1
|
Xu G, Havens CG, Deng Q, Lowenstein C, Samanta D, Vidal B, Behshad E, Russell M, Orth P, Rice CT, Nagilla R, Kirchhoff P, Chen Z, Rej RK, Acharyya RK, Wu D, Wang S, Zhang W, Wu W, Jolivette L, Strickland C, Sui Z, Mohammad HP, Zhang X, Priestley ES. Discovery and Characterization of PVTX-321 as a Potent and Orally Bioavailable Estrogen Receptor Degrader for ER+/HER2- Breast Cancer. J Med Chem 2025. [PMID: 40366756 DOI: 10.1021/acs.jmedchem.5c00223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Estrogen receptor α (ERα) is a key therapeutic target in ER+/HER2- breast cancer, but ESR1 mutations drive resistance to endocrine therapies. Heterobifunctional degraders (HBDs) targeting ERα offer a promising strategy to overcome this resistance. Here, we report PVTX-321 (16a), a potent ER HBD derived from a novel spirocyclic cereblon ligand and an ERα binder. PVTX-321 achieves a DC50 of 0.15 nM in MCF-7 cells and acts as a strong antagonist (IC50 = 59 nM), suppressing proliferation in ERα+ cell lines, including mutant variants (Y537S, D538G). It demonstrates favorable oral bioavailability, dose-dependent ERα degradation in vivo and induces tumor regression at 10 mg/kg (QD) in MCF-7 xenografts. With minimal CYP inhibition and a strong preclinical safety profile, PVTX-321 is a promising candidate for advancing ER+/HER2- breast cancer treatment.
Collapse
Affiliation(s)
- Guozhang Xu
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Courtney G Havens
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Qiaolin Deng
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Cassandra Lowenstein
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Debangshu Samanta
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Brian Vidal
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Elham Behshad
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Mike Russell
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Peter Orth
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Cory T Rice
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Rakesh Nagilla
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Paul Kirchhoff
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhixiang Chen
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Rohan Kalyan Rej
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ranjan Kumar Acharyya
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Dimin Wu
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- The Rogel Cancer Center, Department of Internal Medicine, Department of Pharmacology, and Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Weihong Zhang
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Wenxue Wu
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Larry Jolivette
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Corey Strickland
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Zhihua Sui
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Helai P Mohammad
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - Xuqing Zhang
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| | - E Scott Priestley
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia, Pennsylvania 19406, United States
| |
Collapse
|
2
|
Ferrari P, Schiavone ML, Scatena C, Nicolini A. Molecular Mechanisms and Therapeutic Strategies to Overcome Resistance to Endocrine Therapy and CDK4/6 Inhibitors in Advanced ER+/HER2- Breast Cancer. Int J Mol Sci 2025; 26:3438. [PMID: 40244377 PMCID: PMC11989623 DOI: 10.3390/ijms26073438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Approximately 70-80% of breast cancers are estrogen receptor-positive (ER+), with 65% of these cases also being progesterone receptor-positive (ER+PR+). In most cases of ER+ advanced breast cancer, endocrine therapy (ET) serves as the first-line treatment, utilizing various drugs that inhibit ER signaling. These include tamoxifen, a selective estrogen receptor modulator (SERM); fulvestrant, a selective estrogen receptor degrader (SERD); and aromatase inhibitors (AIs), which block estrogen synthesis. However, intrinsic or acquired hormone resistance eventually develops, leading to disease progression. The combination of ET with cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6is) has been shown to significantly increase progression-free survival (PFS) and, in some cases, overall survival (OS). CDK4/6is works by arresting the cell cycle in the G1 phase, preventing DNA synthesis, and enhancing the efficacy of ET. This review highlights the key mechanisms of resistance to ET, whether used alone or in combination with biological agents, as well as emerging therapeutic strategies aimed at overcoming resistance. Addressing ET resistance remains a work in progress, and in the near future, better patient selection for different therapeutic approaches is expected through the identification of more precise biological and genetic markers. In particular, liquid biopsy may provide a real-time portrait of the disease, offering insights into mechanisms driving ET resistance and cancer progression.
Collapse
Affiliation(s)
- Paola Ferrari
- Department of Oncology, Pisa University Hospital, Via Roma 57, 56126 Pisa, Italy; (C.S.); (A.N.)
| | - Maria Luisa Schiavone
- Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Cristian Scatena
- Department of Oncology, Pisa University Hospital, Via Roma 57, 56126 Pisa, Italy; (C.S.); (A.N.)
- Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Andrea Nicolini
- Department of Oncology, Pisa University Hospital, Via Roma 57, 56126 Pisa, Italy; (C.S.); (A.N.)
| |
Collapse
|
3
|
Neill NE, Mauro LA, Pennisi A. Novel Estrogen Receptor - Targeted Therapies in Hormone-Receptor Positive Breast Cancer. Curr Treat Options Oncol 2025; 26:302-312. [PMID: 40163189 DOI: 10.1007/s11864-025-01310-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
OPINION STATEMENT Endocrine therapy is the backbone of treatment for HR + /HER2- MBC. The introduction of novel endocrine-based therapies has changed the landscape of metastatic breast cancer care, with even more promising agents on the horizon. Given the consistent success in prolonging PFS and OS, CDK4/6 inhibitors should be used as first-line treatment. Once secondary resistance eventually develops after use of a CDK4/6 inhibitor, use of monotherapy with either AI or fulvestrant has shown poor outcome. For example, in the control group of the EMERALD trial, in which all the patients were required to have previously received a CDK4/6 inhibitor, median progression-free survival with endocrine therapy was only 1.9 months. Based on the emerging evidence, molecular profiling of tissue or liquid biopsy at progression of disease is crucial to select future therapy. For patients whose tumors harbor ESR1 mutations, oral SERDs are the preferred option. For those with PIK3CA or AKT1 mutation or PTEN inactivation, combination therapy with the AKT pathway inhibitor capivasertib is recommended. Alpelisib, the first AKT1 inhibitor approved in combination therapy with fulvestrant in PIK3CA mutated tumors only, is now less in favor given its challenging side effect profile. When mutations are not present, options include combination therapy with the mTOR inhibitor everolimus or changing endocrine therapy and continuing a CDK 4/6 inhibitor. In patients with short response to CDK4/6 inhibitors suggesting endocrine resistant disease, chemotherapy or antibody-drug conjugates should be considered. With better understanding of the mechanisms of resistance to CDK4/6 inhibitors, additional mutations could be identified and potentially targeted in order to provide individualized treatment options. Optimal sequencing of treatment options depends on several factors: (1) the presence of specific molecular aberrations; (2) previous treatment history, duration of response and patient's performance status; (3) balance between maximizing survival benefits with quality of life/toxicities; (4) disease burden. In the upcoming years, we anticipate FDA approvals for more of the SERD molecules both in monotherapy and in combination therapy which will continue to expand the options available for HR + /HER2- MBC patients.
Collapse
Affiliation(s)
- Nina E Neill
- Inova Schar Cancer Institute, Fairfax, Virginia, USA.
| | | | | |
Collapse
|
4
|
Okwor VC, Okwor JC, Ukwuoma MK, Mitha SB, Nweke MC. Effectiveness of Hormonal Therapy for Post-Menopausal Women with Hormone Receptor-Positive Advanced Breast Cancer: A Systematic Review and Meta-Analysis of Clinical Trials. Med Princ Pract 2025:1-22. [PMID: 40096837 DOI: 10.1159/000545264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 03/09/2025] [Indexed: 03/19/2025] Open
Abstract
OBJECTIVE Breast cancer (BC) cells exhibit mutations over time, conferring resistance to therapeutic approaches. We attempted to ascertain the efficacy of selected hormonal therapy for advanced BC. METHODS This is a systematic review and meta-analysis of clinical trials. We searched Medline, PubMed, Cochrane Library, Web of Science, and others. Studies that investigated the effectiveness of hormonal therapy for HR positive (HR+) advanced BC were included. The outcomes were progression-free survival (PFS), overall survival (OS), and objective response rate (ORR). A random-effect meta-analysis model was employed. The study protocol was registered with the International Prospective Register of Systematic Reviews: CRD42023431939. RESULTS Twenty-one studies were included in the meta-analysis with an overall sample size of 8,482. ORR and PFS between aromatase inhibitors (AIs) and other hormonal therapies: selective oestrogen receptor degrader, selective oestrogen modulator (SERM) and androgen inhibitors showed no significant difference (OR = 1.122 [0.917-1.374], p = 0.263; OR = 0.010 [0.000-1.292], p = 0.063), respectively. Subgroup analysis showed a statistically significant difference in ORR in favour of patients who received SERM compared to AI (OR = 1.362 [1.033-1.795], p = 0.028). For OS, no significant difference was observed among anastrozole, letrozole, and exemestane recepients (OR = 1.718 [0.021-139.128], p = 0.809). CONCLUSION Given the above findings, clinical decisions could be based on factors such as the line of cancer treatment, adverse events, drug dosing, and individual drug benefits. Although newer combination therapies are being adopted, the agents explored in this review are still widely used in clinical practice for HR+ BC.
