such as CD8 + T cells that induces apoptotic activity and can be leveraged for targeted cell killing. CONCLUSIONS Our innovative model allows critical antibody parameters (including target cell binding, internalization, and cytotoxic potential) to be more reliably evaluated in vitro through the creation of an ADC surrogate. Successful incorporation of this AFP could also significantly expand and enhance ADC development pre-clinically, ultimately leading to the accelerated translation of ADC therapies for patients.
Collapse
Affiliation(s)
- Trevor S Anderson
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA
| | - Amanda L McCormick
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA
| | - Savanna L Smith
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA
| | - Devin B Lowe
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA.
| |
Collapse
|
10
|
Bardia A, Jhaveri K, Kalinsky K, Pernas S, Tsurutani J, Xu B, Hamilton E, Im SA, Nowecki Z, Sohn J, Laurentiis MD, Jañez NM, Adamo B, Lee KS, Jung KH, Rubovszky G, Tseng LM, Lu YS, Yuan Y, Maxwell MJ, Haddad V, Khan SS, Rugo HS, Pistilli B. TROPION-Breast01: Datopotamab deruxtecan vs chemotherapy in pre-treated inoperable or metastatic HR+/HER2- breast cancer. Future Oncol 2024; 20:423-436. [PMID: 37387213 DOI: 10.2217/fon-2023-0188] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Abstract
Improving the prognosis for patients with metastatic HR+/HER2- breast cancer remains an unmet need. Patients with tumors that have progressed on endocrine therapy and/or are not eligible for endocrine therapy had limited treatment options beyond chemotherapy. Antibody-drug conjugates are a novel and promising treatment class in this setting. Datopotamab deruxtecan (Dato-DXd) consists of a TROP2-directed humanized IgG1 monoclonal antibody attached via a serum-stable cleavable linker to a topoisomerase I inhibitor payload. TROPION-Breast01 is an ongoing phase III study that is evaluating the efficacy and safety of Dato-DXd compared with investigator's choice of standard-of-care chemotherapy in patients with inoperable or metastatic HR+/HER2- breast cancer who have received one or two prior lines of systemic chemotherapy in the inoperable or metastatic setting. Clinical Trial Registration: NCT05104866 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Aditya Bardia
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Kevin Kalinsky
- Winship Cancer Institute at Emory University, Atlanta, GA, USA
| | - Sonia Pernas
- Institut Català d'Oncologia, IDIBELL, L'Hospitalet, Barcelona, Spain
| | | | - Binghe Xu
- National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Erika Hamilton
- Sarah Cannon Research Institute / Tennessee Oncology, Nashville, TN, USA
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Zbigniew Nowecki
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Joohyuk Sohn
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | - Noelia Martínez Jañez
- Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Barbara Adamo
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Keun Seok Lee
- Center for Breast Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Kyung Hae Jung
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | - Ling-Ming Tseng
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Shen Lu
- National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yuan Yuan
- Formerly City of Hope Comprehensive Cancer Center, Duarte, CA, USA; Currently: Cedars-Sinai Cancer Center, Los Angeles, CA, USA
| | | | | | | | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA, USA
| | | |
Collapse
|
11
|
Dri A, Arpino G, Bianchini G, Curigliano G, Danesi R, De Laurentiis M, Del Mastro L, Fabi A, Generali D, Gennari A, Guarneri V, Santini D, Simoncini E, Zamagni C, Puglisi F. Breaking barriers in triple negative breast cancer (TNBC) - Unleashing the power of antibody-drug conjugates (ADCs). Cancer Treat Rev 2024; 123:102672. [PMID: 38118302 DOI: 10.1016/j.ctrv.2023.102672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/10/2023] [Accepted: 12/13/2023] [Indexed: 12/22/2023]
Abstract
Antibody-drug conjugates (ADCs) represent a novel class of molecules composed of a recombinant monoclonal antibody targeted to a specific cell surface antigen, conjugated to a cytotoxic agent through a cleavable or non-cleavable synthetic linker. The rationale behind the development of ADCs is to overcome the limitations of conventional chemotherapy, such as the narrow therapeutic window and the emergence of resistance mechanisms. ADCs had already revolutionized the treatment algorithm of HER2-positive breast cancer. Currently, emergent non-HER2 targeted ADCs are gaining momentum, with special focus on triple-negative disease therapeutic landscape. Sacituzumab govitecan (SG) is an ADC consisting of a humanized monoclonal antibody hRS7 targeting trophoblast cell surface antigen 2 (Trop2), linked to the topoisomerase I inhibitor SN-38 by a hydrolysable linker. It currently stands as the only non-HER2 targeted ADC that already received approval for the treatment of unresectable locally advanced or metastatic triple negative breast cancer (TNBC) in patients who had received two or more prior systemic therapies, with at least one for advanced disease. The purpose of these review is to analyze the available evidence regarding ADCs in TNBC, alongside with providing an overview on the ongoing and future research horizons in this field.
Collapse
Affiliation(s)
- Arianna Dri
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano (PN), Italy.
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology (IEO), IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michelino De Laurentiis
- Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy; Department of Medical Oncology, Clinical Unit of Medical Oncology, IRCCS Hospital Policlinico San Martino, Genova, Italy
| | - Alessandra Fabi
- Precision Medicine in Breast Cancer Unit, Department of Woman and Child Health and Public Health, IRCCS, Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Daniele Generali
- Department of Medicine, Surgery and Health Sciences, Cattinara Hospital, University of Trieste, Trieste, Italy; Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Alessandra Gennari
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy, Ospedale Maggiore della Caritá, Novara, Italy
| | - Valentina Guarneri
- Medical Oncology 2, Veneto Institute of Oncology (IOV), IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Daniele Santini
- Oncologia Medica A, Policlinico Umberto 1, La Sapienza Università Di Roma, Rome, Italy
| | - Edda Simoncini
- Medical Oncology Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Claudio Zamagni
- Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano (PN), Italy
| |
Collapse
|
12
|
Wei Q, Li P, Yang T, Zhu J, Sun L, Zhang Z, Wang L, Tian X, Chen J, Hu C, Xue J, Ma L, Shimura T, Fang J, Ying J, Guo P, Cheng X. The promise and challenges of combination therapies with antibody-drug conjugates in solid tumors. J Hematol Oncol 2024; 17:1. [PMID: 38178200 PMCID: PMC10768262 DOI: 10.1186/s13045-023-01509-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/06/2023] [Indexed: 01/06/2024] Open
Abstract
Antibody-drug conjugates (ADCs) represent an important class of cancer therapies that have revolutionized the treatment paradigm of solid tumors. To date, many ongoing studies of ADC combinations with a variety of anticancer drugs, encompassing chemotherapy, molecularly targeted agents, and immunotherapy, are being rigorously conducted in both preclinical studies and clinical trial settings. Nevertheless, combination therapy does not always guarantee a synergistic or additive effect and may entail overlapping toxicity risks. Therefore, understanding the current status and underlying mechanisms of ADC combination therapy is urgently required. This comprehensive review analyzes existing evidence concerning the additive or synergistic effect of ADCs with other classes of oncology medicines. Here, we discuss the biological mechanisms of different ADC combination therapy strategies, provide prominent examples, and assess their benefits and challenges. Finally, we discuss future opportunities for ADC combination therapy in clinical practice.
Collapse
Affiliation(s)
- Qing Wei
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
| | - Peijing Li
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Teng Yang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jiayu Zhu
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Sun
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ziwen Zhang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Lu Wang
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xuefei Tian
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, China
- College of Molecular Medicine, Hangzhou Institute for Advanced Study (HIAS), University of Chinese Academy of Sciences, Hangzhou, China
| | - Jiahui Chen
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Can Hu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Junli Xue
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Letao Ma
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Jieer Ying
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China.
| | - Peng Guo
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China.
| | - Xiangdong Cheng
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China.
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, China.
| |
Collapse
|
13
|
Filis P, Zerdes I, Soumala T, Matikas A, Foukakis T. The ever-expanding landscape of antibody-drug conjugates (ADCs) in solid tumors: A systematic review. Crit Rev Oncol Hematol 2023; 192:104189. [PMID: 37866413 DOI: 10.1016/j.critrevonc.2023.104189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/29/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND The advent of targeted therapies signaled novel avenues for more optimal oncological outcomes. Antibody-drug conjugates (ADCs) have risen as a cornerstone of the ever-expanding targeted therapy era. The purpose of this systematic review is to delineate the rapidly evolving clinical landscape of ADCs for solid tumors. METHODS A literature search was performed in Medline, Embase and Cochrane databases for phase II and III clinical trials. Outcomes of interest were the objective response rate, overall survival, progression-free survival and adverse events. RESULTS A total of 92 clinical trials (76 phase II and 16 phase III) evaluated the efficacy and safety of ADCs for a plethora of solid tumors. Out of the 30 investigated ADCs, 8 have received approval by regulatory organizations for solid tumors. Currently, 52 phase III clinical trials for ADCs are ongoing. CONCLUSION ADCs have shown promising results for several solid tumors and various cancer settings.
Collapse
Affiliation(s)
- Panagiotis Filis
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece.
| | - Ioannis Zerdes
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Theodora Soumala
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Alexios Matikas
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
14
|
Wong RSJ, Ong RJM, Lim JSJ. Immune checkpoint inhibitors in breast cancer: development, mechanisms of resistance and potential management strategies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:768-787. [PMID: 38263984 PMCID: PMC10804393 DOI: 10.20517/cdr.2023.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/14/2023] [Accepted: 10/31/2023] [Indexed: 01/25/2024]
Abstract
The use of immune checkpoint inhibitors (ICIs) has increased exponentially in the past decade, although its progress specifically for breast cancer has been modest. The first U.S. Food and Drug Administration approval for ICI in breast cancer came in 2019, eight years after the first-ever approval of an ICI. At present, current indications for ICIs are relevant only to a subset of patients with triple-negative breast cancer, or those displaying high microsatellite instability or deficiency in the mismatch repair protein pathway. With an increasing understanding of the limitations of using ICIs, which stem from breast cancer being innately poorly immunogenic, as well as the presence of various intrinsic and acquired resistance pathways, ongoing trials are evaluating different combination therapies to overcome these barriers. In this review, we aim to describe the development timeline of ICIs and resistance mechanisms limiting their utility, and summarise the available approaches and ongoing trials relevant to overcoming each resistance mechanism.
Collapse
Affiliation(s)
- Rachel SJ Wong
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119228, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Rebecca JM Ong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Joline SJ Lim
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119228, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|
15
|
Sasso J, Tenchov R, Bird R, Iyer KA, Ralhan K, Rodriguez Y, Zhou QA. The Evolving Landscape of Antibody-Drug Conjugates: In Depth Analysis of Recent Research Progress. Bioconjug Chem 2023; 34:1951-2000. [PMID: 37821099 PMCID: PMC10655051 DOI: 10.1021/acs.bioconjchem.3c00374] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Antibody-drug conjugates (ADCs) are targeted immunoconjugate constructs that integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity. Their design allows for higher doses of the cytotoxic drug to be administered, potentially increasing efficacy. They are currently among the most promising drug classes in oncology, with efforts to expand their application for nononcological indications and in combination therapies. Here we provide a detailed overview of the recent advances in ADC research and consider future directions and challenges in promoting this promising platform to widespread therapeutic use. We examine data from the CAS Content Collection, the largest human-curated collection of published scientific information, and analyze the publication landscape of recent research to reveal the exploration trends in published documents and to provide insights into the scientific advances in the area. We also discuss the evolution of the key concepts in the field, the major technologies, and their development pipelines with company research focuses, disease targets, development stages, and publication and investment trends. A comprehensive concept map has been created based on the documents in the CAS Content Collection. We hope that this report can serve as a useful resource for understanding the current state of knowledge in the field of ADCs and the remaining challenges to fulfill their potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
16
|
Lu L, Niu Z, Chao Z, Fu C, Chen K, Shi Y. Exploring the therapeutic potential of ADC combination for triple-negative breast cancer. Cell Mol Life Sci 2023; 80:350. [PMID: 37930428 PMCID: PMC11073441 DOI: 10.1007/s00018-023-04946-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 11/07/2023]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer. Currently, standard treatment options for TNBC are limited to surgery, adjuvant chemotherapy, and radiotherapy. However, these treatment methods are associated with a higher risk of intrinsic or acquired recurrence. Antibody-drug conjugates (ADCs) have emerged as a useful and promising class of cancer therapeutics. ADCs, also known as "biochemical missiles", use a monoclonal antibody (mAb) to target tumor antigens and deliver a cytotoxic drug payload. Currently, several ADCs clinical studies are underway worldwide, including sacituzumab govitecan (SG), which was recently approved by the FDA for the treatment of TNBC. However, due to the fact that only a small portion of TNBC patients respond to ADC therapy and often develop resistance, growing evidence supports the use of ADCs in combination with other treatment strategies to treat TNBC. In this review, we described the current utilization of ADCs and discussed the prospects of ADC combination therapy for TNBC.
Collapse
Affiliation(s)
- Linlin Lu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Zihe Niu
- Department of Soochow University School of Medicine, Soochow University Suzhou, Suzhou, 215000, China
| | - Zhujun Chao
- Department of Soochow University School of Medicine, Soochow University Suzhou, Suzhou, 215000, China
| | - Cuiping Fu
- Department of Respiratory, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, 215006, Jiangsu, China.
| | - Yaqin Shi
- Department of Oncology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
17
|
Criscitiello C, Corti C, De Laurentiis M, Bianchini G, Pistilli B, Cinieri S, Castellan L, Arpino G, Conte P, Di Meco F, Gennari A, Guarneri V, Visani L, Livi L, Marchetti P, Puglisi F, Viale G, Del Mastro L, De Placido S, Curigliano G. Tucatinib's journey from clinical development to clinical practice: New horizons for HER2-positive metastatic disease and promising prospects for brain metastatic spread. Cancer Treat Rev 2023; 120:102618. [PMID: 37639757 DOI: 10.1016/j.ctrv.2023.102618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Approximately 20% of breast cancers (BCs) overexpress human epidermal growth factor receptor 2 (HER2), a transmembrane glycoprotein with tyrosine kinase activity, encoded by ERBB2 gene. Historically, HER2 overexpression has been linked with increased disease recurrence and a worse prognosis. However, the increasing availability of different anti-HER2 compounds and combinations is progressively improving HER2-positive BC outcome, thus requiring expertise to prioritize both overall survival (OS) prolongation and quality of life, without neglecting the accessibility to further treatment lines with a low attrition rate. In this context, tucatinib, an oral tyrosine kinase inhibitor, has recently been granted approval by regulatory agencies based on evidence from the HER2CLIMB, a clinical trial which randomized patients with metastatic BC to receive trastuzumab and capecitabine with either tucatinib or placebo. A distinctive feature of this study was the inclusion of patients with new or active brain metastases (BMs) at study entry, a population traditionally excluded from clinical trials. Thus, HER2CLIMB provides the first solid evidence of an OS benefit in patients with BC and BMs, addressing a long standing unmet medical need, especially given the high incidence of central nervous system metastatic spread in patients with HER2-positive disease. This review provides an overview of the molecular and clinical landscape of tucatinib for the treatment of advanced BC. It focuses on the technological journey that drove the development of this therapeutic innovation, from preclinical data to clinical practice.
Collapse
Affiliation(s)
- Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy.
| | - Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Michelino De Laurentiis
- Department of Breast and Thoracic Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione Pascale', Napoli, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, Ospedale San Raffaele, IRCCS, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Barbara Pistilli
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Saverio Cinieri
- Medical Oncology Unit, Ospedale di Summa A. Perrino, Brindisi, Italy
| | - Lucio Castellan
- Department of Diagnostic and Interventional Neuroradiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Pierfranco Conte
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | | | - Alessandra Gennari
- Medical Oncology, "Maggiore della Carità" University Hospital, Novara, Italy; Department of Translational Medicine (DIMET), University of Eastern Piedmont (UPO), Novara, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Luca Visani
- Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of Florence, Florence, Italy; Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Lorenzo Livi
- Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of Florence, Florence, Italy; Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabio Puglisi
- Department of Medical Oncology, National Cancer Institute, IRCCS, Aviano, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - Giuseppe Viale
- Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy; Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Lucia Del Mastro
- Department of Medical Oncology, Breast Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, Genoa, Italy
| | - Sabino De Placido
- Department of Translational Medicine (DIMET), University of Eastern Piedmont (UPO), Novara, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
18
|
Roy AM, Kumarasamy VM, Dhakal A, O’Regan R, Gandhi S. A review of treatment options in HER2-low breast cancer and proposed treatment sequencing algorithm. Cancer 2023; 129:2773-2788. [PMID: 37349954 PMCID: PMC10478358 DOI: 10.1002/cncr.34904] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/07/2023] [Accepted: 05/15/2023] [Indexed: 06/24/2023]
Abstract
The expansion of the spectrum of human epidermal growth factor receptor 2 (HER2)-status to HER2-low, defined as HER2 expression of 1+ by immunohistochemistry (IHC) or 2+ by IHC without gene amplification, has made a major impact in the field of oncology. The HER2-low expression has emerged as a targetable biomarker, and anti-HER2 antibody-drug conjugate trastuzumab deruxtecan has shown significant survival benefit in pretreated metastatic HER2-low breast cancer (BC). With these recent data, the treatment algorithm for hormone receptor-positive and triple-negative BC needs to be reconsidered, as approximately half of these BCs are HER2-low. Although there are different therapeutic agents for hormone receptor-positive and hormone receptor-negative HER2-low BCs, there is no consensus regarding the sequencing of these agents. In this article, the treatment options for HER2-low BC are enumerated and a treatment sequencing algorithm based on the current clinical evidence proposed.
Collapse
Affiliation(s)
- Arya Mariam Roy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, 14203
| | | | - Ajay Dhakal
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, 14642
| | - Ruth O’Regan
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, 14642
| | - Shipra Gandhi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, 14203
| |
Collapse
|
19
|
Chang HL, Schwettmann B, McArthur HL, Chan IS. Antibody-drug conjugates in breast cancer: overcoming resistance and boosting immune response. J Clin Invest 2023; 133:e172156. [PMID: 37712425 PMCID: PMC10503805 DOI: 10.1172/jci172156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
Antibody-drug conjugates (ADCs) have emerged as a revolutionary therapeutic class, combining the precise targeting ability of monoclonal antibodies with the potent cytotoxic effects of chemotherapeutics. Notably, ADCs have rapidly advanced in the field of breast cancer treatment. This innovative approach holds promise for strengthening the immune system through antibody-mediated cellular toxicity, tumor-specific immunity, and adaptive immune responses. However, the development of upfront and acquired resistance poses substantial challenges in maximizing the effectiveness of these therapeutics, necessitating a deeper understanding of the underlying mechanisms. These mechanisms of resistance include antigen loss, derangements in ADC internalization and recycling, drug clearance, and alterations in signaling pathways and the payload target. To overcome resistance, ongoing research and development efforts are focused on urgently identifying biomarkers, integrating immune therapy approaches, and designing novel cytotoxic payloads. This Review provides an overview of the mechanisms and clinical effectiveness of ADCs, and explores their unique immune-boosting function, while also highlighting the complex resistance mechanisms and safety challenges that must be addressed. A continued focus on how ADCs impact the tumor microenvironment will help to identify new payloads that can improve patient outcomes.
Collapse
Affiliation(s)
- Hannah L. Chang
- Department of Internal Medicine, Division of Hematology and Oncology
- Harold C. Simmons Comprehensive Cancer Center, and
| | - Blake Schwettmann
- Department of Internal Medicine, Division of Hematology and Oncology
- Harold C. Simmons Comprehensive Cancer Center, and
| | - Heather L. McArthur
- Department of Internal Medicine, Division of Hematology and Oncology
- Harold C. Simmons Comprehensive Cancer Center, and
| | - Isaac S. Chan
- Department of Internal Medicine, Division of Hematology and Oncology
- Harold C. Simmons Comprehensive Cancer Center, and
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
20
|
Fabi A, Carbognin L, Botticelli A, Paris I, Fuso P, Savastano MC, La Verde N, Strina C, Pedersini R, Guarino S, Curigliano G, Criscitiello C, Raffaele M, Beano A, Franco A, Valerio MR, Verderame F, Fontana A, Haspinger ER, Caldara A, Di Leone A, Tortora G, Giannarelli D, Scambia G. Real-world ANASTASE study of atezolizumab+nab-paclitaxel as first-line treatment of PD-L1-positive metastatic triple-negative breast cancer. NPJ Breast Cancer 2023; 9:73. [PMID: 37684252 PMCID: PMC10491680 DOI: 10.1038/s41523-023-00579-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The combination of atezolizumab and nab-paclitaxel is recommended in the EU as first-line treatment for PD-L1-positive metastatic triple-negative breast cancer (mTNBC), based on the results of phase III IMpassion130 trial. However, 'real-world' data on this combination are limited. The ANASTASE study (NCT05609903) collected data on atezolizumab plus nab-paclitaxel in PD-L1-positive mTNBC patients enrolled in the Italian Compassionate Use Program. A retrospective analysis was conducted in 29 Italian oncology centers among patients who completed at least one cycle of treatment. Data from 52 patients were gathered. Among them, 21.1% presented de novo stage IV; 78.8% previously received (neo)adjuvant treatment; 55.8% patients had only one site of metastasis; median number of treatment cycles was five (IQR: 3-8); objective response rate was 42.3% (95% CI: 28.9-55.7%). The median time-to-treatment discontinuation was 5 months (95% CI: 2.8-7.1); clinical benefit at 12 months was 45.8%. The median duration of response was 12.7 months (95% CI: 4.1-21.4). At a median follow-up of 20 months, the median progression-free survival was 6.3 months (95% CI: 3.9-8.7) and the median time to next treatment or death was 8.1 months (95% CI: 5.5-10.7). At 12 months and 24 months, the overall survival rates were 66.3% and 49.1%, respectively. The most common immune-related adverse events included rash (23.1%), hepatitis (11.5%), thyroiditis (11.5%) and pneumonia (9.6%). Within the ANASTASE study, patients with PD-L1-positive mTNBC treated with first-line atezolizumab plus nab-paclitaxel achieved PFS and ORR similar to those reported in the IMpassion130 study, with no unexpected adverse events.
Collapse
Affiliation(s)
- Alessandra Fabi
- Precision Medicine Unit in Senology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Luisa Carbognin
- Division of Gynecology Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Botticelli
- Medical Oncology Unit, La Sapienza, University of Rome, Policlinico Umberto I, Rome, Italy
| | - Ida Paris
- Division of Gynecology Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paola Fuso
- Division of Gynecology Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Nicla La Verde
- Medical Oncology Unit, ASST Fatebenefratelli Sacco PO Luigi Sacco - Polo Universitario, Milan, Italy
| | - Carla Strina
- Medical Oncology Unit Azienda Socio-Sanitaria Territoriale Cremona, Cremona, Italy
| | | | - Stefania Guarino
- Medical Oncology Unit Santa Maria della Misericordia Hospital, Urbino, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
| | - Mimma Raffaele
- Presidio Cassia Sant'andrea, Dipartimento Oncologico, Asl Roma1, Rome, Italy
| | - Alessandra Beano
- Department of Medical Oncology1, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Antonio Franco
- Breast Unit, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | | | - Andrea Fontana
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | | | | | - Alba Di Leone
- Breast Unit, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Medical Oncology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Diana Giannarelli
- Epidemiology and Biostatistics Facility, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Giovanni Scambia
- Division of Gynecology Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
21
|
Mark C, Lee JS, Cui X, Yuan Y. Antibody-Drug Conjugates in Breast Cancer: Current Status and Future Directions. Int J Mol Sci 2023; 24:13726. [PMID: 37762027 PMCID: PMC10531043 DOI: 10.3390/ijms241813726] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Antibody drug conjugates (ADCs) are novel medications that combine monoclonal antibodies with cytotoxic payloads, enabling the selective delivery of potent drugs to cancer cells expressing specific surface antigens. This targeted strategy seeks to optimize treatment effectiveness while reducing the risk of systemic toxicity, distinguishing ADCs from conventional chemotherapy. The rapid growth in ADC research has led to numerous developments and approvals for cancer treatment, with significant impacts on the management of breast cancer. ADCs like T-DXd for HER2-low disease and sacituzumab govitecan for triple negative breast cancer (TNBC) have provided valuable options for challenging subtypes of breast cancer. However, essential questions still need to be addressed, including the optimal order of ADCs amidst the growing number of newly developed ones and strategies to overcome resistance mechanisms. Preclinical studies have shed light on potential resistance mechanisms, emphasizing the potential benefit of combinational approaches with other agents such as immune checkpoint inhibitors (ICIs) and targeted tyrosine kinase inhibitors (TKIs) to enhance treatment effectiveness. Additionally, personalized approaches based on molecular profiling hold promise in tailoring ADC treatments to individual tumors, identifying unique molecular markers for each patient to optimize treatment efficacy while minimizing side effects.
Collapse
Affiliation(s)
- Cynthia Mark
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jin Sun Lee
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xiaojiang Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yuan Yuan
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
22
|
D’Arienzo A, Verrazzo A, Pagliuca M, Napolitano F, Parola S, Viggiani M, Caputo R, Puglisi F, Giuliano M, Del Mastro L, Arpino G, De Laurentiis M, Montemurro F. Toxicity profile of antibody-drug conjugates in breast cancer: practical considerations. EClinicalMedicine 2023; 62:102113. [PMID: 37554126 PMCID: PMC10404866 DOI: 10.1016/j.eclinm.2023.102113] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023] Open
Abstract
Antibody-drug conjugates (ADCs) represent a novel and evolving class of antineoplastic agents, constituted by monoclonal antibody linked to biologically active drugs, delivering cytotoxic compounds at the tumor site, reducing the likelihood of systemic exposure and toxicity. They are generally well tolerated, nevertheless some predictable adverse reactions need careful monitoring and timely approach. These include neutropenia, nausea and vomiting, alopecia, diarrhea, left ventricular dysfunction, ILD/pneumonitis. The mechanisms leading to drug-associated toxicities are summarized, and prophylaxis protocols and appropriate management strategies are proposed, based on current literature. This review aims to collect the most updated evidence on toxicities potentially occurring during breast cancer treatment with approved or under clinical investigation (advanced stage) ADCs. A focus is dedicated to monitoring protocols and clinical management, aimed at preventing and/or promptly address relevant problems, in order to avoid premature discontinuation or improper dose reduction.
Collapse
Affiliation(s)
- Andrea D’Arienzo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Annarita Verrazzo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Via Mezzocannone 4, Naples 80138, Italy
| | - Martina Pagliuca
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Via Mezzocannone 4, Naples 80138, Italy
- Molecular Predictors and New Targets in Oncology Unit 981, Gustave Roussy, 114 Rue Édouard-Vaillant, Villejuif 94805, France
| | - Fabiana Napolitano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Sara Parola
- Oncology Unit, PO di San Felice a Cancello, Via Roma 349, San Felice a Cancello, Caserta 81021, Italy
| | - Martina Viggiani
- Department of Oncology, HFR Fribourg-Cantonal Hospital, Chemin des Pensionnats 2-6, Fribourg 1708, Switzerland
| | - Roberta Caputo
- Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola 52, Naples 80131, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Via Palladio 8, Udine 33100, Italy
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Via Franco Gallini 2, Aviano, Pordenone 33081, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Lucia Del Mastro
- Department of Medical Oncology, UO Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, Genova 16132, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Michelino De Laurentiis
- Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola 52, Naples 80131, Italy
| | - Filippo Montemurro
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 -KM 3.95, Candiolo, Torino 10060, Italy
| |
Collapse
|
23
|
Morrison L, Okines A. Systemic Therapy for Metastatic Triple Negative Breast Cancer: Current Treatments and Future Directions. Cancers (Basel) 2023; 15:3801. [PMID: 37568617 PMCID: PMC10417818 DOI: 10.3390/cancers15153801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Until recently, despite its heterogenous biology, metastatic triple negative breast cancer (TNBC) was treated as a single entity, with successive lines of palliative chemotherapy being the only systemic option. Significant gene expression studies have demonstrated the diversity of TNBC, but effective differential targeting of the four main (Basal-like 1 and 2, mesenchymal and luminal androgen receptor) molecular sub-types has largely eluded researchers. The introduction of immunotherapy, currently useful only for patients with PD-L1 positive cancers, led to the stratification of first-line therapy using this immunohistochemical biomarker. Germline BRCA gene mutations can also be targeted with PARP inhibitors in both the adjuvant and metastatic settings. In contrast, the benefit of the anti-Trop-2 antibody-drug conjugate (ADC) Sacituzumab govitecan (SG) does not appear confined to patients with tumours expressing high levels of Trop-2, leading to its potential utility for any patient with an estrogen receptor (ER)-negative, HER2-negative advanced breast cancer (ABC). Most recently, low levels of HER2 expression, detected in up to 60% of TNBC, predicts benefit from the potent HER2-directed antibody-drug conjugate trastuzumab deruxtecan (T-DXd), defining an additional treatment option for this sub-group. Regrettably, despite recent advances, the median survival of TNBC continues to lag far behind the approximately 5 years now expected for patients with ER-positive or HER2-positive breast cancers. We review the data supporting immunotherapy, ADCs, and targeted agents in subgroups of patients with TNBC, and current clinical trials that may pave the way to further advances in this challenging disease.
Collapse
Affiliation(s)
| | - Alicia Okines
- Breast Unit, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
| |
Collapse
|
24
|
Guven DC, Yildirim HC, Kus F, Erul E, Kertmen N, Dizdar O, Aksoy S. Optimal adjuvant treatment strategies for TNBC patients with residual disease after neoadjuvant treatment. Expert Rev Anticancer Ther 2023; 23:1049-1059. [PMID: 37224429 DOI: 10.1080/14737140.2023.2218090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/22/2023] [Indexed: 05/26/2023]
Abstract
INTRODUCTION The therapeutic armamentarium for the neoadjuvant treatment of triple-negative breast cancer (TNBC) has significantly expanded with the hopes of improving pathological complete response (pCR) rates and the possibility of a cure. However, the data on optimal adjuvant treatment strategies for patients with residual disease after neoadjuvant treatment is limited. AREAS COVERED We discuss the available data on adjuvant treatment for residual TNBC after neoadjuvant treatment considering clinical trials. Additionally, we discuss ongoing trials to give perspectives on how the field may evolve in the next decade. EXPERT OPINION The available data support the use of adjuvant capecitabine for all patients and either adjuvant capecitabine or olaparib for patients with germline BRCA1 and BRCA2 mutations, according to availability. The CREATE-X study of capecitabine and OlympiA study of olaparib demonstrated disease-free and overall survival benefits. There is an unmet need for studies comparing these two options for patients with germline BRCA mutations. Further research is needed to delineate the use of immunotherapy in the adjuvant setting, molecular targeted therapy for patients with molecular alterations other than germline BRCA mutation, combinations, and antibody-drug conjugates to further improve outcomes.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Hasan Cagri Yildirim
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Fatih Kus
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Enes Erul
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Neyran Kertmen
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Omer Dizdar
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
25
|
Tarantino P, Ricciuti B, Pradhan SM, Tolaney SM. Optimizing the safety of antibody-drug conjugates for patients with solid tumours. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00783-w. [PMID: 37296177 DOI: 10.1038/s41571-023-00783-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
Over the past 5 years, improvements in the design of antibody-drug conjugates (ADCs) have enabled major advances that have reshaped the treatment of several advanced-stage solid tumours. Considering the intended rationale behind the design of ADCs, which is to achieve targeted delivery of cytotoxic molecules by linking them to antibodies targeting tumour-specific antigens, ADCs would be expected to be less toxic than conventional chemotherapy. However, most ADCs are still burdened by off-target toxicities that resemble those of the cytotoxic payload as well as on-target toxicities and other poorly understood and potentially life-threatening adverse effects. Given the rapid expansion in the clinical indications of ADCs, including use in curative settings and various combinations, extensive efforts are ongoing to improve their safety. Approaches currently being pursued include clinical trials optimizing the dose and treatment schedule, modifications of each ADC component, identification of predictive biomarkers for toxicities, and the development of innovative diagnostic tools. In this Review, we describe the determinants of the toxicities of ADCs in patients with solid tumours, highlighting key strategies that are expected to improve tolerability and enable improvements in the treatment outcomes of patients with advanced-stage and those with early stage cancers in the years to come.
Collapse
Affiliation(s)
- Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Biagio Ricciuti
- Harvard Medical School, Boston, MA, USA
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shan M Pradhan
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Shastry M, Gupta A, Chandarlapaty S, Young M, Powles T, Hamilton E. Rise of Antibody-Drug Conjugates: The Present and Future. Am Soc Clin Oncol Educ Book 2023; 43:e390094. [PMID: 37229614 DOI: 10.1200/edbk_390094] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Antibody-drug conjugates (ADCs) embody a simple, but elegant, vision for cancer therapy-the delivery of a potent cytotoxic agent to tumor cells with minimal damage to normal cells-so-called smart chemo. Although there were significant challenges in achieving this milestone culminating in the first Food and Drug Administration approval in 2000, subsequent advancements in technology have led to rapid drug development with regulatory approvals for ADCs targeting a variety of tumor types. The most successful application for solid tumors has been in breast cancer, with ADCs becoming the standard of care across traditional human epidermal growth factor receptor 2 (HER2)+, hormone receptor+ (HR+) and triple-negative disease subtypes. Moreover, the improved features and gains in potency with the development of ADCs have expanded the treatment-eligible population to those with low/heterogeneous expression of the target antigen on the tumor with trastuzumab deruxtecan or in the case of sacituzumab govitecan, agnostic to target expression. Despite their antibody-directed homing, these novel agents come with their share of toxicities obligating appropriate patient selection and vigilant monitoring while on treatment. As more ADCs are included in the treatment armamentarium, mechanisms of resistance need to be studied and understood for optimal sequencing. Modifying the payload to use immune-stimulating agents or combination therapies with immunotherapy and other effective targeted therapies may further extend the utility of these agents in the treatment of solid tumors.
Collapse
Affiliation(s)
| | - Avantika Gupta
- Department of Medicine, Human Oncology and Pathogenesis Program, MSKCC, New York, NY
| | - Sarat Chandarlapaty
- Department of Medicine, Human Oncology and Pathogenesis Program, MSKCC, New York, NY
| | - Matthew Young
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Thomas Powles
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Erika Hamilton
- Sarah Cannon Research Institute, Nashville, TN
- Tennessee Oncology, Nashville, TN
| |
Collapse
|
27
|
Passalacqua MI, Rizzo G, Santarpia M, Curigliano G. 'Why is survival with triple negative breast cancer so low? insights and talking points from preclinical and clinical research'. Expert Opin Investig Drugs 2022; 31:1291-1310. [PMID: 36522800 DOI: 10.1080/13543784.2022.2159805] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Triple negative breast cancer is typically related to poor prognosis, early metastasis, and high recurrence rate. Intrinsic and extrinsic biological features of TNBC and resistance mechanisms to conventional therapies can support its aggressive behavior, characterizing TNBC how extremely heterogeneous. Novel combination strategies are under investigation, including immunotherapeutic agents, anti-drug conjugates, PARP inhibitors, and various targeting agents, exploring, in the meanwhile, possible predictive biomarkers to correctly select patients for the optimal treatment for their specific subtype. AREAS COVERED This article examines the main malignity characteristics across different subtype, both histological and molecular, and the resistance mechanisms, both primary and acquired, to different drugs explored in the landscape of TNBC treatment, that lead TNBC to still has high mortality rate. EXPERT OPINION The complexity of TNBC is not only the main reason of its aggressivity, but its heterogeneity should be exploited in terms of therapeutics opportunities, combining agents with different mechanism of action, after a correct selection by biologic or molecular biomarkers. The main goal is to understand what TNBC really is and to act selectively on its characteristics, with a personalized anticancer treatment.
Collapse
Affiliation(s)
- Maria Ilenia Passalacqua
- Division of Early Drug Development for Innovative Therapies, Ieo, European Institute of Oncology Irccs, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy.,Medical Oncology Unit, Department of Human Pathology G Barresi, University of Messina, Messina, Italy
| | - Graziella Rizzo
- Division of Early Drug Development for Innovative Therapies, Ieo, European Institute of Oncology Irccs, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy.,Medical Oncology Unit, Department of Human Pathology G Barresi, University of Messina, Messina, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology G Barresi, University of Messina, Messina, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, Ieo, European Institute of Oncology Irccs, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| |
Collapse
|
28
|
Massa D, Tosi A, Rosato A, Guarneri V, Dieci MV. Multiplexed In Situ Spatial Protein Profiling in the Pursuit of Precision Immuno-Oncology for Patients with Breast Cancer. Cancers (Basel) 2022; 14:4885. [PMID: 36230808 PMCID: PMC9562913 DOI: 10.3390/cancers14194885] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of many solid tumors. In breast cancer (BC), immunotherapy is currently approved in combination with chemotherapy, albeit only in triple-negative breast cancer. Unfortunately, most patients only derive limited benefit from ICIs, progressing either upfront or after an initial response. Therapeutics must engage with a heterogeneous network of complex stromal-cancer interactions that can fail at imposing cancer immune control in multiple domains, such as in the genomic, epigenomic, transcriptomic, proteomic, and metabolomic domains. To overcome these types of heterogeneous resistance phenotypes, several combinatorial strategies are underway. Still, they can be predicted to be effective only in the subgroups of patients in which those specific resistance mechanisms are effectively in place. As single biomarker predictive performances are necessarily suboptimal at capturing the complexity of this articulate network, precision immune-oncology calls for multi-omics tumor microenvironment profiling in order to identify unique predictive patterns and to proactively tailor combinatorial treatments. Multiplexed single-cell spatially resolved tissue analysis, through precise epitope colocalization, allows one to infer cellular functional states in view of their spatial organization. In this review, we discuss-through the lens of the cancer-immunity cycle-selected, established, and emerging markers that may be evaluated in multiplexed spatial protein panels to help identify prognostic and predictive patterns in BC.
Collapse
Affiliation(s)
- Davide Massa
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Anna Tosi
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
| |
Collapse
|
29
|
Drago JZ, Ferraro E, Abuhadra N, Modi S. Beyond HER2: Targeting the ErbB receptor family in breast cancer. Cancer Treat Rev 2022; 109:102436. [PMID: 35870237 PMCID: PMC10478787 DOI: 10.1016/j.ctrv.2022.102436] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/28/2022]
Abstract
Targeting the HER2 oncogene represents one of the greatest advances in the treatment of breast cancer. HER2 is one member of the ERBB-receptor family, which includes EGFR (HER1), HER3 and HER4. In the presence or absence of underling genomic aberrations such as mutations or amplification events, intricate interactions between these proteins on the cell membrane lead to downstream signaling that encourages cancer growth and proliferation. In this Review, we contextualize efforts to pharmacologically target the ErbB receptor family beyond HER2, with a focus on EGFR and HER3. Preclinical and clinical efforts are synthesized. We discuss successes and failures of this approach to date, summarize lessons learned, and propose a way forward that invokes new therapeutic modalities such as antibody drug conjugates (ADCs), combination strategies, and patient selection through rational biomarkers.
Collapse
Affiliation(s)
- Joshua Z Drago
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA.
| | - Emanuela Ferraro
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nour Abuhadra
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA
| |
Collapse
|
30
|
朱 以, 王 志. [Current Progress and Future Developments of Antibody Drug Conjugates
in Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:468-476. [PMID: 35899443 PMCID: PMC9346152 DOI: 10.3779/j.issn.1009-3419.2022.102.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022]
Abstract
Antibody drug conjugates (ADCs) are a novel class of anti-cancer drugs, which combined the specificity of monoclonal antibodies with the cytotoxic palyload via the linkers. Many ADCs have not only verified impressive activity in a variety of cancers, including breast cancer and hematological system tumors, but also in lung cancer. The aim of this study was to provide informations for practice by summarizing the mechanism of action, clinical application and problems and challenges of ADCs.
.
Collapse
Affiliation(s)
- 以香 朱
- />100021 北京,国家癌症中心,国家肿瘤临床医学研究中心,中国医学科学院北京协和医学院肿瘤医院内科CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
| | - 志杰 王
- />100021 北京,国家癌症中心,国家肿瘤临床医学研究中心,中国医学科学院北京协和医学院肿瘤医院内科CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing 100021, China
| |
Collapse
|