1
|
Corti C, Rugo HS, Tolaney SM. Decoding Clinical Trials in Metastatic Breast Cancer: Practical Insights for Optimal Therapy Sequencing. Am Soc Clin Oncol Educ Book 2025; 45:e100053. [PMID: 40367401 DOI: 10.1200/edbk-25-100053] [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: 05/16/2025]
Abstract
The art of sequencing therapy in the management of breast cancer is a multifaceted challenge that demands the careful integration of clinical trial data, real-world evidence, and individualized patient factors to guide treatment decisions. As the therapeutic landscape evolves rapidly with new agents and combinations, clinicians are confronted with critical decisions on how best to order treatments to maximize benefit, minimize toxicity, and preserve future options. For patients with estrogen receptor-positive (ER+) disease, this review discusses how emerging resistance patterns after cyclin-dependent kinase 4 and 6 inhibitors require careful re-evaluation of subsequent endocrine and targeted therapies, as well as chemotherapy, emphasizing the need for evidence-based strategies and ethical crossover designs in clinical trials. In addition, for both ER+ and ER- metastatic breast cancer (MBC) with nonoverexpressed human epidermal growth factor receptor 2 (HER2), this review highlights pivotal trials investigating antibody-drug conjugates (ADCs)-including trastuzumab deruxtecan, sacituzumab govitecan, and datopotamab deruxtecan-and the challenges related to control arm selection and crossover that may affect outcome interpretation. Finally, for patients with HER2-positive disease, the review explores first-line and maintenance strategies-including insights from landmark trials like CLEOPATRA and PATINA-and addresses the impact of brain metastases on sequencing decisions. By critically appraising current data and identifying gaps in biomarker-guided and sequencing-specific strategies, this review provides practical insights to inform clinical practice and optimize personalized treatment plans for patients with MBC.
Collapse
Affiliation(s)
- Chiara Corti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
2
|
Gadaleta-Caldarola G, Lanotte L, Santoro AN, Pinto A, Gadaleta-Caldarola A, Giacomelli L, Fedele P. Predictive Factors of Antibody-Drug Conjugate Treatment in Metastatic Breast Cancer: A Narrative Review. Cancers (Basel) 2024; 16:4082. [PMID: 39682268 DOI: 10.3390/cancers16234082] [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: 10/21/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have revolutionized the treatment landscape for metastatic breast cancer, offering targeted delivery of cytotoxic agents with improved efficacy and tolerability compared to conventional chemotherapy. This narrative review explores key predictive factors influencing the efficacy of ADCs, focusing on HER2-targeted therapies, such as trastuzumab emtansine and trastuzumab deruxtecan, as well as sacituzumab govitecan for triple-negative breast cancer. HER2 expression, TROP-2 levels, hormone receptor status, and the tumor microenvironment emerge as critical biomarkers for patient selection and therapeutic outcomes. Additionally, we discuss resistance mechanisms, such as antigen loss, impaired drug internalization, and the role of circulating tumor DNA in predicting ADC response. Finally, future perspectives on the sequential use of ADCs and potential combination therapies are highlighted, along with emerging agents targeting alternative antigens like HER3 and LIV-1. Overall, identifying predictive biomarkers and overcoming resistance mechanisms are essential for optimizing the use of ADCs in metastatic breast cancer, thereby improving patient outcomes.