Collapse
Affiliation(s)
- Vitalis C Okwor
- Department of Radiation and Clinical Oncology, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Juliet C Okwor
- Department of Chemical Pathology, College of Medicine, University of Nigeria Ituku-Ozalla Campus, Enugu, Nigeria
| | - Maryjane K Ukwuoma
- Department of Physiotherapy, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Sara B Mitha
- Information specialist, Steve Biko Library, Durban University of Technology, Durban, South Africa
| | - Martins C Nweke
- Information specialist, Steve Biko Library, Durban University of Technology, Durban, South Africa
- Department of Physiotherapy, David Umahi Federal University of Health Sciences, Uburu, Nigeria
| |
Collapse
|
5
|
Haki M, Bayat R. Innovative Approaches for Molecular Targeted Therapy of Breast Cancer: Interfering with Various Pathway Signaling. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2025; 14:533-551. [PMID: 40123585 PMCID: PMC11927153 DOI: 10.22088/ijmcm.bums.14.1.533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/26/2024] [Indexed: 03/25/2025]
Abstract
Breast cancer encompasses a diverse array of conditions classified as hormone receptor-positive, human epidermal growth factor receptor 2-positive, and triple-negative breast cancer subtypes, dictating treatment approaches. The therapeutic strategies commonly involve addressing estrogen receptors (ER) and HER2, which have exhibited efficacy in managing cancer. Nevertheless, the prevalence of resistance to these therapies, whether inherent or acquired, persists despite the introduction of novel treatment modalities. Progress in comprehending the biology of tumors has facilitated the identification of fresh targets, such as inhibitors targeting different pathways like phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR), cell-cycle regulation, heat shock protein, and epigenetic pathways, demonstrating encouraging outcomes in clinical experiments. For example, the mTOR inhibitor everolimus has been sanctioned for ER+ breast cancer and resistance to aromatase inhibitors in the advanced or metastatic phase. Triple-negative breast cancer, characterized by the absence of estrogen receptors, progesterone receptors, and HER2, currently lacks established targeted therapies. While poly (ADP-ribose) polymerase inhibitors exhibit effectiveness in BRCA-related cancers, their efficiency in addressing triple-negative breast cancer residues is uncertain. This paper furnishes a comprehensive outline of the principal targeted therapies presently employed or under exploration for breast cancer treatment within the three clinical subsets.
Collapse
Affiliation(s)
- Mahyar Haki
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Reihaneh Bayat
- Shahid Beheshti University of Medical Sciences and Health Services, Taleghani Hospital, Tehran, Iran.
| |
Collapse
|
6
|
Agostini M, Mandrioli A, Zamagni C. Fulvestrant Monotherapy After CDK4/6 Inhibitors in Metastatic Breast Cancer Patients: A Real-Life Experience. Cancers (Basel) 2024; 16:4179. [PMID: 39766078 PMCID: PMC11674525 DOI: 10.3390/cancers16244179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The treatment of hormone receptor positive (HR+), HER-2 negative metastatic breast cancer (MBC) has radically changed over the last few years. CDK4/6 inhibitors combined with endocrine therapy have become the standard of care as a front-line therapeutic approach, conferring a significant improvement in progression-free survival and overall survival compared to traditional endocrine therapy (ET) alone. However, the wide administration of these drugs in clinical practice paved the way for the emergence of new intrinsic and acquired mechanisms of resistance that seem to compromise second-line treatment effectiveness. In this context, fulvestrant monotherapy appears disqualified. Materials and Methods: we evaluated a population of 30 women currently treated in our oncology unit with HR+/HER2- metastatic breast cancer, receiving fulvestrant 500 mg every 28 days after progression to first-line therapy with CDK 4/6 inhibitors combined with aromatase inhibitors. Results: Of 30 patients observed, 23 progressed to fulvestrant with a median PFS of 3.7 months (range 1-9.7 months). Conclusions: our real-life experience suggests that second-line fulvestrant monotherapy confers very poor disease control and is quite an inadequate therapeutic option. CDK4/6i administration beyond progression could possibly be considered as more valid option, in the absence of targetable mutations or newer, more effective drugs.
Collapse
Affiliation(s)
- Margherita Agostini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Anna Mandrioli
- Breast and Gyncological Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Claudio Zamagni
- Breast and Gyncological Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| |
Collapse
|
7
|
Neven P, Stahl N, Vidal M, Martín M, Kaufman PA, Harbeck N, Hunt KK, Carter S, Bidard FC, Fasching PA, Aftimos P, Wheatley D, Hamilton E, Aft R, Kulkarni S, Schmid P, Bhave M, Ismail-Khan R, Karacsonyi C, Estrem ST, Nguyen B, Ozbek U, Yuen E, Rodrik-Outmezguine V, Ciruelos E. A Preoperative Window-of-Opportunity Study of Oral SERD, Imlunestrant, in Newly Diagnosed ER-Positive, HER2-Negative Early Breast Cancer: Results from the EMBER-2 Study. Clin Cancer Res 2024; 30:5304-5313. [PMID: 39377773 PMCID: PMC11609621 DOI: 10.1158/1078-0432.ccr-24-2113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE Imlunestrant is an oral selective estrogen receptor degrader with favorable safety and preliminary efficacy in patients with advanced breast cancer. Pharmacodynamic (PD) biomarker data can optimize drug dosing; in this study, we present PD data from the EMBER-2 study. PATIENTS AND METHODS Postmenopausal women with untreated, operable estrogen receptor (ER)-positive, HER2-negative early breast cancer were randomized to 400 versus 800 mg of imlunestrant daily for ∼2 weeks before surgery. A single arm study tested a daily dose of 200 mg. PD biomarker changes (ER, progesterone receptor, Ki-67 by IHC, and mRNA expression of ER-related genes) were evaluated in paired tumor samples (pre-/posttreatment). Safety and pharmacokinetics were also assessed. RESULTS Among evaluable paired samples (n = 75), PD profiles demonstrated consistent ER targeting between 400- and 800-mg doses, with less toxicity at the 400-mg dose. Although inducing the lowest rate of complete cell-cycle arrest, PD and pharmacokinetic results were similar for the 200-mg dose. CONCLUSIONS EMBER-2 combined with existing phase I data has identified 400 mg as the optimal imlunestrant dose.
Collapse
Affiliation(s)
- Patrick Neven
- Leuven Cancer Institute, Universitaire Ziekenhuizen (UZ), Leuven, Belgium
| | | | - Maria Vidal
- Department of Medical Oncology, Hospital Clinic, Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Miguel Martín
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | | | - Nadia Harbeck
- Breast Center, Department of Obstetrics and Gynecology, and Comprehensive Cancer Center CCCLMU, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Kelly K. Hunt
- Department of Breast Surgical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Stacey Carter
- Division of Surgical Oncology, Baylor College of Medicine, Lester and Sue Smith Breast Center, Dan L. Duncan Comprehensive Cancer Center, Houston, Texas
| | | | - Peter A. Fasching
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Philippe Aftimos
- Department of Medical Oncology, Institute Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | - Rebecca Aft
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Swati Kulkarni
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Peter Schmid
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Manali Bhave
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | | | | | | | - Bastien Nguyen
- Loxo@Lilly, Lilly Corporate Center, Indianapolis, Indiana
| | - Umut Ozbek
- Eli Lilly and Company, Indianapolis, Indiana
| | - Eunice Yuen
- Eli Lilly and Company, Indianapolis, Indiana
| | | | - Eva Ciruelos
- Medical Oncology Department, Breast Cancer Unit, University Hospital 12 de Octubre, Madrid, Spain
- Medical Oncology Department, Breast Cancer Unit, HM Hospitales, Madrid, Spain
| |
Collapse
|
8
|
O'Brien SR, Edmonds CE, Ward RE, Taunk NK, Pantel AR, Mankoff DA. Update on 18F-Fluoroestradiol. Semin Nucl Med 2024; 54:812-826. [PMID: 39368910 DOI: 10.1053/j.semnuclmed.2024.09.001] [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: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 10/07/2024]
Abstract
18F-16α-Fluoroestradiol (18F-FES) is a radiolabeled estrogen analogue positron emission tomography (PET) imaging agent that binds to the estrogen receptor (ER) in the nucleus of ER-expressing cells. Proof-of-concept studies of 18F-FES demonstrated expected correlation between tumoral 18F-FES-positivity on PET-imaging and ER+ status assessed on biopsy samples by radioligand binding and immunohistochemistry. After decades of study, 18F-FES PET/CT gained clinical approval in 2016 in France and 2020 in the United States for use in patients with ER+ metastatic or recurrent breast cancer. ER+ as assessed by 18F-FES PET/CT has been shown to serve as a biomarker, identifying metastatic breast cancer patients who may respond to endocrine therapy and those who are unlikely to respond. In 2023, the Society of Nuclear Medicine and Molecular Imaging (SNMMI) published Appropriate Use Criteria for 18F-FES PET/CT, identifying four indications in which use of 18F-FES PET/CT was "appropriate": (1) To assess functional ER status in metastatic lesions unfavorable to biopsy or when biopsy is nondiagnostic, (2) To detect ER status when other imaging tests are equivocal or suspicious, and at (3) initial diagnosis of metastatic disease or (4) progression of metastatic disease, for considering endocrine therapy. This article reviews the foundations of 18F-FES imaging, including normal distribution, false positives, and false negatives, and describes the most up-to-date clinical uses as well as emerging research in breast cancer and other patient populations.
Collapse
Affiliation(s)
- Sophia R O'Brien
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA; Department of Radiology, Division of Breast Imaging, Hospital of the University of Pennsylvania, Philadelphia, PA.
| | - Christine E Edmonds
- Department of Radiology, Division of Breast Imaging, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Rebecca E Ward
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Neil K Taunk
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Austin R Pantel
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - David A Mankoff
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
9
|
Turner NC, Im SA, Saura C, Juric D, Loibl S, Kalinsky K, Schmid P, Loi S, Sunpaweravong P, Musolino A, Li H, Zhang Q, Nowecki Z, Leung R, Thanopoulou E, Shankar N, Lei G, Stout TJ, Hutchinson KE, Schutzman JL, Song C, Jhaveri KL. Inavolisib-Based Therapy in PIK3CA-Mutated Advanced Breast Cancer. N Engl J Med 2024; 391:1584-1596. [PMID: 39476340 DOI: 10.1056/nejmoa2404625] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
BACKGROUND Inavolisib is a highly potent and selective inhibitor of the alpha isoform of the p110 catalytic subunit of the phosphatidylinositol 3-kinase complex (encoded by PIK3CA) that also promotes the degradation of mutated p110α. Inavolisib plus palbociclib-fulvestrant has shown synergistic activity in preclinical models and promising antitumor activity in early-phase trials. METHODS In a phase 3, double-blind, randomized trial, we compared first-line inavolisib (at an oral dose of 9 mg once daily) plus palbociclib-fulvestrant (inavolisib group) with placebo plus palbociclib-fulvestrant (placebo group) in patients with PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor 2 (HER2)-negative locally advanced or metastatic breast cancer who had had relapse during or within 12 months after the completion of adjuvant endocrine therapy. The primary end point was progression-free survival as assessed by the investigator. RESULTS A total of 161 patients were assigned to the inavolisib group and 164 to the placebo group; the median follow-up was 21.3 months and 21.5 months, respectively. The median progression-free survival was 15.0 months (95% confidence interval [CI], 11.3 to 20.5) in the inavolisib group and 7.3 months (95% CI, 5.6 to 9.3) in the placebo group (hazard ratio for disease progression or death, 0.43; 95% CI, 0.32 to 0.59; P<0.001). An objective response occurred in 58.4% of the patients in the inavolisib group and in 25.0% of those in the placebo group. The incidence of grade 3 or 4 neutropenia was 80.2% in the inavolisib group and 78.4% in the placebo group; grade 3 or 4 hyperglycemia, 5.6% and 0%, respectively; grade 3 or 4 stomatitis or mucosal inflammation, 5.6% and 0%; and grade 3 or 4 diarrhea, 3.7% and 0%. No grade 3 or 4 rash was observed. Discontinuation of any trial agent because of adverse events occurred in 6.8% of the patients in the inavolisib group and in 0.6% of those in the placebo group. CONCLUSIONS In patients with PIK3CA-mutated, hormone receptor-positive, HER2-negative locally advanced or metastatic breast cancer, inavolisib plus palbociclib-fulvestrant led to significantly longer progression-free survival than placebo plus palbociclib-fulvestrant, with a greater incidence of toxic effects. The percentage of patients who discontinued any trial agent because of adverse events was low. (Funded by F. Hoffmann-La Roche; INAVO120 ClinicalTrials.gov number, NCT04191499.).
Collapse
Affiliation(s)
- Nicholas C Turner
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Seock-Ah Im
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Cristina Saura
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Dejan Juric
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Sibylle Loibl
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Kevin Kalinsky
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Peter Schmid
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Sherene Loi
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Patrapim Sunpaweravong
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Antonino Musolino
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Huiping Li
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Qingyuan Zhang
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Zbigniew Nowecki
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Roland Leung
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Eirini Thanopoulou
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Noopur Shankar
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Guiyuan Lei
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Thomas J Stout
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Katherine E Hutchinson
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Jennifer L Schutzman
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Chunyan Song
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| | - Komal L Jhaveri
- From the Royal Marsden Hospital and Institute of Cancer Research (N.C.T.) and the Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London (P. Schmid), London, and Roche, Welwyn Garden City (E.T., G.L.) - all in the United Kingdom; Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, South Korea (S.-A.I.); Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona (C. Saura); Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston (D.J.); Winship Cancer Institute at Emory University, Atlanta (K.K.); Genentech, San Francisco (N.S., T.J.S., K.E.H., J.L.S., C. Song); the Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College - both in New York (K.L.J.); the German Breast Group, Neu-Isenburg, and the Center for Hematology and Oncology Bethanien, Goethe University, Frankfurt - both in Germany (S. Loibl); the Division of Cancer Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, and the Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, VIC - both in Australia (S. Loi); the Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand (P. Sunpaweravong); the Department of Medicine, University of Parma, Parma, and the Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori," Meldola - both in Italy (A.M.); the Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing (H.L.), Harbin Medical University, Harbin (Q.Z.), and the University Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (R.L.) - all in China; and Maria Skłodowska-Curie Institute of Oncology, Warsaw, Poland (Z.N.)
| |
Collapse
|
10
|
Huang H, Wei T, Zhang A, Zhang H, Kong L, Li Y, Li F. Trends in the incidence and survival of women with hormone receptor-positive breast cancer from 1990 to 2019: a large population-based analysis. Sci Rep 2024; 14:23690. [PMID: 39390094 PMCID: PMC11467179 DOI: 10.1038/s41598-024-74746-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
Hormone receptor-positive breast cancer (BC) is the most prevalent subtype of BC and is generally correlated with a favorable prognosis. This study aimed to determine the incidence and survival trends among women diagnosed with hormone receptor-positive BC between 1990 and 2019. Female patients with hormone receptor-positive BC for calendar years 1990-2019 were obtained from the Surveillance, Epidemiology, and End Results (SEER) database and categorized into six diagnostic groups according to the year of diagnosis. Age-adjusted incidence rates (IRs) were calculated using joinpoint regression. We used the Kaplan-Meier method and multivariate Cox regression analyses to determine the association between diagnostic groups, and overall survival (OS) and BC-specific survival (BCSS). The final analysis included 370,729 women, among whom 37,943 (10.2%), 49,266 (13.3%), 55,652 (15.0%), 64,451 (17.4%), 77,127 (20.8%), and 86,290 (23.3%) were diagnosed between 1990 and 1994, 1995-1999, 2000-2004, 2005-2009, 2010-2014, and 2015-2019, respectively. Within the overall cohort, IRs gradually increased from 70 per 100,000 in 1990 to 113 per 100,000 in 2019 (average annual percent change, 1.59%; 95% CI, 1.18-1.99). Multivariate Cox regression analysis revealed that the survival outcomes gradually improved over nearly three decades among hormone receptor-positive BC patients, with a 0.8% and 1.3% decrease in risk for all-cause and BC-specific mortality each year, respectively. Compared to 1990-1994, hormone receptor-positive BC patients diagnosed in 2015-2019 had a 22% lower risk of all-cause death (hazard ratio [HR], 0.78; 95% CI, 0.76-0.81) and a 27% lower risk of BC-specific death (HR, 0.73; 95% CI, 0.70-0.76). The development of treatment strategies within the past three decades, especially endocrine therapy, may contribute to the continuous improvement of clinical outcomes in patients with hormone receptor-positive BC.
Collapse
Affiliation(s)
- Hongbo Huang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Tingting Wei
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Aijie Zhang
- Health Management Center of University-Town Hospital Affiliated to Chongqing Medical University, Chongqing, China
| | - Heng Zhang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Lingquan Kong
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Yunhai Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| | - Fan Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
11
|
Gough SM, Flanagan JJ, Teh J, Andreoli M, Rousseau E, Pannone M, Bookbinder M, Willard R, Davenport K, Bortolon E, Cadelina G, Gordon D, Pizzano J, Macaluso J, Soto L, Corradi J, Digianantonio K, Drulyte I, Morgan A, Quinn C, Békés M, Ferraro C, Chen X, Wang G, Dong H, Wang J, Langley DR, Houston J, Gedrich R, Taylor IC. Oral Estrogen Receptor PROTAC Vepdegestrant (ARV-471) Is Highly Efficacious as Monotherapy and in Combination with CDK4/6 or PI3K/mTOR Pathway Inhibitors in Preclinical ER+ Breast Cancer Models. Clin Cancer Res 2024; 30:3549-3563. [PMID: 38819400 PMCID: PMC11325148 DOI: 10.1158/1078-0432.ccr-23-3465] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 04/11/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
PURPOSE Estrogen receptor (ER) alpha signaling is a known driver of ER-positive (ER+)/human epidermal growth factor receptor 2 negative (HER2-) breast cancer. Combining endocrine therapy (ET) such as fulvestrant with CDK4/6, mTOR, or PI3K inhibitors has become a central strategy in the treatment of ER+ advanced breast cancer. However, suboptimal ER inhibition and resistance resulting from the ESR1 mutation dictates that new therapies are needed. EXPERIMENTAL DESIGN A medicinal chemistry campaign identified vepdegestrant (ARV-471), a selective, orally bioavailable, and potent small molecule PROteolysis-TArgeting Chimera (PROTAC) degrader of ER. We used biochemical and intracellular target engagement assays to demonstrate the mechanism of action of vepdegestrant, and ESR1 wild-type (WT) and mutant ER+ preclinical breast cancer models to demonstrate ER degradation-mediated tumor growth inhibition (TGI). RESULTS Vepdegestrant induced ≥90% degradation of wild-type and mutant ER, inhibited ER-dependent breast cancer cell line proliferation in vitro, and achieved substantial TGI (87%-123%) in MCF7 orthotopic xenograft models, better than those of the ET agent fulvestrant (31%-80% TGI). In the hormone independent (HI) mutant ER Y537S patient-derived xenograft (PDX) breast cancer model ST941/HI, vepdegestrant achieved tumor regression and was similarly efficacious in the ST941/HI/PBR palbociclib-resistant model (102% TGI). Vepdegestrant-induced robust tumor regressions in combination with each of the CDK4/6 inhibitors palbociclib, abemaciclib, and ribociclib; the mTOR inhibitor everolimus; and the PI3K inhibitors alpelisib and inavolisib. CONCLUSIONS Vepdegestrant achieved greater ER degradation in vivo compared with fulvestrant, which correlated with improved TGI, suggesting vepdegestrant could be a more effective backbone ET for patients with ER+/HER2- breast cancer.
Collapse
Affiliation(s)
| | | | - Jessica Teh
- Arvinas Operations, Inc., New Haven, Connecticut.
| | | | | | | | | | - Ryan Willard
- Arvinas Operations, Inc., New Haven, Connecticut.
| | | | | | | | | | | | | | - Leofal Soto
- Arvinas Operations, Inc., New Haven, Connecticut.
| | - John Corradi
- Arvinas Operations, Inc., New Haven, Connecticut.
| | | | - Ieva Drulyte
- Thermo Fisher Scientific, Materials and Structural Analysis, Eindhoven, Netherlands.
| | | | - Connor Quinn
- Arvinas Operations, Inc., New Haven, Connecticut.
| | - Miklós Békés
- Arvinas Operations, Inc., New Haven, Connecticut.
| | | | - Xin Chen
- Arvinas Operations, Inc., New Haven, Connecticut.
| | - Gan Wang
- Arvinas Operations, Inc., New Haven, Connecticut.
| | - Hanqing Dong
- Arvinas Operations, Inc., New Haven, Connecticut.
| | - Jing Wang
- Arvinas Operations, Inc., New Haven, Connecticut.
| | | | - John Houston
- Arvinas Operations, Inc., New Haven, Connecticut.
| | | | | |
Collapse
|
12
|
Qureshi Z, Jamil A, Altaf F, Siddique R, Adilovic E, Fatima E, Shah S. Elacestrant in the treatment landscape of ER-positive, HER2-negative, ESR1-mutated advanced breast cancer: a contemporary narrative review. Ann Med Surg (Lond) 2024; 86:4624-4633. [PMID: 39118705 PMCID: PMC11305773 DOI: 10.1097/ms9.0000000000002293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/11/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer with ESR1 mutations presents a significant therapeutic challenge due to its adaptive resistance mechanisms to chemotherapy, especially endocrine treatment. Elacestrant, a novel oral selective estrogen receptor degrader (SERD), has emerged as a promising agent in this treatment-resistant era. Method A comprehensive search was conducted on pivotal clinical trials, including the RAD1901-005 Trial, EMERALD TRIAL, ELIPSE, and ELEVATE, focusing on their methodologies, patient populations, treatment regimens, and outcomes. Discussion This narrative review describes the available preclinical and clinical evidence on elacestrant, focusing on its pharmacodynamics, pharmacokinetics, efficacy, and safety within the existing literature. Elacestrant has demonstrated excellent activity against ESR1 mutations associated with resistance to first-line endocrine therapies. Clinical trials have shown improved progression-free survival in patients with advanced ER+/HER2-, ESR1-mutated breast cancer. Safety profiles indicate a tolerable side effect spectrum consistent with other agents. Its oral bioavailability offers a convenient alternative to injectable SERDs, with potential implications for patient adherence and quality of life. The review also discusses the comparative efficacy of elacestrant relative to existing endocrine therapies and its possible use in combination regimens. Conclusion Ongoing clinical trials assessing elacestrant and other SERDs will yield data that might aid clinicians in determining the optimal selection and order of endocrine treatment drugs for ER+ breast cancer. The integration of targeted and immunotherapeutic agents with traditional chemotherapy represents a pivotal shift in Breast Cancer treatment, moving towards more personalized and effective regimens.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University
| | - Abdur Jamil
- Department of Medicine, Samaritan Medical Centre
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY, USA
| | | | | | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Shivendra Shah
- Department of Medicine, Nepalgunj Medical College, Chisapani, Nepal
| |
Collapse
|
13
|
Al-Ruwishan A, Amer B, Salem A, Abdi A, Chimpandu N, Esa A, Melemenis A, Saleem MZ, Mathew R, Gamallat Y. Advancements in Understanding the Hide-and-Seek Strategy of Hibernating Breast Cancer Cells and Their Implications in Oncology from a Broader Perspective: A Comprehensive Overview. Curr Issues Mol Biol 2024; 46:8340-8367. [PMID: 39194709 DOI: 10.3390/cimb46080492] [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: 06/10/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Despite recent advancements in technology, breast cancer still poses a significant threat, often resulting in fatal consequences. While early detection and treatments have shown some promise, many breast cancer patients continue to struggle with the persistent fear of the disease returning. This fear is valid, as breast cancer cells can lay dormant for years before remerging, evading traditional treatments like a game of hide and seek. The biology of these dormant breast cancer cells presents a crucial yet poorly understood challenge in clinical settings. In this review, we aim to explore the mysterious world of dormant breast cancer cells and their significant impact on patient outcomes and prognosis. We shed light on the elusive role of the G9a enzyme and many other epigenetic factors in breast cancer recurrence, highlighting its potential as a target for eliminating dormant cancer cells and preventing disease relapse. Through this comprehensive review, we not only emphasise the urgency of unravelling the dynamics of dormant breast cancer cells to improve patient outcomes and advance personalised oncology but also provide a guide for fellow researchers. By clearly outlining the clinical and research gaps surrounding dormant breast cancer cells from a molecular perspective, we aim to inspire further exploration of this critical area, ultimately leading to improved patient care and treatment strategies.
Collapse
Affiliation(s)
- Aiman Al-Ruwishan
- Space for Research Initiative, Research Horizons, London NW10 2PU, UK
| | - Bushra Amer
- Department of Family Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Ahmed Salem
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, 53210 Pardubice, Czech Republic
| | - Ahmed Abdi
- Independent Researcher, Uxbridge UB9 6JH, UK
| | | | | | | | - Muhammad Zubair Saleem
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Roselit Mathew
- Department of Oncology, Biochemistry and Molecular Biology, and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Yaser Gamallat
- Department of Oncology, Biochemistry and Molecular Biology, and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
14
|
Ferro A, Campora M, Caldara A, De Lisi D, Lorenzi M, Monteverdi S, Mihai R, Bisio A, Dipasquale M, Caffo O, Ciribilli Y. Novel Treatment Strategies for Hormone Receptor (HR)-Positive, HER2-Negative Metastatic Breast Cancer. J Clin Med 2024; 13:3611. [PMID: 38930141 PMCID: PMC11204965 DOI: 10.3390/jcm13123611] [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: 05/09/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Estrogen receptor (ER)-positive breast cancer (BC) is the most common BC subtype. Endocrine therapy (ET) targeting ER signaling still remains the mainstay treatment option for hormone receptor (HR)-positive BC either in the early or in advanced setting, including different strategies, such as the suppression of estrogen production or directly blocking the ER pathway through SERMs-selective estrogen receptor modulators-or SERDs-selective estrogen receptor degraders. Nevertheless, the development of de novo or acquired endocrine resistance still remains challenging for oncologists. The use of novel ET combined with targeted drugs, such as cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors, has significantly improved long-term outcome rates, thus changing the therapeutic algorithm for metastatic BC (MBC) and recently the therapeutic strategy in the adjuvant setting for early high-risk BC. Eluding the resistance to CDK4/6 inhibitors combined with ET is currently an unmet medical need, and there is disagreement concerning the best course of action for patients who continue to progress after this combination approach. Genetic changes in the tumor along its growth uncovered by genomic profiling of recurrent and/or metastatic lesions through tumor and/or liquid biopsies may predict the response or resistance to specific agents, suggesting the best therapeutic strategy for each patient by targeting the altered ER-dependent pathway (novel oral SERDs and a new generation of anti-estrogen agents) or alternative ER-independent signaling pathways such as PI3K/AKT/mTOR or tyrosine kinase receptors (HER2 mutations or HER2 low status) or by inhibiting pathways weakened through germline BRCA1/2 mutations. These agents are being investigated as single molecules and in combination with other target therapies, offering promising weapons to overcome or avoid treatment failure and propose increasingly more personalized treatment approaches. This review presents novel insights into ET and other targeted therapies for managing metastatic HR+/HER2- BC by exploring potential strategies based on clinical evidence and genomic profiling following the failure of the CDK4/6i and ET combination.
Collapse
Affiliation(s)
- Antonella Ferro
- Medical Oncology and Breast Unit, Santa Chiara Hospital, APSS Trento, 38122 Trento, Italy; (A.C.); (D.D.L.); (M.L.); (S.M.); (M.D.)
| | - Michela Campora
- Department of Pathology, Santa Chiara Hospital, APSS Trento, 38122 Trento, Italy;
| | - Alessia Caldara
- Medical Oncology and Breast Unit, Santa Chiara Hospital, APSS Trento, 38122 Trento, Italy; (A.C.); (D.D.L.); (M.L.); (S.M.); (M.D.)
| | - Delia De Lisi
- Medical Oncology and Breast Unit, Santa Chiara Hospital, APSS Trento, 38122 Trento, Italy; (A.C.); (D.D.L.); (M.L.); (S.M.); (M.D.)
| | - Martina Lorenzi
- Medical Oncology and Breast Unit, Santa Chiara Hospital, APSS Trento, 38122 Trento, Italy; (A.C.); (D.D.L.); (M.L.); (S.M.); (M.D.)
| | - Sara Monteverdi
- Medical Oncology and Breast Unit, Santa Chiara Hospital, APSS Trento, 38122 Trento, Italy; (A.C.); (D.D.L.); (M.L.); (S.M.); (M.D.)
| | - Raluca Mihai
- Department of Pathology, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK;
| | - Alessandra Bisio
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (A.B.); (Y.C.)
| | - Mariachiara Dipasquale
- Medical Oncology and Breast Unit, Santa Chiara Hospital, APSS Trento, 38122 Trento, Italy; (A.C.); (D.D.L.); (M.L.); (S.M.); (M.D.)
| | - Orazio Caffo
- Medical Oncology, Santa Chiara Hospital, APSS Trento, 38122 Trento, Italy;
| | - Yari Ciribilli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (A.B.); (Y.C.)
| |
Collapse
|
15
|
Zhao Y, Wang S, Lv S, Liu X, Li W, Song Y, Rong D, Zheng P, Huang H, Zheng H. Combined oral low-dose cyclophosphamide endocrine therapy may improve clinical response among patients with metastatic breast cancer via Tregs in TLSs. Sci Rep 2024; 14:13432. [PMID: 38862586 PMCID: PMC11166640 DOI: 10.1038/s41598-024-64042-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/04/2024] [Indexed: 06/13/2024] Open
Abstract
Despite limited research on refractory and/or endocrine therapy failure in elderly metastatic breast cancer (MBC) patients, a prior study showed that low-dose oral cyclophosphamide (CY) can improve the overall survival rate of MBC patients, possibly through the immunoregulation of regulatory T cells (Tregs). We preliminarily investigated the combination of endocrine therapy (ET) with oral low-dose CY as salvage therapy in elderly patients via peripheral blood regulatory T-cell analyses. In addition, we evaluated the associations of tumor tertiary lymphoid structures (TLSs) with therapeutic outcomes. HR+/HER2- advanced breast cancer patients who received low-dose CY combined with ET or ET only from April 2015 to August 2021 were enrolled in this retrospective study. The primary outcome was the clinical control rate (CCR), and the secondary outcome was progression-free survival (PFS). Circulating T lymphocyte subpopulations represented by Tregs were monitored during treatment by flow cytometry methods. TLSs wereconfirmed by hematoxylin-eosin staining of pretreatment specimens, and CD3, CD4, and Foxp3 were detected using Opal multicolor immunofluorescence. A total of 85 patients who received CY + ET and 50 patients who received ET only were enrolled, the percentage of patients who received CCR was 73% (62/85) vs. 70% (45/50), and the objective response rate (ORR) was 28% (24/85) vs. 24% (12/50). No deaths occurred during the study period. The mean PFS time was 13 vs. 11 months (P = 0.03). In the CY + ET group, decreases in CD4+/CD25+/Foxp3+ T cells (P < 0.001) were favorable for both clinical control and prolonged PFS (P < 0.001). Compared with patients without TLSs, those with TLSs were more likely to have better clinical control and PFS (mean time = 6 months), and a greater number of Treg cells during TLS pretreatment correlated with longer PFS (P = 0.043). Oral low-dose CY combined with standard ET exerts immunological effects by decreasing Treg levels to achieve improved clinical responses. Moreover, patients with TLSs might benefit more from such therapy than those without TLSs, and a high Treg cell count in TLSs before treatment predicts better therapeutic efficacy.
Collapse
Affiliation(s)
- Yuze Zhao
- Department of Medical Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, 10 Tieyi Rd, Beijing, 100038, China
| | - Shuo Wang
- Department of Medical Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, 10 Tieyi Rd, Beijing, 100038, China
| | - Shuzhen Lv
- Breast Department, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xiaojun Liu
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Weiping Li
- Department of Pharmacology, Shanxi Medical University Fenyang College, Fenyang, 032200, China
| | - Yuguang Song
- Department of Medical Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, 10 Tieyi Rd, Beijing, 100038, China
| | - Dongwen Rong
- Department of Medical Oncology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Peiming Zheng
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Hongyan Huang
- Department of Medical Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, 10 Tieyi Rd, Beijing, 100038, China.
| | - Huixia Zheng
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
16
|
Lainé M, Greene ME, Kurleto JD, Bozek G, Leng T, Huggins RJ, Komm BS, Greene GL. Lasofoxifene as a potential treatment for aromatase inhibitor-resistant ER-positive breast cancer. Breast Cancer Res 2024; 26:95. [PMID: 38849889 PMCID: PMC11161925 DOI: 10.1186/s13058-024-01843-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/17/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Breast cancers treated with aromatase inhibitors (AIs) can develop AI resistance, which is often driven by estrogen receptor-alpha (ERα/ESR1) activating mutations, as well as by ER-independent signaling pathways. The breast ER antagonist lasofoxifene, alone or combined with palbociclib, elicited antitumor activities in a xenograft model of ER + metastatic breast cancer (mBC) harboring ESR1 mutations. The current study investigated the activity of LAS in a letrozole-resistant breast tumor model that does not have ESR1 mutations. METHODS Letrozole-resistant, MCF7 LTLT cells tagged with luciferase-GFP were injected into the mammary duct inguinal glands of NSG mice (MIND model; 6 mice/group). Mice were randomized to vehicle, lasofoxifene ± palbociclib, fulvestrant ± palbociclib, or palbociclib alone 2-3 weeks after cell injections. Tumor growth and metastases were monitored with in vivo and ex vivo luminescence imaging, terminal tumor weight measurements, and histological analysis. The experiment was repeated with the same design and 8-9 mice in each treatment group. RESULTS Western blot analysis showed that the MCF7 LTLT cells had lower ERα and higher HER2 expressions compared with normal MCF7 cells. Lasofoxifene ± palbociclib, but not fulvestrant, significantly reduced primary tumor growth versus vehicle as assessed by in vivo imaging of tumors at study ends. Percent tumor area in excised mammary glands was significantly lower for lasofoxifene plus palbociclib versus vehicle. Ki67 staining showed decreased overall tumor cell proliferation with lasofoxifene ± palbociclib. The lasofoxifene + palbociclib combination was also associated with significantly fewer bone metastases compared with vehicle. Similar results were observed in the repeat experiment. CONCLUSIONS In a mouse model of letrozole-resistant breast cancer with no ESR1 mutations, reduced levels of ERα, and overexpression of HER2, lasofoxifene alone or combined with palbociclib inhibited primary tumor growth more effectively than fulvestrant. Lasofoxifene plus palbociclib also reduced bone metastases. These results suggest that lasofoxifene alone or combined with a CDK4/6 inhibitor may offer benefits to patients who have ER-low and HER2-positive, AI-resistant breast cancer, independent of ESR1 mutations.
Collapse
Affiliation(s)
- Muriel Lainé
- The Ben May Department for Cancer Research, The University of Chicago, 929 East 57th Street GCIS W421C, Chicago, IL, 60637, USA
| | - Marianne E Greene
- The Ben May Department for Cancer Research, The University of Chicago, 929 East 57th Street GCIS W421C, Chicago, IL, 60637, USA
| | - Justyna D Kurleto
- The Ben May Department for Cancer Research, The University of Chicago, 929 East 57th Street GCIS W421C, Chicago, IL, 60637, USA
| | - Grazyna Bozek
- The Ben May Department for Cancer Research, The University of Chicago, 929 East 57th Street GCIS W421C, Chicago, IL, 60637, USA
| | - Tiffany Leng
- The Ben May Department for Cancer Research, The University of Chicago, 929 East 57th Street GCIS W421C, Chicago, IL, 60637, USA
| | - Rosemary J Huggins
- The Ben May Department for Cancer Research, The University of Chicago, 929 East 57th Street GCIS W421C, Chicago, IL, 60637, USA
| | | | - Geoffrey L Greene
- The Ben May Department for Cancer Research, The University of Chicago, 929 East 57th Street GCIS W421C, Chicago, IL, 60637, USA.
| |
Collapse
|
17
|
Corti C, Batra-Sharma H, Kelsten M, Shatsky RA, Garrido-Castro AC, Gradishar WJ. Systemic Therapy in Breast Cancer. Am Soc Clin Oncol Educ Book 2024; 44:e432442. [PMID: 39013124 DOI: 10.1200/edbk_432442] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Therapeutic advances in breast cancer have significantly improved outcomes in recent decades. In the early setting, there has been a gradual shift from adjuvant-only to neoadjuvant strategies, with a growing focus on customizing post-neoadjuvant treatments through escalation and de-escalation based on pathologic response. At the same time, the transition from a pre-genomic to a post-genomic era, utilizing specific assays in the adjuvant setting and targeted sequencing in the advanced stage, has deepened our understanding of disease biology and aided in identifying molecular markers associated with treatment benefit. Finally, the introduction of new drug classes such as antibody-drug conjugates, and the incorporation in the (neo)adjuvant setting of therapies previously investigated in the advanced stage, like immunotherapy and CDK4-6 inhibitors, poses new challenges in treatment sequencing.
Collapse
Affiliation(s)
- Chiara Corti
- Dana Farber Cancer Institute, Harvard University, Boston, MA
| | | | - Max Kelsten
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | | | - William J Gradishar
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| |
Collapse
|
18
|
Sharaf B, Hajahjeh A, Bani Hani H, Abdel-Razeq H. Next generation selective estrogen receptor degraders in postmenopausal women with advanced-stage hormone receptors-positive, HER2-negative breast cancer. Front Oncol 2024; 14:1385577. [PMID: 38800404 PMCID: PMC11116652 DOI: 10.3389/fonc.2024.1385577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Breast cancer is the most prevalent malignancy in women, and is characterized by its heterogeneity; exhibiting various subgroups identifiable through molecular biomarkers that also serve as predictive indicators. More than two thirds of breast tumors are classified as luminal with positive hormone receptors (HR), indicating that cancer cells proliferation is promoted by hormones. Endocrine therapies play a vital role in the effective treatment of breast cancer by manipulating the signaling of estrogen receptors (ER), leading to a reduction in cell proliferation and growth rate. Selective estrogen receptor modulators (SERMs), such as tamoxifen and toremifene, function by blocking estrogen's effects. Aromatase inhibitors (AI), including anastrozole, letrozole and exemestane, suppress estrogen production. On the other hand, selective estrogen receptor degraders (SERDs), like fulvestrant, act by blocking and damaging estrogen receptors. Tamoxifen and AI are widely used both in early- and advanced-stage disease, while fulvestrant is used as a single agent or in combination with other agents like the cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors (palbociclib, abemaciclib, ribociclib) or alpelisib for advanced-stage disease. Currently, SERDs are recognized as an effective therapeutic approach for the treatment of ER-positive breast cancer, showing proficiency in reducing and blocking ER signaling. This review aims to outline the ongoing development of novel oral SERDs from a practical therapeutic perspective, enhancing our understanding of the mechanisms of action underlying these compounds.
Collapse
Affiliation(s)
- Baha’ Sharaf
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | | | - Hira Bani Hani
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
- School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
19
|
Vasseur A, Cabel L, Hego C, Takka W, Trabelsi Grati O, Renouf B, Lerebours F, Loirat D, Brain E, Cottu P, Sablin MP, Pierga JY, Callens C, Renault S, Bidard FC. Fulvestrant and everolimus efficacy after CDK4/6 inhibitor: a prospective study with circulating tumor DNA analysis. Oncogene 2024; 43:1214-1222. [PMID: 38413796 PMCID: PMC11014798 DOI: 10.1038/s41388-024-02986-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/29/2024]
Abstract
In a prospective study (NCT02866149), we assessed the efficacy of fulvestrant and everolimus in CDK4/6i pre-treated mBC patients and circulating tumor DNA (ctDNA) changes throughout therapy. Patients treated with fulvestrant and everolimus had their ctDNA assessed at baseline, after 3-5 weeks and at disease progression. Somatic mutations were identified in archived tumor tissues by targeted NGS and tracked in cell-free DNA by droplet digital PCR. ctDNA detection was then associated with clinicopathological characteristics and patients' progression-free survival (PFS), overall survival (OS) and best overall response (BOR). In the 57 included patients, median PFS and OS were 6.8 (95%CI [5.03-11.5]) and 38.2 (95%CI [30.0-not reached]) months, respectively. In 47 response-evaluable patients, BOR was a partial response or stable disease in 15 (31.9%) and 11 (23.4%) patients, respectively. Among patients with trackable somatic mutation and available plasma sample, N = 33/47 (70.2%) and N = 19/36 (52.8%) had ctDNA detected at baseline and at 3 weeks, respectively. ctDNA detection at baseline and PIK3CA mutation had an adverse prognostic impact on PFS and OS in multivariate analysis. This prospective cohort study documents the efficacy of fulvestrant and everolimus in CDK4/6i-pretreated ER + /HER2- mBC and highlights the clinical validity of early ctDNA changes as pharmacodynamic biomarker.
Collapse
Affiliation(s)
- Antoine Vasseur
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
- Circulating Tumor Biomarkers Laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France
| | - Luc Cabel
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
| | - Caroline Hego
- Circulating Tumor Biomarkers Laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France
| | - Wissam Takka
- Circulating Tumor Biomarkers Laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France
| | - Olfa Trabelsi Grati
- Department of Genetics, Institut Curie, Paris Sciences & Lettres University, Paris, France
| | | | - Florence Lerebours
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
| | - Delphine Loirat
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
| | - Etienne Brain
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
| | - Paul Cottu
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
| | - Marie-Paule Sablin
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
| | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France
- Université Paris Cité, Paris, France
| | - Céline Callens
- Department of Genetics, Institut Curie, Paris Sciences & Lettres University, Paris, France
| | - Shufang Renault
- Circulating Tumor Biomarkers Laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France.
| | - François-Clément Bidard
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud, France.
- Circulating Tumor Biomarkers Laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France.
- UVSQ, Paris-Saclay University, Saint Cloud, France.
| |
Collapse
|
20
|
Ma CX, Suman VJ, Sanati S, Vij K, Anurag M, Leitch AM, Unzeitig GW, Hoog J, Fernandez-Martinez A, Fan C, Gibbs RA, Watson MA, Dockter TJ, Hahn O, Guenther JM, Caudle A, Crouch E, Tiersten A, Mita M, Razaq W, Hieken TJ, Wang Y, Rimawi MF, Weiss A, Winer EP, Hunt KK, Perou CM, Ellis MJ, Partridge AH, Carey LA. Endocrine-Sensitive Disease Rate in Postmenopausal Patients With Estrogen Receptor-Rich/ERBB2-Negative Breast Cancer Receiving Neoadjuvant Anastrozole, Fulvestrant, or Their Combination: A Phase 3 Randomized Clinical Trial. JAMA Oncol 2024; 10:362-371. [PMID: 38236590 PMCID: PMC10797521 DOI: 10.1001/jamaoncol.2023.6038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/29/2023] [Indexed: 01/19/2024]
Abstract
Importance Adding fulvestrant to anastrozole (A+F) improved survival in postmenopausal women with advanced estrogen receptor (ER)-positive/ERBB2 (formerly HER2)-negative breast cancer. However, the combination has not been tested in early-stage disease. Objective To determine whether neoadjuvant fulvestrant or A+F increases the rate of pathologic complete response or ypT1-2N0/N1mic/Ki67 2.7% or less residual disease (referred to as endocrine-sensitive disease) over anastrozole alone. Design, Setting, and Participants A phase 3 randomized clinical trial assessing differences in clinical and correlative outcomes between each of the fulvestrant-containing arms and the anastrozole arm. Postmenopausal women with clinical stage II to III, ER-rich (Allred score 6-8 or >66%)/ERBB2-negative breast cancer were included. All analyses were based on data frozen on March 2, 2023. Interventions Patients received anastrozole, fulvestrant, or a combination for 6 months preoperatively. Tumor Ki67 was assessed at week 4 and optionally at week 12, and if greater than 10% at either time point, the patient switched to neoadjuvant chemotherapy or immediate surgery. Main Outcomes and Measures The primary outcome was the endocrine-sensitive disease rate (ESDR). A secondary outcome was the percentage change in Ki67 after 4 weeks of neoadjuvant endocrine therapy (NET) (week 4 Ki67 suppression). Results Between February 2014 and November 2018, 1362 female patients (mean [SD] age, 65.0 [8.2] years) were enrolled. Among the 1298 evaluable patients, ESDRs were 18.7% (95% CI, 15.1%-22.7%), 22.8% (95% CI, 18.9%-27.1%), and 20.5% (95% CI, 16.8%-24.6%) with anastrozole, fulvestrant, and A+F, respectively. Compared to anastrozole, neither fulvestrant-containing regimen significantly improved ESDR or week 4 Ki67 suppression. The rate of week 4 or week 12 Ki67 greater than 10% was 25.1%, 24.2%, and 15.7% with anastrozole, fulvestrant, and A+F, respectively. Pathologic complete response/residual cancer burden class I occurred in 8 of 167 patients and 17 of 167 patients, respectively (15.0%; 95% CI, 9.9%-21.3%), after switching to neoadjuvant chemotherapy due to week 4 or week 12 Ki67 greater than 10%. PAM50 subtyping derived from RNA sequencing of baseline biopsies available for 753 patients (58%) identified 394 luminal A, 304 luminal B, and 55 nonluminal tumors. A+F led to a greater week 4 Ki67 suppression than anastrozole alone in luminal B tumors (median [IQR], -90.4% [-95.2 to -81.9%] vs -76.7% [-89.0 to -55.6%]; P < .001), but not luminal A tumors. Thirty-six nonluminal tumors (65.5%) had a week 4 or week 12 Ki67 greater than 10%. Conclusions and Relevance In this randomized clinical trial, neither fulvestrant nor A+F significantly improved the 6-month ESDR over anastrozole in ER-rich/ERBB2-negative breast cancer. Aromatase inhibition remains the standard-of-care NET. Differential NET response by PAM50 subtype in exploratory analyses warrants further investigation. Trial Registration ClinicalTrials.gov Identifier: NCT01953588.
Collapse
Affiliation(s)
- Cynthia X. Ma
- Washington University School of Medicine, St Louis, Missouri
| | - Vera J. Suman
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota
| | - Souzan Sanati
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Kiran Vij
- Washington University School of Medicine, St Louis, Missouri
| | | | | | | | - Jeremy Hoog
- Washington University School of Medicine, St Louis, Missouri
| | | | - Cheng Fan
- University of North Carolina at Chapel Hill
| | | | - Mark A. Watson
- Washington University School of Medicine, St Louis, Missouri
| | - Travis J. Dockter
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota
| | - Olwen Hahn
- University of Chicago, Chicago, Illinois
| | | | | | - Erika Crouch
- Washington University School of Medicine, St Louis, Missouri
| | | | - Monica Mita
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Wajeeha Razaq
- University of Oklahoma Health Sciences Center, Oklahoma City
| | | | - Yang Wang
- Presbyterian Kaseman Hospital, Albuquerque, New Mexico
| | | | - Anna Weiss
- University of Rochester, Rochester, New York
| | | | | | | | | | | | | |
Collapse
|
21
|
Kingston B, Pearson A, Herrera-Abreu MT, Sim LX, Cutts RJ, Shah H, Moretti L, Kilburn LS, Johnson H, Macpherson IR, Ring A, Bliss JM, Hou Y, Toy W, Katzenellenbogen JA, Chandarlapaty S, Turner NC. ESR1 F404 Mutations and Acquired Resistance to Fulvestrant in ESR1-Mutant Breast Cancer. Cancer Discov 2024; 14:274-289. [PMID: 37982575 PMCID: PMC10850945 DOI: 10.1158/2159-8290.cd-22-1387] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 09/18/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023]
Abstract
Fulvestrant is used to treat patients with hormone receptor-positive advanced breast cancer, but acquired resistance is poorly understood. PlasmaMATCH Cohort A (NCT03182634) investigated the activity of fulvestrant in patients with activating ESR1 mutations in circulating tumor DNA (ctDNA). Baseline ESR1 mutations Y537S are associated with poor outcomes and Y537C with good outcomes. Sequencing of baseline and EOT ctDNA samples (n = 69) revealed 3/69 (4%) patients acquired novel ESR1 F404 mutations (F404L, F404I, and F404V), in cis with activating mutations. In silico modeling revealed that ESR1 F404 contributes to fulvestrant binding to estrogen receptor-alpha (ERα) through a pi-stacking bond, with mutations disrupting this bond. In vitro analysis demonstrated that single F404L, E380Q, and D538G models were less sensitive to fulvestrant, whereas compound mutations D538G + F404L and E380Q + F404L were resistant. Several oral ERα degraders were active against compound mutant models. We have identified a resistance mechanism specific to fulvestrant that can be targeted by treatments in clinical development. SIGNIFICANCE Novel F404 ESR1 mutations may be acquired to cause overt resistance to fulvestrant when combined with preexisting activating ESR1 mutations. Novel combinations of mutations in the ER ligand binding domain may cause drug-specific resistance, emphasizing the potential of similar drug-specific mutations to impact the efficacy of oral ER degraders in development. This article is featured in Selected Articles from This Issue, p. 201.
Collapse
Affiliation(s)
- Belinda Kingston
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Alex Pearson
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Maria Teresa Herrera-Abreu
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Li-Xuan Sim
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Rosalind J. Cutts
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Heena Shah
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Laura Moretti
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Lucy S. Kilburn
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Hannah Johnson
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Iain R. Macpherson
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Alistair Ring
- Breast Unit, The Royal Marsden Hospital, London, United Kingdom
| | - Judith M. Bliss
- Clinical Trials and Statistics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Yingwei Hou
- Department of Chemistry and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Weiyi Toy
- Memorial Sloan Kettering Cancer Center, New York City, New York
- Department of Medicine, Weill Cornell Medical College, New York City, New York
| | - John A. Katzenellenbogen
- Department of Chemistry and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Sarat Chandarlapaty
- Memorial Sloan Kettering Cancer Center, New York City, New York
- Department of Medicine, Weill Cornell Medical College, New York City, New York
| | - Nicholas C. Turner
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
- Breast Unit, The Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
22
|
Wang B, Ma M, Dai Y, Yu P, Ye L, Wang W, Sha C, Yang H, Yang Y, Zhu Y, Dong L, Wei S, Wang L, Tian J, Wang H. A novel scaffold long-acting selective estrogen receptor antagonist and degrader with superior preclinical profile against ER+ breast cancer. Eur J Med Chem 2024; 264:115934. [PMID: 38007911 DOI: 10.1016/j.ejmech.2023.115934] [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: 08/20/2023] [Revised: 10/12/2023] [Accepted: 11/03/2023] [Indexed: 11/28/2023]
Abstract
Breast cancer is one of the most common malignant tumors in women worldwide, with the majority of cases showing expression of estrogen receptors (ERs). Although drugs targeting ER have significantly improved survival rates in ER-positive patients, drug resistance remains an unmet clinical need. Fulvestrant, which overcomes selective estrogen receptor modulator (SERM) and AI (aromatase inhibitor) resistance, is currently the only long-acting selective estrogen receptor degrader (SERD) approved for both first and second-line settings. However, it fails to achieve satisfactory efficacy due to its poor solubility. Therefore, we designed and synthesized a series of novel scaffold (THC) derivatives, identifying their activities as ER antagonists and degraders. G-5b, the optimal compound, exhibited binding, antagonistic, degradation or anti-proliferative activities comparable to fulvestrant in ER+ wild type and mutants breast cancer cells. Notably, G-5b showed considerably improved stability and solubility. Research into the underlying mechanism indicated that G-5b engaged the proteasome pathway to degrade ER, subsequently inhibiting the ER signaling pathway and leading to the induction of apoptosis and cell cycle arrest events. Furthermore, G-5b displayed superior in vivo pharmacokinetics and pharmacodynamics properties, coupled with a favorable safety profile in the MCF-7 tamoxifen-resistant (MCF-7/TR) tumor xenograft model. Collectively, G-5b has emerged as a highly promising lead compound, offering potent antagonistic and degradation activities, positioning it as a novel long-acting SERD worthy of further refinement and optimization.
Collapse
Affiliation(s)
- Bingsi Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Mingxu Ma
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Yusen Dai
- R & D Center, Luye Pharma Group Ltd., Yantai, 264003, China
| | - Pengfei Yu
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Liang Ye
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Wenyan Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Chunjie Sha
- R & D Center, Luye Pharma Group Ltd., Yantai, 264003, China
| | - Huijie Yang
- R & D Center, Luye Pharma Group Ltd., Yantai, 264003, China
| | - Yingjie Yang
- R & D Center, Luye Pharma Group Ltd., Yantai, 264003, China
| | - Yunjing Zhu
- R & D Center, Luye Pharma Group Ltd., Yantai, 264003, China
| | - Lin Dong
- R & D Center, Luye Pharma Group Ltd., Yantai, 264003, China
| | - Shujuan Wei
- R & D Center, Luye Pharma Group Ltd., Yantai, 264003, China
| | - Linlin Wang
- Department of Food Engineering, Shandong Business Institute, Yantai, 264670, China.
| | - Jingwei Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China.
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China.
| |
Collapse
|
23
|
Huppert LA, Gumusay O, Idossa D, Rugo HS. Systemic therapy for hormone receptor-positive/human epidermal growth factor receptor 2-negative early stage and metastatic breast cancer. CA Cancer J Clin 2023; 73:480-515. [PMID: 36939293 DOI: 10.3322/caac.21777] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 01/09/2023] [Accepted: 02/01/2023] [Indexed: 03/21/2023] Open
Abstract
Hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative breast cancer is defined by the presence of the estrogen receptor and/or the progesterone receptor and the absence of HER2 gene amplification. HR-positive/HER2-negative breast cancer accounts for 65%-70% of all breast cancers, and incidence increases with increasing age. Treatment varies by stage, and endocrine therapy is the mainstay of treatment in both early stage and late-stage disease. Combinations with cyclin-dependent kinase 4/6 inhibitors have reduced distant recurrence in the early stage setting and improved overall survival in the metastatic setting. Chemotherapy is used based on stage and tumor biology in the early stage setting and after endocrine resistance for advanced disease. New therapies, including novel endocrine agents and antibody-drug conjugates, are now changing the treatment landscape. With the availability of new treatment options, it is important to define the optimal sequence of treatment to maximize clinical benefit while minimizing toxicity. In this review, the authors first discuss the pathologic and molecular features of HR-positive/HER2-negative breast cancer and mechanisms of endocrine resistance. Then, they discuss current and emerging therapies for both early stage and metastatic HR-positive/HER2-negative breast cancer, including treatment algorithms based on current data.
Collapse
Affiliation(s)
- Laura A Huppert
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Ozge Gumusay
- Department of Medical Oncology, Acibadem University, School of Medicine, Istanbul, Turkey
| | - Dame Idossa
- Masonic Comprehensive Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hope S Rugo
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| |
Collapse
|
24
|
Jaki T, Burdon A, Chen X, Mozgunov P, Zheng H, Baird R. Early phase clinical trials in oncology: Realising the potential of seamless designs. Eur J Cancer 2023; 189:112916. [PMID: 37301716 PMCID: PMC7614750 DOI: 10.1016/j.ejca.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND The pharmaceutical industry's productivity has been declining over the last two decades and high attrition rates and reduced regulatory approvals are being seen. The development of oncology drugs is particularly challenging with low rates of approval for novel treatments when compared with other therapeutic areas. Reliably establishing the potential of novel treatment and the corresponding optimal dosage is a key component to ensure efficient overall development. A growing interest lies in terminating developments of poor treatments quickly while enabling accelerated development for highly promising interventions. METHODS One approach to reliably establish the optimal dosage and the potential of a novel treatment and thereby improve efficiency in the drug development pathway is the use of novel statistical designs that make efficient use of the data collected. RESULTS In this paper, we discuss different (seamless) strategies for early oncology development and illustrate their strengths and weaknesses through real trial examples. We provide some directions for good practices in early oncology development, discuss frequently seen missed opportunities for improved efficiency and some future opportunities that have yet to fully develop their potential in early oncology treatment development. DISCUSSION Modern methods for dose-finding have the potential to shorten and improve dose-finding and only small changes to current approaches are required to realise this potential.
Collapse
Affiliation(s)
- Thomas Jaki
- MRC Biostatistics Unit, University of Cambridge, UK; University of Regensburg, Germany.
| | | | - Xijin Chen
- MRC Biostatistics Unit, University of Cambridge, UK
| | | | - Haiyan Zheng
- MRC Biostatistics Unit, University of Cambridge, UK
| | | |
Collapse
|
25
|
Mittal A, Molto Valiente C, Tamimi F, Schlam I, Sammons S, Tolaney SM, Tarantino P. Filling the Gap after CDK4/6 Inhibitors: Novel Endocrine and Biologic Treatment Options for Metastatic Hormone Receptor Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15072015. [PMID: 37046675 PMCID: PMC10093251 DOI: 10.3390/cancers15072015] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023] Open
Abstract
The rise of cyclin-dependent kinase (CDK)4/6 inhibitors has rapidly reshaped treatment algorithms for hormone receptor (HR)-positive metastatic breast cancer, with endocrine treatment (ET) plus a CDK4/6-inhibitor currently representing the standard of care in the first line setting. However, treatment selection for those patients experiencing progression while on ET + CDK4/6-inhibitors remains challenging due to the suboptimal activity or significant toxicities of the currently available options. There is also a paucity of data regarding the efficacy of older regimens, such as everolimus + exemestane, post-CDK4/6 inhibition. In this setting of high unmet need, several clinical trials of novel drugs have recently reported encouraging results: the addition of the AKT-inhibitor capivasertib to fulvestrant demonstrated a significant improvement in progression-free survival (PFS); the oral selective estrogen receptor degrader (SERD) elacestrant prolonged PFS compared to traditional ET in a phase 3 trial, particularly among patients with detectable ESR1 mutations; finally, PARP inhibitors are available treatment options for patients with pathogenic BRCA1/2 germline mutations. Overall, a plethora of novel endocrine and biologic treatment options are finally filling the gap between first-line ET and later line chemotherapy. In this review article, we recapitulate the activity of these novel treatment options and their potential role in future treatment algorithms.
Collapse
Affiliation(s)
- Abhenil Mittal
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center; Toronto, ON M5G 2C1, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Consolacion Molto Valiente
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center; Toronto, ON M5G 2C1, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Faris Tamimi
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center; Toronto, ON M5G 2C1, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Ilana Schlam
- Department of Hematology and Oncology, Tufts Medical Center, Boston, MA 02111, USA
| | - Sarah Sammons
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Sara M. Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Oncology and Onco-Hematology, University of Milan, 20122 Milan, Italy
- Correspondence: ; Tel.: +1-631-632-3800
| |
Collapse
|
26
|
Zagami P, Nicolò E, Corti C, Valenza C, Curigliano G. New Concepts in Cardio-Oncology. Cancer Treat Res 2023; 188:303-341. [PMID: 38175351 DOI: 10.1007/978-3-031-33602-7_12] [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] [Indexed: 01/05/2024]
Abstract
Cancer and cardiovascular disease are the two major causes of morbidity and mortality in worldwide. Discovering new therapeutic agents for the management of breast cancer (BC) has increased the numbers of cancer survivors but with the risk of cardiovascular adverse events (CV-AEs). All drugs can potentially damage the cardiovascular system, with different types of clinical manifestations from ischemic myocardial disease to vasculitis, thrombosis or pericarditis. An early detection of CV-AEs guarantees an earlier treatment, which is associated with better outcomes. Cardio-oncology field enlarged its studies to improve prevention, monitoring and treatment of all cardiotoxic manifestations related to old or modern oncological agents. A multidisciplinary approach with a close partnership between oncologists and cardiologists is essential for an optimal management and therapeutic decision-making. The aim of this chapter is to review all types of cardiotoxic manifestations related to novel and old agents approved for treatment of BC patients including chemotherapy, anti-HER2 agents, cyclin-dependent kinase 4/6 inhibitors, PolyADP-ribose polymerase (PARP) inhibitors, antiangiogenic drugs and immunotherapy. We also focused our discussion on prevention, monitoring, treatment, and management of CV-AEs.
Collapse
Affiliation(s)
- Paola Zagami
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy.
- Department of Oncology and Hematology, University of Milano, Milan, Italy.
| | - Eleonora Nicolò
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milano, Milan, Italy
| | - Chiara Corti
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milano, Milan, Italy
| | - Carmine Valenza
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milano, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milano, Milan, Italy
| |
Collapse
|
27
|
Varella L, Cristofanilli M. Evaluating Elacestrant in the Management of ER-Positive, HER2-Negative Advanced Breast Cancer: Evidence to Date. Onco Targets Ther 2023; 16:189-196. [PMID: 36993871 PMCID: PMC10041978 DOI: 10.2147/ott.s400563] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Breast cancer remains the second leading cause of cancer mortality in women. Endocrine therapy is the backbone treatment for hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer, the most common subtype. Although several endocrine therapy agents are available, essentially all HR-positive metastatic breast cancers will become resistant to these drugs. ESR1 mutations represent an important mechanism of resistance to aromatase inhibitors. Elacestrant is a novel oral selective estrogen receptor degrader (SERD) that selectively binds to the estrogen receptor in breast cancer cells, inhibiting tumor growth. Preclinical data suggested greater efficacy of elacestrant in combination with cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) or everolimus. In a Phase III clinical trial, elacestrant demonstrated a significant although modest improvement in median progression-free survival (PFS) compared to standard of care endocrine therapy in patients with HR-positive, HER2-negative advanced breast cancer. Importantly, there was also a significant benefit in patients with ESR1 mutations, which led to the FDA approval of elacestrant in this patient group. Elacestrant was generally well tolerated, with main side effects being upper gastrointestinal symptoms. There are several ongoing clinical trials evaluating the efficacy of elacestrant in the early setting as well as in combination with other targeted agents in the treatment of metastatic breast cancer. Other novel oral SERDs are also currently being evaluated in the treatment of HR-positive breast cancer. Results of ongoing clinical trials with these drugs will help clinicians decide the best sequence and combination of endocrine therapy agents.
Collapse
Affiliation(s)
- Leticia Varella
- Division of Medical Oncology, Internal Medicine Department, Weill Cornell Medicine, New York, NY, USA
- Correspondence: Leticia Varella, Tel +1 646 962 9888, Fax +1 646 962 0193, Email
| | - Massimo Cristofanilli
- Division of Medical Oncology, Internal Medicine Department, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
28
|
Patient-centered dosing: oncologists' perspectives about treatment-related side effects and individualized dosing for patients with metastatic breast cancer (MBC). Breast Cancer Res Treat 2022; 196:549-563. [PMID: 36198984 DOI: 10.1007/s10549-022-06755-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/18/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE Although metastatic breast cancer (MBC) is treatable, it is not curable and most patients remain on treatment indefinitely. While oncologists commonly prescribe the recommended starting dose (RSD) from the FDA-approved label, patient tolerance may differ from that seen in clinical trials. We report on a survey of medical oncologists' perspectives about treatment-related toxicity and willingness to discuss flexible dosing with patients. METHODS We disseminated a confidential survey via social media/email in Spring 2021. Eligible respondents needed to be US-based medical oncologists with experience treating patients with MBC. RESULTS Of 131 responses, 119 were eligible. Physicians estimated that 47% of their patients reported distressing treatment-related side effects; of these, 15% visited the Emergency Room/hospital and 37% missed treatment. 74% (n = 87) of doctors reported improvement of patient symptoms after dose reduction. 87% (n = 104) indicated that they had ever, if appropriate, initiated treatment at lower doses. Most (85%, n = 101) respondents did not believe that the RSD is always more effective than a lower dose and 97% (n = 115) were willing to discuss individualized dosing with patients. CONCLUSION Treatment-related side effects are prevalent among patients with MBC, resulting in missed treatments and acute care visits. To help patients tolerate treatment, oncologists may decrease initial and/or subsequent doses. The majority of oncologists reject the premise that a higher dose is always superior and are willing to discuss individualized dosing with patients. Given potential improvements regarding quality of life and clinical care, dose modifications should be part of routine shared decision-making between patients and oncologists.
Collapse
|
29
|
Elacestrant demonstrates strong anti-estrogenic activity in PDX models of estrogen-receptor positive endocrine-resistant and fulvestrant-resistant breast cancer. NPJ Breast Cancer 2022; 8:125. [PMID: 36446866 PMCID: PMC9709100 DOI: 10.1038/s41523-022-00483-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/07/2022] [Indexed: 12/02/2022] Open
Abstract
The selective oestrogen receptor (ER) degrader (SERD), fulvestrant, is limited in its use for the treatment of breast cancer (BC) by its poor oral bioavailability. Comparison of the orally bioavailable investigational SERD elacestrant, versus fulvestrant, demonstrates both drugs impact tumour growth of ER+ patient-derived xenograft models harbouring several ESR1 mutations but that elacestrant is active after acquired resistance to fulvestrant. In cell line models of endocrine sensitive and resistant breast cancer both drugs impact the ER-cistrome, ER-interactome and transcription of oestrogen-regulated genes similarly, confirming the anti-oestrogenic activity of elacestrant. The addition of elacestrant to CDK4/6 inhibitors enhances the antiproliferative effect compared to monotherapy. Furthermore, elacestrant inhibits the growth of palbociclib-resistant cells. Lastly, resistance to elacestrant involves Type-I and Type-II receptor tyrosine kinases which are amenable to therapeutic targeting. Our data support the wider clinical testing of elacestrant.
Collapse
|
30
|
Curtaz CJ, Kiesel L, Meybohm P, Wöckel A, Burek M. Anti-Hormonal Therapy in Breast Cancer and Its Effect on the Blood-Brain Barrier. Cancers (Basel) 2022; 14:cancers14205132. [PMID: 36291916 PMCID: PMC9599962 DOI: 10.3390/cancers14205132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
The molecular receptor status of breast cancer has implications for prognosis and long-term metastasis. Although metastatic luminal B-like, hormone-receptor-positive, HER2−negative, breast cancer causes brain metastases less frequently than other subtypes, though tumor metastases in the brain are increasingly being detected of this patient group. Despite the many years of tried and tested use of a wide variety of anti-hormonal therapeutic agents, there is insufficient data on their intracerebral effectiveness and their ability to cross the blood-brain barrier. In this review, we therefore summarize the current state of knowledge on anti-hormonal therapy and its intracerebral impact and effects on the blood-brain barrier in breast cancer.
Collapse
Affiliation(s)
- Carolin J. Curtaz
- Department of Gynecology and Obstetrics, University Hospital Würzburg, 97080 Würzburg, Germany
- Correspondence:
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, University Hospital of Münster, 48143 Münster, Germany
| | - Patrick Meybohm
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Malgorzata Burek
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
31
|
Downton T, Zhou F, Segara D, Jeselsohn R, Lim E. Oral Selective Estrogen Receptor Degraders (SERDs) in Breast Cancer: Advances, Challenges, and Current Status. Drug Des Devel Ther 2022; 16:2933-2948. [PMID: 36081610 PMCID: PMC9447452 DOI: 10.2147/dddt.s380925] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/20/2022] [Indexed: 11/23/2022] Open
Abstract
Several endocrine therapies are currently available for the treatment of estrogen receptor (ER) positive breast cancer, but the clinical benefit of these agents is limited by endocrine therapy drug resistance. A common mechanism of endocrine therapy resistance is ESR1 mutations. The first-generation selective estrogen receptor degrader (SERD) fulvestrant has activity against ESR1 mutant tumors but requires intramuscular injection and has poor bioavailability that precludes optimal drug dosing. This led to the development of second-generation SERDs which are potent and have improved oral bioavailability and pharmacokinetics. Several of these oral SERDs are now in phase III trials in both the early and advanced ER positive breast cancer settings. This review summarizes the background of oral SERD development, the current status and future perspectives.
Collapse
Affiliation(s)
- Teesha Downton
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Fiona Zhou
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Davendra Segara
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | | | - Elgene Lim
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Correspondence: Elgene Lim, