51
|
Cogliati V, Capici S, Pepe FF, di Mauro P, Riva F, Cicchiello F, Maggioni C, Cordani N, Cerrito MG, Cazzaniga ME. How to Treat HR+/HER2- Metastatic Breast Cancer Patients after CDK4/6 Inhibitors: An Unfinished Story. Life (Basel) 2022; 12:378. [PMID: 35330128 PMCID: PMC8954717 DOI: 10.3390/life12030378] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 12/19/2022] Open
Abstract
CDK4/6 inhibitors in association with endocrine therapy represent the best therapeutic choice for either endocrine-sensitive or resistant hormone-receptor-positive advanced breast cancer patients. On the contrary, the optimal therapeutic strategy after the failure of CDK4/6 inhibitors-based treatment still remains an open question worldwide. In this review, we analyze the most studied mechanisms of resistance to CDK4/6 inhibitors treatment, as well as the most significant results of retrospective and prospective trials in the setting of progression after CDK4/6 inhibitors, to provide the reader a comprehensive overview from both a preclinical and especially a clinical perspective. In our opinion, an approach based on a deeper knowledge of resistance mechanisms to CDK4/6 inhibitors, but also on a careful analysis of what is done in clinical practice, can lead to a better definition of prospective randomized trials, to implement a personalized sequence approach, based on molecular analyses.
Collapse
Affiliation(s)
- Viola Cogliati
- Phase 1 Research Centre, ASST Monza, 20900 Monza, MB, Italy; (S.C.); (F.F.P.); (M.E.C.)
| | - Serena Capici
- Phase 1 Research Centre, ASST Monza, 20900 Monza, MB, Italy; (S.C.); (F.F.P.); (M.E.C.)
| | - Francesca Fulvia Pepe
- Phase 1 Research Centre, ASST Monza, 20900 Monza, MB, Italy; (S.C.); (F.F.P.); (M.E.C.)
| | - Pierluigi di Mauro
- Oncology Unit, ASST Monza, 20900 Monza, MB, Italy; (P.d.M.); (F.R.); (F.C.); (C.M.)
| | - Francesca Riva
- Oncology Unit, ASST Monza, 20900 Monza, MB, Italy; (P.d.M.); (F.R.); (F.C.); (C.M.)
| | - Federica Cicchiello
- Oncology Unit, ASST Monza, 20900 Monza, MB, Italy; (P.d.M.); (F.R.); (F.C.); (C.M.)
| | - Claudia Maggioni
- Oncology Unit, ASST Monza, 20900 Monza, MB, Italy; (P.d.M.); (F.R.); (F.C.); (C.M.)
| | - Nicoletta Cordani
- School of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, MB, Italy; (N.C.); (M.G.C.)
| | - Maria Grazia Cerrito
- School of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, MB, Italy; (N.C.); (M.G.C.)
| | - Marina Elena Cazzaniga
- Phase 1 Research Centre, ASST Monza, 20900 Monza, MB, Italy; (S.C.); (F.F.P.); (M.E.C.)
- School of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, MB, Italy; (N.C.); (M.G.C.)
| |
Collapse
|
52
|
Thomssen C, Fehm TN, Stickeler E, Fasching PA, Janni W, Kolberg-Liedtke C, Kolberg HC, Lüftner D, Müller V, Schütz F, Belleville E, Bader S, Untch M, Welslau M, Thill M, Hartkopf AD, Tesch H, Ditsch N, Lux MP, Wöckel A, Aktas B, Schneeweiss A, Würstlein R. Update Breast Cancer 2021 Part 4 - Prevention and Early Stages. Geburtshilfe Frauenheilkd 2022; 82:206-214. [PMID: 35169388 PMCID: PMC8837401 DOI: 10.1055/a-1724-9639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/18/2021] [Indexed: 12/18/2022] Open
Abstract
This past year has seen new and effective options for further improving treatment outcome in many patients with early-stage breast cancer. Patients with hormone receptor-positive disease benefited significantly from the addition of the CDK4/6 inhibitor abemaciclib to endocrine adjuvant therapy. In triple-negative disease, data were presented for two treatment regimens. Patients with advanced disease (stage 2 and 3) benefit from neoadjuvant treatment with the immune checkpoint inhibitor pembrolizumab in combination with standard chemotherapy, regardless of PD-L1 expression. When neoadjuvant therapy has failed to achieve the desired remission in BRCA1 and BRCA2 mutations, the administration of the PARP inhibitor olaparib has demonstrated an impressive response. Other data address translational issues in HER2-positive breast cancer and neoadjuvant therapy approaches with the oral SERD giredestrant and the PARP inhibitor talazoparib. This review presents and analyses the findings of this year' s most important study outcomes.