Collapse
Affiliation(s)
| | - Laura Lanotte
- Oncology Unit, "Mons. A. R. Dimiccoli" Hospital, 70051 Barletta, Italy
| | | | - Antonello Pinto
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| | | | | | - Palma Fedele
- Oncology Unit, "Dario Camberlingo" Hospital, 72021 Francavilla Fontana, Italy
| |
Collapse
|
3
|
Zouein J, Karam E, Strickler JH, Kourie HR. Trastuzumab-Deruxtecan: Redefining HER2 as a Tumor Agnostic Biomarker. Target Oncol 2024; 19:705-710. [PMID: 38963654 DOI: 10.1007/s11523-024-01079-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Trastuzumab deruxtecan (T-DXd) is an antibody-drug conjugate (ADC) targeting HER2-positive malignancies across various tumor types. Through its unique composition, T-DXd achieves selective payload delivery, inducing cell death and halting tumor progression. Clinical trials initially investigated T-DXd's efficacy in HER2-positive advanced or metastatic breast, gastric, lung, and colorectal cancers; however, recent results from the DESTINY-PanTumor02 trial further underscore T-DXd's versatility, prompting T-DXd's US FDA approval for HER2-positive (immunohistochemistry [IHC] 3+) solid tumors. Moreover, in addition to T-DXd's efficacy against brain metastasis, T-DXd is showing promising results in HER2-low and HER2-ultra-low metastatic breast cancer, indicating a broader population of patients who may benefit.
Collapse
Affiliation(s)
- Joseph Zouein
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.
- Hematology-Oncology Department, Faculty of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon.
| | - Elias Karam
- Départements de Médecine Oncologique, Gustave Roussy, 94805, Villejuif, France
- Hematology-Oncology Department, Faculty of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - John H Strickler
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Hampig Raphael Kourie
- Hematology-Oncology Department, Faculty of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| |
Collapse
|
4
|
Theocharopoulos C, Ziogas IA, Douligeris CC, Efstathiou A, Kolorizos E, Ziogas DC, Kontis E. Antibody-drug conjugates for hepato-pancreato-biliary malignancies: "Magic bullets" to the rescue? Cancer Treat Rev 2024; 129:102806. [PMID: 39094332 DOI: 10.1016/j.ctrv.2024.102806] [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: 05/19/2024] [Revised: 07/17/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Hepato-Pancreato-Biliary (HPB) malignancies constitute a highly aggressive group of cancers that have a dismal prognosis. Patients not amenable to curative intent surgical resection are managed with systemic chemotherapy which, however, confers little survival benefit. Antibody-Drug Conjugates (ADCs) are tripartite compounds that merge the intricate selectivity and specificity of monoclonal antibodies with the cytodestructive potency of attached supertoxic payloads. In view of the unmet need for drugs that will enhance the survival rates of HPB cancer patients, the assessment of ADCs for treating HPB malignancies has become the focus of extensive clinical and preclinical investigation, showing encouraging preliminary results. In the current review, we offer a comprehensive overview of the growing body of evidence on ADC approaches tested for HPB malignancies. Starting from a concise discussion of the functional principles of ADCs, we summarize here all available data from preclinical and clinical studies evaluating ADCs in HPB cancers.
Collapse
Affiliation(s)
| | - Ioannis A Ziogas
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | | | | | | | - Dimitrios C Ziogas
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens 11527, Greece
| | - Elissaios Kontis
- Department of Surgery, Metaxa Cancer Hospital, Piraeus 18537, Greece
| |
Collapse
|
5
|
Klümper N, Tran NK, Zschäbitz S, Hahn O, Büttner T, Roghmann F, Bolenz C, Zengerling F, Schwab C, Nagy D, Toma M, Kristiansen G, Heers H, Ivanyi P, Niegisch G, Grunewald CM, Darr C, Farid A, Schlack K, Abbas M, Aydogdu C, Casuscelli J, Mokry T, Mayr M, Niedersüß-Beke D, Rausch S, Dietrich D, Saal J, Ellinger J, Ritter M, Alajati A, Kuppe C, Meeks J, Vera Badillo FE, Nakauma-González JA, Boormans J, Junker K, Hartmann A, Grünwald V, Hölzel M, Eckstein M. NECTIN4 Amplification Is Frequent in Solid Tumors and Predicts Enfortumab Vedotin Response in Metastatic Urothelial Cancer. J Clin Oncol 2024; 42:2446-2455. [PMID: 38657187 PMCID: PMC11227306 DOI: 10.1200/jco.23.01983] [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: 09/12/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 04/26/2024] Open
Abstract
PURPOSE The anti-NECTIN4 antibody-drug conjugate enfortumab vedotin (EV) is approved for patients with metastatic urothelial cancer (mUC). However, durable benefit is only achieved in a small, yet uncharacterized patient subset. NECTIN4 is located on chromosome 1q23.3, and 1q23.3 gains represent frequent copy number variations (CNVs) in urothelial cancer. Here, we aimed to evaluate NECTIN4 amplifications as a genomic biomarker to predict EV response in patients with mUC. MATERIALS AND METHODS We established a NECTIN4-specific fluorescence in situ hybridization (FISH) assay to assess the predictive value of NECTIN4 CNVs in a multicenter EV-treated mUC patient cohort (mUC-EV, n = 108). CNVs were correlated with membranous NECTIN4 protein expression, EV treatment responses, and outcomes. We also assessed the prognostic value of NECTIN4 CNVs measured in metastatic biopsies of non-EV-treated mUC (mUC-non-EV, n = 103). Furthermore, we queried The Cancer Genome Atlas (TCGA) data sets (10,712 patients across 32 cancer types) for NECTIN4 CNVs. RESULTS NECTIN4 amplifications are frequent genomic events in muscle-invasive bladder cancer (TCGA bladder cancer data set: approximately 17%) and mUC (approximately 26% in our mUC cohorts). In mUC-EV, NECTIN4 amplification represents a stable genomic alteration during metastatic progression and associates with enhanced membranous NECTIN4 protein expression. Ninety-six percent (27 of 28) of patients with NECTIN4 amplifications demonstrated objective responses to EV compared with 32% (24 of 74) in the nonamplified subgroup (P < .001). In multivariable Cox analysis adjusted for age, sex, and Bellmunt risk factors, NECTIN4 amplifications led to a 92% risk reduction for death (hazard ratio, 0.08 [95% CI, 0.02 to 0.34]; P < .001). In the mUC-non-EV, NECTIN4 amplifications were not associated with outcomes. TCGA Pan-Cancer analysis demonstrated that NECTIN4 amplifications occur frequently in other cancers, for example, in 5%-10% of breast and lung cancers. CONCLUSION NECTIN4 amplifications are genomic predictors of EV responses and long-term survival in patients with mUC.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
| | - Ngoc Khanh Tran
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Stefanie Zschäbitz
- Department of Medical Oncology, National Center for Tumor Disease (NCT), University Hospital, Heidelberg, Germany
| | - Oliver Hahn
- Department of Urology and Pediatric Urology, Julius Maximilians University Medical Center of Würzburg, Würzburg, Germany
| | - Thomas Büttner
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Florian Roghmann
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Department of Urology, Marien Hospital, Ruhr-University Bochum, Herne, Germany
| | - Christian Bolenz
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Department of Urology and Pediatric Urology, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Friedemann Zengerling
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Department of Urology and Pediatric Urology, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Constantin Schwab
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Dora Nagy
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Marieta Toma
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Glen Kristiansen
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Hendrik Heers
- Department of Urology, University Hospital Marburg, Marburg, Germany
| | - Philipp Ivanyi
- Department of Hemostaseology, Oncology and Stem Cell Transplantation, Medical University Hannover, Hannover, Germany
| | - Günter Niegisch
- Department of Urology, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Christopher Darr
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Arian Farid
- Department of Urology, University Medical Center Göttingen, Göttingen, Germany
| | - Katrin Schlack
- Department of Urology, University Hospital Münster, Münster, Germany
| | - Mahmoud Abbas
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Can Aydogdu
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jozefina Casuscelli
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Theresa Mokry
- Department of Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Mayr
- Clinic Ottakring, Institute of Pathology and Microbiology, Wien, Austria
| | | | - Steffen Rausch
- Department of Urology, Eberhard Karls University, Tübingen, Germany
| | - Dimo Dietrich
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Bonn, Germany
| | - Jonas Saal
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- Medical Clinic III for Oncology, Hematology, Immune-Oncology and Rheumatology, University Medical Center Bonn (UKB), Bonn, Germany
| | - Jörg Ellinger
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Manuel Ritter
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
| | - Abdullah Alajati
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Christoph Kuppe
- Institute of Experimental Medicine and Systems Biology and Division of Nephrology, RWTH Aachen University, Aachen, Germany
| | - Joshua Meeks
- Department of Urology, Feinberg School of Medicine, Chicago, IL
| | | | - J. Alberto Nakauma-González
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Joost Boormans
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Kerstin Junker
- Department of Urology and Pediatric Urology, Saarland University, Homburg, Germany
| | - Arndt Hartmann
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Center for Cancer Research (Bayerisches Zentrum für Krebsforschung, BZKF), Erlangen, Germany
| | - Viktor Grünwald
- Clinic for Internal Medicine (Tumor Research) and Clinic for Urology, Interdisciplinary Genitourinary Oncology at the West-German Cancer Center, Essen University Hospital, Essen, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Markus Eckstein
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Center for Cancer Research (Bayerisches Zentrum für Krebsforschung, BZKF), Erlangen, Germany
| |
Collapse
|
6
|
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
|
7
|
Antonarelli G, Corti C, Tarantino P, Salimbeni BT, Zagami P, Marra A, Trapani D, Tolaney S, Cortes J, Curigliano G. Management of patients with HER2-positive metastatic breast cancer after trastuzumab deruxtecan failure. ESMO Open 2023; 8:101608. [PMID: 37467660 PMCID: PMC10372550 DOI: 10.1016/j.esmoop.2023.101608] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/11/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023] Open
Abstract
The current treatment of patients with human epidermal growth factor receptor 2 (HER2)-positive advanced breast cancer (ABC) has been greatly impacted in the past decade by the introduction of antibody-drug conjugates (ADCs), which represent a relatively novel therapeutic class with the peculiar ability to deliver otherwise overtly toxic chemotherapeutics to tumor sites by exploiting the specificities of monoclonal antibodies. Indeed, drug engineering refinements in ADC design, such as through the introduction of cleavable linkers and hydrophobic payloads, resulted in improved patient outcomes in recent years. Two different ADCs, namely trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd), have already entered clinical practice for the treatment of HER2-positive ABC. In this scenario, T-DXd has shown to portend better survival outcomes compared to T-DM1, while leaving a large unsought area of unmet medical need upon T-DXd failure. Treatment decision and benefit of cancer drugs following T-DXd still represent an area of clinical controversy, where a preclinical investigation and clinical development should be prioritized. As the pace of innovation is currently accelerating, and with novel ADC formulations advancing in early-phase clinical trials, the whole BC field is changing at an unprecedented rate, with potential broadenings of therapeutic indications. In this review, we present the clinical landscape of HER2-positive advanced BC and discuss our vision on how to tackle T-DXd resistance, providing a perspective on the priority areas of the cancer research in this setting.
Collapse
Affiliation(s)
- G Antonarelli
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan; Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy. https://twitter.com/GabrAnton
| | - C Corti
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan; Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy. https://twitter.com/CCortiMD
| | - P Tarantino
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan; Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA. https://twitter.com/PTarantinoMD
| | - B T Salimbeni
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan; Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
| | - P Zagami
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan; Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy. https://twitter.com/paolazagam
| | - A Marra
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy. https://twitter.com/antoniomarraMD
| | - D Trapani
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan; Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy. https://twitter.com/darioT_
| | - S Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA. https://twitter.com/stolaney1
| | - J Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona; Medical Scientia Innovation Research (MedSIR), Barcelona; Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain.
| | - G Curigliano
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan; Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy.
| |
Collapse
|
8
|
Guidi L, Valenza C, Trapani D, Curigliano G. Antibody-drug conjugates targeting HER2-mutant or HER2-(non) amplified solid tumors: is HER2 an agnostic target? Expert Opin Biol Ther 2023; 23:1039-1042. [PMID: 37897696 DOI: 10.1080/14712598.2023.2277915] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/27/2023] [Indexed: 10/30/2023]
Affiliation(s)
- Lorenzo Guidi
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- 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, Milan, Italy
- 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, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
9
|
Corti C, Boscolo Bielo L, Schianca AC, Salimbeni BT, Criscitiello C, Curigliano G. Future potential targets of antibody-drug conjugates in breast cancer. Breast 2023; 69:312-322. [PMID: 36996620 PMCID: PMC10063401 DOI: 10.1016/j.breast.2023.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/06/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Metastatic breast cancer (BC) remains an incurable disease. Besides endocrine and targeted agents, chemotherapy is still a relevant therapeutic option for this disease. Recently, antibody-drug conjugates (ADCs) have shown to overcome the lack of tumor specificity and systemic toxicity typically associated with traditional chemotherapies, thus improving the therapeutic index. To effectively exploit this technological breakthrough, identification of optimal target antigens (Ags) is of utmost importance. To make the ideal target, differential expression of target Ags between healthy and cancer tissues, as well as specific mechanisms of ADC internalization after Ag-antibody interaction are required. Therefore, several in silico strategies to identify and characterize new promising candidate Ags have been developed. If initial in vitro and in vivo positive data are documented, thus providing a biological rationale for further Ag investigation, early phase clinical trials are designed. In BC, these strategies have already led to the development of effective ADCs, namely trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd) and sacituzumab govitecan (SG), primarily targeting HER2 and TROP-2. However, promising new Ags are currently under investigation, with encouraging results especially coming from targeting HER3, FRα, Tissue Factor, LIV-1, ROR1-2, and B7-H4. In this review, we describe the landscape of emergent and future potential targets (i.e., other than HER2 and TROP-2) investigated in BC for ADC development. Predominant target expression, function, preclinical rationale, potential clinical implication, as well as preliminary clinical trial results are provided.
Collapse
Affiliation(s)
- Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy.
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Ambra Carnevale Schianca
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Beatrice Taurelli Salimbeni
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, 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 Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
10
|
Ivanova M, Porta FM, Giugliano F, Frascarelli C, Sajjadi E, Venetis K, Cursano G, Mazzarol G, Guerini-Rocco E, Curigliano G, Criscitiello C, Fusco N. Breast Cancer with Brain Metastasis: Molecular Insights and Clinical Management. Genes (Basel) 2023; 14:1160. [PMID: 37372340 DOI: 10.3390/genes14061160] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Breast cancer is the most frequently diagnosed malignancy worldwide and the leading cause of cancer-related death among women. Brain metastases are a primary contributor to mortality, as they often go undetected until late stages due to their dormant nature. Moreover, the clinical management of brain metastases is complicated by the relevant issue of blood-brain barrier penetration. The molecular pathways involved in the formation, progression, and colonization of primary breast tumors and subsequent brain metastases are diverse, posing significant hurdles due to the heterogeneous nature of breast cancer subtypes. Despite advancements in primary breast cancer treatments, the prognosis for patients with brain metastases remains poor. In this review, we aim to highlight the biological mechanisms of breast cancer brain metastases by evaluating multi-step genetic pathways and to discuss currently available and emerging treatment strategies to propose a prospective overview of the management of this complex disease.
Collapse
Affiliation(s)
- Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Francesca Maria Porta
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- School of Pathology, University of Milan, 20122 Milan, Italy
| | - Federica Giugliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giovanni Mazzarol
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
11
|
Tateo V, Marchese PV, Mollica V, Massari F, Kurzrock R, Adashek JJ. Agnostic Approvals in Oncology: Getting the Right Drug to the Right Patient with the Right Genomics. Pharmaceuticals (Basel) 2023; 16:ph16040614. [PMID: 37111371 PMCID: PMC10144220 DOI: 10.3390/ph16040614] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
(1) Background: The oncology field has drastically changed with the advent of precision medicine, led by the discovery of druggable genes or immune targets assessed through next-generation sequencing. Biomarker-based treatments are increasingly emerging, and currently, six tissue-agnostic therapies are FDA-approved. (2) Methods: We performed a review of the literature and reported the trials that led to the approval of tissue-agnostic treatments and ongoing clinical trials currently investigating novel biomarker-based approaches. (3) Results: We discussed the approval of agnostic treatments: pembrolizumab and dostarlimab for MMRd/MSI-H, pembrolizumab for TMB-H, larotrectinib and entrectinib for NTRK-fusions, dabrafenib plus trametinib for BRAF V600E mutation, and selpercatinib for RET fusions. In addition, we reported novel clinical trials of biomarker-based approaches, including ALK, HER2, FGFR, and NRG1. (4) Conclusions: Precision medicine is constantly evolving, and with the improvement of diagnostic tools that allow a wider genomic definition of the tumor, tissue-agnostic targeted therapies are a promising treatment strategy tailored to the specific tumor genomic profile, leading to improved survival outcomes.
Collapse
Affiliation(s)
- Valentina Tateo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Paola Valeria Marchese
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40127 Bologna, Italy
| | - Razelle Kurzrock
- MCW Cancer Center, Milwaukee, WI 53226, USA
- WIN Consortium, San Diego, CA 92093, USA
- Department of Oncology, University of Nebraska, Omaha, NE 68198, USA
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
12
|
Corti C, Giugliano F, Nicolò E, Tarantino P, Criscitiello C, Curigliano G. HER2-Low Breast Cancer: a New Subtype? Curr Treat Options Oncol 2023; 24:468-478. [PMID: 36971965 DOI: 10.1007/s11864-023-01068-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2023] [Indexed: 03/29/2023]
Abstract
OPINION STATEMENT Breast cancer (BC) guidelines subdivide the disease into three main groups, namely hormone receptor (HR)-positive HER2-negative, HER2-positive, and triple-negative BC (TNBC). The natural history of the HER2-positive subtype has changed since the introduction of HER-targeted therapies, which demonstrated benefit only in case of HER2 overexpression (IHC, score 3+) or gene amplification. Such observation may depend on direct drug inhibition of HER2 downstream signaling, which is needed for survival and proliferation in HER2-addicted BC. Clinically focused categories cannot comprehensively describe biology, as almost half of the currently defined HER2-negative BCs show some degree of IHC expression and have been recently renamed as HER2-low. Why? As technological breakthroughs enable the synthesis of antibody-drug conjugates (ADCs), target antigens may be viewed not only as a biological switch to be turned on-off by targeted drugs but also as an anchor for ADC docking and tethering. As trastuzumab deruxtecan (T-DXd) has already proven in the clinical trial DESTINY-Breast04, even fewer HER2 available receptors on cancer cells may be sufficient for a clinical benefit. So, for HR-negative HER2-low subtype (~40% of TNBCs), though only 58 patients had been enrolled in DESTINY-Breast04, the observed benefit, together with the dismal prognosis of TNBC, warrants the use of T-DXd. Notably, another topoisomerase-based ADC, sacituzumab govitecan, has already been granted approval for pretreated TNBC (ASCENT). As no head-to-head comparison has been performed, the choice relies on regulatory approvals at the time of patient assessment, critical appraisal of available evidence, and careful evaluation of possible cross-resistance with sequential use of ADCs. As for HR-positive HER2-low disease (~60% of HR-positive tumors), DESTINY-Breast04 provides solid evidence for T-DXd prioritization in either second or third treatment lines. Although the remarkable activity observed in this setting favorably compares with outcomes observed in treatment-naive patients, the ongoing DESTINY-Breast06 will clarify the role of T-DXd in this population.
Collapse
|
13
|
García-Foncillas J. Precision Oncology: Next Steps. Arch Med Res 2022; 53:867-868. [PMID: 36473804 DOI: 10.1016/j.arcmed.2022.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Jesús García-Foncillas
- Hospital Universitario Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
14
|
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: 1.7] [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
|