Collapse
Affiliation(s)
- Christoph Thomssen
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Tanja N. Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Elmar Stickeler
- Department of Gynecology and Obstetrics, RWTH University Hospital Aachen, Aachen, Germany
| | - Peter A. Fasching
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen,
Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Cornelia Kolberg-Liedtke
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
- palleos healthcare, Wiesbaden, Germany
- Phaon Scientific, Wiesbaden, Germany
| | | | - Diana Lüftner
- Charité University Hospital, Department of Hematology, Oncology and Tumour Immunology, University Medicine Berlin, Berlin, Germany
| | - Volkmar Müller
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | - Florian Schütz
- Gynäkologie und Geburtshilfe, Diakonissen-Stiftungs-Krankenhaus Speyer, Speyer, Germany
| | | | - Simon Bader
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen,
Germany
| | - Michael Untch
- Clinic for Gynecology and Obstetrics, Breast Cancer Center, Genecologic Oncology Center, Helios Klinikum Berlin Buch, Berlin, Germany
| | | | - Marc Thill
- Agaplesion Markus Krankenhaus, Department of Gynecology and Gynecological Oncology, Frankfurt am Main, Germany
| | - Andreas D. Hartkopf
- Department of Obstetrics and Gynecology, University of Tübingen, Tübingen, Germany
| | - Hans Tesch
- Oncology Practice at Bethanien Hospital, Frankfurt am Main, Germany
| | - Nina Ditsch
- Department of Gynecology and Obstetrics, University Hospital Augsburg, Augsburg, Germany
| | - Michael P. Lux
- Klinik für Gynäkologie und Geburtshilfe, Frauenklinik St. Louise, Paderborn, St. Josefs-Krankenhaus, Salzkotten, St. Vincenz Krankenhaus GmbH, Paderborn, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | - Bahriye Aktas
- Klinik und Poliklinik für Gynäkologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Rachel Würstlein
- Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, Munich, Germany
| |
Collapse
|
53
|
Lüftner D, Schütz F, Stickeler E, Fasching PA, Janni W, Kolberg-Liedtke C, Kolberg HC, Thomssen C, Müller V, Fehm TN, Belleville E, Bader S, Untch M, Welslau M, Thill M, Tesch H, Ditsch N, Lux MP, Wöckel A, Aktas B, Schneeweiss A, Würstlein R, Hartkopf AD. Update Breast Cancer 2021 Part 5 - Advanced Breast Cancer. Geburtshilfe Frauenheilkd 2022; 82:215-225. [PMID: 35169389 PMCID: PMC8837406 DOI: 10.1055/a-1724-9569] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/18/2021] [Indexed: 12/13/2022] Open
Abstract
Despite the COVID 19 pandemic and mostly virtual congresses, innovation in the treatment of breast cancer patients continues at an unabated pace. This review summarises the current developments. Initial overall survival data for CDK4/6 inhibitor treatment in combination with an aromatase inhibitor as the first advanced line of therapy in treatment-naive postmenopausal patients have been published. Similarly, a trial comparing trastuzumab-deruxtecan versus trastuzumab-emtansine revealed a clear benefit regarding progression-free survival. Understanding of biomarkers making checkpoint inhibitor therapy particularly effective is increasing, and new compounds such as oral selective estrogen receptor destabilisers (SERDs) are entering clinical development and completing the first phase III trials.
Collapse
Affiliation(s)
- Diana Lüftner
- Charité University Hospital, Department of Hematology, Oncology and Tumour Immunology, University Medicine Berlin, Berlin, Germany
| | - Florian Schütz
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Elmar Stickeler
- Department of Gynecology and Obstetrics, RWTH University Hospital Aachen, Aachen, Germany
| | - Peter A. Fasching
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen,
Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Cornelia Kolberg-Liedtke
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
- palleos healthcare, Wiesbaden, Germany
- Phaon Scientific, Wiesbaden, Germany
| | | | - Christoph Thomssen
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Volkmar Müller
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | - Tanja N. Fehm
- Gynäkologie und Geburtshilfe, Diakonissen-Stiftungs-Krankenhaus Speyer, Speyer, Germany
| | | | - Simon Bader
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen,
Germany
| | - Michael Untch
- Clinic for Gynecology and Obstetrics, Breast Cancer Center, Genecologic Oncology Center, Helios Klinikum Berlin Buch, Berlin, Germany
| | | | - Marc Thill
- Agaplesion Markus Krankenhaus, Department of Gynecology and Gynecological Oncology, Frankfurt am Main, Germany
| | - Hans Tesch
- Oncology Practice at Bethanien Hospital, Frankfurt am Main, Germany
| | - Nina Ditsch
- Department of Gynecology and Obstetrics, University Hospital Augsburg, Augsburg, Germany
| | - Michael P. Lux
- Klinik für Gynäkologie und Geburtshilfe, Frauenklinik St. Louise, Paderborn, St. Josefs-Krankenhaus, Salzkotten, St. Vincenz Krankenhaus GmbH, Paderborn, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | - Bahriye Aktas
- Klinik und Poliklinik für Gynäkologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Rachel Würstlein
- Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, Munich, Germany
| | - Andreas D. Hartkopf
- Department of Obstetrics and Gynecology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
54
|
Lloyd MR, Wander SA, Hamilton E, Razavi P, Bardia A. Next-generation selective estrogen receptor degraders and other novel endocrine therapies for management of metastatic hormone receptor-positive breast cancer: current and emerging role. Ther Adv Med Oncol 2022; 14:17588359221113694. [PMID: 35923930 PMCID: PMC9340905 DOI: 10.1177/17588359221113694] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Endocrine therapy (ET) is a pivotal strategy to manage early- and advanced-stage estrogen receptor-positive (ER+) breast cancer. In patients with metastatic breast cancer (MBC), progression of disease inevitably occurs due to the presence of acquired or intrinsic resistance mechanisms. ET resistance can be driven by ligand-independent, ER-mediated signaling that promotes tumor proliferation in the absence of hormone, or ER-independent oncogenic signaling that circumvents endocrine regulated transcription pathways. Estrogen receptor 1 (ESR1) mutations induce constitutive ER activity and upregulate ER-dependent gene transcription, provoking resistance to estrogen deprivation and aromatase inhibitor therapy. The role ESR1 mutations play in regulating response to other therapies, such as the selective estrogen receptor degrader (SERD) fulvestrant and the available CDK4/6 inhibitors, is less clear. Novel oral SERDs and other next-generation ETs are in clinical development for ER+ breast cancer as single agents and in combination with established targeted therapies. Recent results from the phase III EMERALD trial demonstrated improved outcomes with the oral SERD elacestrant compared to standard anti-estrogen therapies in ER+ MBC after prior progression on ET, and other agents have shown promise in both the laboratory and early-phase clinical trials. In this review, we will discuss the emerging data related to oral SERDs and other novel ET in managing ER+ breast cancer. As clinical data continue to mature on these next-generation ETs, important questions will emerge related to the optimal sequence of therapeutic options and the genomic and molecular landscape of resistance to these agents.
Collapse
Affiliation(s)
- Maxwell R. Lloyd
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Seth A. Wander
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Erika Hamilton
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | - Pedram Razavi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, 10 North Grove Street, Harvard Medical School, Boston, MA 02114-2621, USA
| |
Collapse
|
55
|
Patel JM, Jeselsohn RM. Estrogen Receptor Alpha and ESR1 Mutations in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:171-194. [DOI: 10.1007/978-3-031-11836-4_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
56
|
Saatci O, Huynh-Dam KT, Sahin O. Endocrine resistance in breast cancer: from molecular mechanisms to therapeutic strategies. J Mol Med (Berl) 2021; 99:1691-1710. [PMID: 34623477 PMCID: PMC8611518 DOI: 10.1007/s00109-021-02136-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/20/2021] [Accepted: 09/06/2021] [Indexed: 12/31/2022]
Abstract
Estrogen receptor-positive (ER +) breast cancer accounts for approximately 75% of all breast cancers. Endocrine therapies, including selective ER modulators (SERMs), aromatase inhibitors (AIs), and selective ER down-regulators (SERDs) provide substantial clinical benefit by reducing the risk of disease recurrence and mortality. However, resistance to endocrine therapies represents a major challenge, limiting the success of ER + breast cancer treatment. Mechanisms of endocrine resistance involve alterations in ER signaling via modulation of ER (e.g., ER downregulation, ESR1 mutations or fusions); alterations in ER coactivators/corepressors, transcription factors (TFs), nuclear receptors and epigenetic modulators; regulation of signaling pathways; modulation of cell cycle regulators; stress signaling; and alterations in tumor microenvironment, nutrient stress, and metabolic regulation. Current therapeutic strategies to improve outcome of endocrine-resistant patients in clinics include inhibitors against mechanistic target of rapamycin (mTOR), cyclin-dependent kinase (CDK) 4/6, and the phosphoinositide 3-kinase (PI3K) subunit, p110α. Preclinical studies reveal novel therapeutic targets, some of which are currently tested in clinical trials as single agents or in combination with endocrine therapies, such as ER partial agonists, ER proteolysis targeting chimeras (PROTACs), next-generation SERDs, AKT inhibitors, epidermal growth factor receptor 1 and 2 (EGFR/HER2) dual inhibitors, HER2 targeting antibody-drug conjugates (ADCs) and histone deacetylase (HDAC) inhibitors. In this review, we summarize the established and emerging mechanisms of endocrine resistance, alterations during metastatic recurrence, and discuss the approved therapies and ongoing clinical trials testing the combination of novel targeted therapies with endocrine therapy in endocrine-resistant ER + breast cancer patients.
Collapse
Affiliation(s)
- Ozge Saatci
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715, Sumter Street, CLS609D, Columbia, SC, 29208, USA
| | - Kim-Tuyen Huynh-Dam
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715, Sumter Street, CLS609D, Columbia, SC, 29208, USA
| | - Ozgur Sahin
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715, Sumter Street, CLS609D, Columbia, SC, 29208, USA.
| |
Collapse
|
57
|
Nabieva N, Fasching PA. Endocrine Treatment for Breast Cancer Patients Revisited-History, Standard of Care, and Possibilities of Improvement. Cancers (Basel) 2021; 13:5643. [PMID: 34830800 PMCID: PMC8616153 DOI: 10.3390/cancers13225643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 01/16/2023] Open
Abstract
PURPOSE OF REVIEW Due to the findings of current studies and the approval of novel substances for the therapy of hormone-receptor-positive breast cancer patients, the established standards of endocrine treatment are changing. The purpose of this review is to give an overview of the history of endocrine treatment, to clarify its role in the present standard of care, and to discuss the possibilities of improvement. RECENT FINDINGS Tamoxifen, aromatase inhibitors, and fulvestrant are the main drugs that have been used for decades in the therapy of hormone-receptor-positive breast cancer patients. However, since a relevant number of women suffer at some point from disease recurrence or progression, several novel substances are being investigated to overcome resistance mechanisms by interfering with certain signaling pathways, such as the PI3K/AKT/mTOR or the CDK4/6 pathways. mTOR and CDK4/6 inhibitors were the first drugs approved for this purpose and many more are in development. SUMMARY Endocrine treatment is one of the best tolerable cancer therapies available. Continuous investigation serves to improve patients' outcomes and modernize the current standard of care. Considering the resistance mechanisms and substances analyzed against these, endocrine treatment of hormone-receptor-positive breast cancer is on the brink of a new era.
Collapse
Affiliation(s)
- Naiba Nabieva
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
- Novartis Oncology, Novartis Pharma GmbH, 90429 Nuremberg, Germany
| | - Peter A. Fasching
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| |
Collapse
|
58
|
Wilson BE, Cescon DW. Accelerating drug access from advanced to early breast cancer: the special case of oral selective estrogen receptor degraders. Curr Opin Oncol 2021; 33:538-546. [PMID: 34555836 DOI: 10.1097/cco.0000000000000786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW For hormone receptor positive breast cancer, the development of endocrine resistance commonly occurs, presenting as either disease progression in the metastatic setting or recurrence during or following adjuvant endocrine therapy. Various mechanisms of resistance have been described. In order to reduce or overcome endocrine resistance, there has been substantial interest in developing potent and orally bioavailable selective estrogen receptor degraders (SERDs) for metastatic disease and select patients with early-stage estrogen receptor positive breast cancer. RECENT FINDINGS At least 11 oral SERDs have entered clinical development. We review current studies in both the metastatic and neoadjuvant/adjuvant setting and present the available evidence of benefit and toxicity for these novel agents. Further characterization of changes to tissue-based biomarkers such as estrogen receptor, progesterone receptor and Ki67 expression and blood-based biomarkers such as ctDNA and estrogen receptor 1 mutation may help to refine therapeutic strategies, combinations, and patient selection to identify women who are most likely to benefit from these novel endocrine agents. SUMMARY Although SERDs have clear therapeutic potential based on nonclinical studies and have demonstrated early signs of activity in phase I and II studies in the metastatic setting, ongoing research is needed to clarify when and in whom these agents may have greatest clinical benefit.
Collapse
Affiliation(s)
- Brooke E Wilson
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.,University of New South Wales, Kensington, New South Wales, Australia
| | - David W Cescon
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
59
|
Jhaveri K, Juric D, Yap YS, Cresta S, Layman RM, Duhoux FP, Terret C, Takahashi S, Huober J, Kundamal N, Sheng Q, Balbin A, Ji Y, He W, Crystal A, De Vita S, Curigliano G. A Phase I Study of LSZ102, an Oral Selective Estrogen Receptor Degrader, with or without Ribociclib or Alpelisib, in Patients with Estrogen Receptor-Positive Breast Cancer. Clin Cancer Res 2021; 27:5760-5770. [PMID: 34433648 PMCID: PMC9401512 DOI: 10.1158/1078-0432.ccr-21-1095] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/08/2021] [Accepted: 08/19/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE Data are sparse for oral selective estrogen receptor (ER) degraders (SERD) in cancer treatment. The investigational oral SERD LSZ102 was assessed in monotherapy and combination use in a phase I study. PATIENTS AND METHODS A phase I, multicenter, open-label dose-escalation study (NCT02734615) of LSZ102 alone (arm A; n = 77) or with ribociclib (arm B; n = 78) or alpelisib (arm C; n = 43) in heavily pretreated adults with histologically confirmed ER-positive breast cancer and prior disease progression. Arm A received LSZ102 200-900 mg/day; arm B, LSZ102 200-600 mg/day plus ribociclib 300-600 mg/day; arm C, LSZ102 300-450 mg/day plus alpelisib 200-300 mg/day. Key outcomes were dose-limiting toxicities (DLT) in the first 28-day treatment cycle, adverse events (AE), laboratory parameters, pharmacokinetics, biopsy ER protein, and investigator-assessed clinical response (RECIST v1.1). RESULTS The most common AEs were gastrointestinal. Treatment-related serious AEs occurred in 10% of participants (19/198), mostly in arm C [10/43 (23%)]. DLTs occurred in: arm A, 5% (4/77); arm B, 3% (2/78); and arm C, 19% (8/43). LSZ102 exposure was slightly greater than dose proportional. On-treatment biopsy ER reductions were observed, with a trend toward an LSZ102 dose response. Objective response rates (95% confidence interval) were: arm A, 1.3% (0.0-7.0); arm B, 16.9% (9.3-27.1); and arm C, 7.0% (1.5-19.1), and clinical benefit rates 7.8% (2.9-16.2), 35.1% (24.5-46.8), and 20.9% (10.0-36.0), respectively. CONCLUSIONS LSZ102 was well tolerated alone and with ribociclib and had a manageable safety profile with alpelisib. Preliminary clinical activity was observed in combination use.
Collapse
Affiliation(s)
- Komal Jhaveri
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York.
| | - Dejan Juric
- Massachusetts General Hospital, Boston, Massachusetts
| | | | - Sara Cresta
- Fondazione IRCCS-Istituto Nazionale dei Tumori, Milan, Italy
| | - Rachel M Layman
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | | | | | | | - Jens Huober
- Department of Gynecology, Breast Center, University of Ulm, Ulm, Germany
| | - Nicole Kundamal
- Novartis Institutes for Biomedical Research, East Hanover, New Jersey
| | - Qing Sheng
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Alejandro Balbin
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Yan Ji
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Wei He
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Adam Crystal
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Serena De Vita
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan and Istituto Europeo di Oncologia-IRCCS, Division of Early Drug Development, Milan, Italy.
| |
Collapse
|
60
|
Chen YC, Yu J, Metcalfe C, De Bruyn T, Gelzleichter T, Malhi V, Perez-Moreno PD, Wang X. Latest generation estrogen receptor degraders for the treatment of hormone receptor-positive breast cancer. Expert Opin Investig Drugs 2021; 31:515-529. [PMID: 34694932 DOI: 10.1080/13543784.2021.1983542] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The selective estrogen receptor degrader (SERD) and full receptor antagonist provides an important therapeutic option for hormone receptor (HR)-positive breast cancer. Endocrine therapies include tamoxifen, a selective estrogen receptor modulator (SERM), that exhibits receptor agonist and antagonist activity, and aromatase inhibitors that block estrogen biosynthesis but which demonstrate acquired resistance. Fulvestrant, the only currently approved SERD, is limited by poor drug-like properties. A key focus for improving disease management has been development of oral SERDs with optimized target occupancy and potency and superior clinical efficacy. AREAS COVERED Using PubMed, clinicaltrials.gov, and congress websites, this review explored the preclinical development and clinical pharmacokinetics from early phase clinical studies (2015 or later) of novel oral SERDs, including giredestrant, amcenestrant, camizestrant, elacestrant, and rintodestrant. EXPERT OPINION Numerous oral SERDs are in clinical development, aiming to form the core endocrine therapy for HR-positive breast cancer. Through property- and structure-based drug design of estrogen receptor-binding, antagonism, degradation, anti-proliferation, and pharmacokinetic properties, these SERDs have distinct profiles which impact clinical dosing, efficacy, and safety. Assuming preliminary safety and activity data are confirmed in phase 3 trials, these promising agents could further improve the management, outcomes, and quality of life in HR-positive breast cancer.
Collapse
Affiliation(s)
- Ya-Chi Chen
- Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Jiajie Yu
- Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Ciara Metcalfe
- Discovery Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Tom De Bruyn
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Thomas Gelzleichter
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, USA
| | - Vikram Malhi
- Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | | | - Xiaojing Wang
- Discovery Chemistry, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
61
|
Brett JO, Spring LM, Bardia A, Wander SA. ESR1 mutation as an emerging clinical biomarker in metastatic hormone receptor-positive breast cancer. Breast Cancer Res 2021; 23:85. [PMID: 34392831 PMCID: PMC8365900 DOI: 10.1186/s13058-021-01462-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/20/2021] [Indexed: 11/10/2022] Open
Abstract
In metastatic hormone receptor-positive breast cancer, ESR1 mutations are a common cause of acquired resistance to the backbone of therapy, estrogen deprivation by aromatase inhibition. How these mutations affect tumor sensitivity to established and novel therapies are active areas of research. These therapies include estrogen receptor-targeting agents, such as selective estrogen receptor modulators, covalent antagonists, and degraders (including tamoxifen, fulvestrant, and novel agents), and combination therapies, such as endocrine therapy plus CDK4/6, PI3K, or mTORC1 inhibition. In this review, we summarize existing knowledge surrounding the mechanisms of action of ESR1 mutations and roles in resistance to aromatase inhibition. We then analyze the recent literature on how ESR1 mutations affect outcomes in estrogen receptor-targeting and combination therapies. For estrogen receptor-targeting therapies such as tamoxifen and fulvestrant, ESR1 mutations cause relative resistance in vitro but do not clearly lead to resistance in patients, making novel agents in this category promising. Regarding combination therapies, ESR1 mutations nullify any aromatase inhibitor component of the combination. Thus, combinations using endocrine alternatives to aromatase inhibition, or combinations where the non-endocrine component is efficacious as monotherapy, are still effective against ESR1 mutations. These results emphasize the importance of investigating combinatorial resistance, challenging as these efforts are. We also discuss future directions and open questions, such as studying the differences among distinct ESR1 mutations, asking how to adjust clinical decisions based on molecular surveillance testing, and developing novel therapies that are effective against ESR1 mutations.
Collapse
Affiliation(s)
- Jamie O Brett
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Laura M Spring
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA, 02114, USA
| | - Aditya Bardia
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA, 02114, USA
| | - Seth A Wander
- Harvard Medical School, Boston, MA, USA.
- Department of Medical Oncology, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
62
|
Chien TJ. A review of the endocrine resistance in hormone-positive breast cancer. Am J Cancer Res 2021; 11:3813-3831. [PMID: 34522451 PMCID: PMC8414389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/25/2021] [Indexed: 06/13/2023] Open
Abstract
Hormone-positive breast cancer (BC) is a unique heterogeneous disease with a favorable prognosis compared to other types of breast cancer. As tumor biology influences the prognosis and clinical treatment, a deep understanding of how the molecular mechanisms regulate hormone sensitivity or resistance is critical in improving the efficacy and overcoming the endocrine resistance. This article comprehensively reviews the endocrine resistance in hormone-positive BC from a molecular and genetic perspective, encompassing the updated treatment and developing direction. This review includes the mechanisms of hormone resistance, which vary from epigenetic changes, crosstalk between signaling networks, cell cycle aberrance, and even change in the tumor microenvironment (TME) or stem cell. These mechanisms may contribute to treatment resistance. Current targeted therapy for hormone-resistant tumors includes PI3K/AKT/mTOR and cdk4/6 inhibitors. Several relevant pathways, biomarkers, and predictor genes have also been identified. Immunotherapy so far has a relatively less crucial role in hormone-positive than in triple-negative BC. Furthermore, the methodology to identify the PDL1 is not standardized. In a molecule and gene study, next-generation sequencing with circulating tumor DNA (ctDNA) has recently appeared as a sensitive and minimally invasive tool worth investigating.
Collapse
Affiliation(s)
- Tsai-Ju Chien
- Division of Hemato-Oncology, Department of Internal Medicine, Branch of Zhong-Zhou, Taipei City HospitalTaipei, Taiwan
- Division of Hemato-Oncology, Department of Internal Medicine, Branch of Jen-Ai, Taipei City HospitalTaipei, Taiwan
- Institute of Traditional Medicine, National Yang-Ming Chiao Tung UniversityTaipei, Taiwan
| |
Collapse
|
63
|
Current and emerging estrogen receptor-targeted therapies for the treatment of breast cancer. Essays Biochem 2021; 65:985-1001. [PMID: 34328178 DOI: 10.1042/ebc20200174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/18/2022]
Abstract
Nearly 80% of all breast cancers are estrogen receptor positive (ER+) and require the activity of this transcription factor for tumor growth and survival. Thus, endocrine therapies, which target the estrogen signaling axis, have and will continue to be the cornerstone of therapy for patients diagnosed with ER+ disease. Several inhibitors of ER activity exist, including aromatase inhibitors (AIs), selective estrogen receptor modulators (SERMs), selective estrogen receptor degraders/down-regulators (SERDs), and ER proteolysis-targeting chimeras (ER PROTACs); drugs which differ in the mechanism(s) by which they inhibit this signaling pathway. Notwithstanding their significant impact on the management of this disease, resistance to existing endocrine therapies remains a major impediment to durable clinical responses. Although the mechanisms of resistance are complex and varied, dependence on ER is typically retained after progression on SERMs and AIs, suggesting that ER remains a bona fide therapeutic target. The discovery and development of orally bioavailable drugs that eliminate ER expression (SERDs and ER PROTACs) will likely aid in treating this growing patient population. All of the existing endocrine therapies were developed with the intent of inhibiting the cancer cell intrinsic actions of ER and/or with the objective of achieving extreme estrogen deprivation and most achieve that goal. A longstanding question that remains to be addressed, however, is how actions of existing interventions extrinsic to the cancer cells influence tumor biology. We believe that these issues need to be addressed in the development of strategies to develop the next generation of ER-modulators optimized for positive activities in both cancer cells and other cells within the tumor microenvironment (TME).
Collapse
|
64
|
Hernando C, Ortega-Morillo B, Tapia M, Moragón S, Martínez MT, Eroles P, Garrido-Cano I, Adam-Artigues A, Lluch A, Bermejo B, Cejalvo JM. Oral Selective Estrogen Receptor Degraders (SERDs) as a Novel Breast Cancer Therapy: Present and Future from a Clinical Perspective. Int J Mol Sci 2021; 22:ijms22157812. [PMID: 34360578 PMCID: PMC8345926 DOI: 10.3390/ijms22157812] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 01/21/2023] Open
Abstract
Estrogen receptor-positive (ER+) is the most common subtype of breast cancer. Endocrine therapy is the fundamental treatment against this entity, by directly or indirectly modifying estrogen production. Recent advances in novel compounds, such as cyclin-dependent kinase 4/6 inhibitors (CDK4/6i), or phosphoinositide 3-kinase (PI3K) inhibitors have improved progression-free survival and overall survival in these patients. However, some patients still develop endocrine resistance after or during endocrine treatment. Different underlying mechanisms have been identified as responsible for endocrine treatment resistance, where ESR1 gene mutations are one of the most studied, outstanding from others such as somatic alterations, microenvironment involvement and epigenetic changes. In this scenario, selective estrogen receptor degraders/downregulators (SERD) are one of the weapons currently in research and development against aromatase inhibitor- or tamoxifen-resistance. The first SERD to be developed and approved for ER+ breast cancer was fulvestrant, demonstrating also interesting activity in ESR1 mutated patients in the second line treatment setting. Recent investigational advances have allowed the development of new oral bioavailable SERDs. This review describes the evolution and ongoing studies in SERDs and new molecules against ER, with the hope that these novel drugs may improve our patients’ future landscape.
Collapse
Affiliation(s)
- Cristina Hernando
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
- Correspondence: (C.H.); (J.M.C.)
| | - Belén Ortega-Morillo
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
| | - Marta Tapia
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
| | - Santiago Moragón
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
| | - María Teresa Martínez
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
| | - Pilar Eroles
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, 28029 Madrid, Spain
- Departamento de Fisiología, Universidad de València, 46010 Valencia, Spain
| | - Iris Garrido-Cano
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
| | - Anna Adam-Artigues
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
| | - Ana Lluch
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, 28029 Madrid, Spain
| | - Begoña Bermejo
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, 28029 Madrid, Spain
| | - Juan Miguel Cejalvo
- Hospital Clínico de València, Instituto de Investigación INCLIVA, 46010 Valencia, Spain; (B.O.-M.); (M.T.); (S.M.); (M.T.M.); (I.G.-C.); (A.A.-A.); (A.L.); (B.B.)
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, 28029 Madrid, Spain
- Correspondence: (C.H.); (J.M.C.)
| |
Collapse
|
65
|
Crimini E, Repetto M, Aftimos P, Botticelli A, Marchetti P, Curigliano G. Precision medicine in breast cancer: From clinical trials to clinical practice. Cancer Treat Rev 2021; 98:102223. [PMID: 34049187 DOI: 10.1016/j.ctrv.2021.102223] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Breast cancer (BC) is the most common cancer in women and, despite the undeniable improvements in the outcome of these patients obtained in the last decade, the discovery and the validation of new actionable molecular targets represent a priority. ESCAT permits to rank molecular alterations in different classes according to their evidence of actionability in a specific cancer type, assisting clinicians in their therapeutical decisions. MAIN: ERBB2, PIK3CA and germline BRCA1/2 alterations are biomarkers prospectively validated in BC, driving the selection of targeted therapies, and are therefore classified in the highest level of evidence (Ia). Agnostic biomarkers, namely microsatellite instability, NTRK fusions and high tumor mutational burden, demonstrated similar activity across different tumor types and are consequently ranked in tier Ic. In tier II are classified alterations that still need confirmatory prospective studies but for which evidence of efficacy is available. Somatic BRCA1/2 mutations, germline PALB2 mutations, HER2-low expression, ERBB2 mutations, PTEN deletions, AKT1 mutations, ESR1 resistance mutations satisfy the requirements to be classified in this tier. In tier III are ranked various molecular alterations for which there is evidence of actionability in other tumors (IIIa) or that have similar functional impact in the same gene or pathway of a tier I alteration, without clinical data (IIIb). In tier IV are listed the molecular alterations for which only preclinical studies are available. CONCLUSION In this review we report the most relevant molecular targets in BC, ordered pursuant to their pathway and classified in concordance with ESCAT.
Collapse
Affiliation(s)
- Edoardo Crimini
- European Institute of Oncology, IRCCS, 20141 Milan, Italy; Department of Oncology and Hematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Matteo Repetto
- European Institute of Oncology, IRCCS, 20141 Milan, Italy; Department of Oncology and Hematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Philippe Aftimos
- Institut Jules Bordet - Université Libre de Bruxelles, Brussels Belgium
| | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, 20141 Milan, Italy; Department of Oncology and Hematology (DIPO), University of Milan, 20122 Milan, Italy.
| |
Collapse
|
66
|
McDonnell DP, Wardell SE, Chang CY, Norris JD. Next-Generation Endocrine Therapies for Breast Cancer. J Clin Oncol 2021; 39:1383-1388. [PMID: 33705209 DOI: 10.1200/jco.20.03565] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC
| | - Suzanne E Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC
| | - John D Norris
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
67
|
Liefaard MC, Lips EH, Wesseling J, Hylton NM, Lou B, Mansi T, Pusztai L. The Way of the Future: Personalizing Treatment Plans Through Technology. Am Soc Clin Oncol Educ Book 2021; 41:1-12. [PMID: 33793316 DOI: 10.1200/edbk_320593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Advances in tissue analysis methods, image analysis, high-throughput molecular profiling, and computational tools increasingly allow us to capture and quantify patient-to patient variations that impact cancer risk, prognosis, and treatment response. Statistical models that integrate patient-specific information from multiple sources (e.g., family history, demographics, germline variants, imaging features) can provide individualized cancer risk predictions that can guide screening and prevention strategies. The precision, quality, and standardization of diagnostic imaging are improving through computer-aided solutions, and multigene prognostic and predictive tests improved predictions of prognosis and treatment response in various cancer types. A common theme across many of these advances is that individually moderately informative variables are combined into more accurate multivariable prediction models. Advances in machine learning and the availability of large data sets fuel rapid progress in this field. Molecular dissection of the cancer genome has become a reality in the clinic, and molecular target profiling is now routinely used to select patients for various targeted therapies. These technology-driven increasingly more precise and quantitative estimates of benefit versus risk from a given intervention empower patients and physicians to tailor treatment strategies that match patient values and expectations.
Collapse
Affiliation(s)
- Marte C Liefaard
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nola M Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA
| | - Bin Lou
- Digital Technology and Innovation, Siemens Healthineers, Princeton, NJ
| | - Tommaso Mansi
- Digital Technology and Innovation, Siemens Healthineers, Princeton, NJ
| | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, CT
| |
Collapse
|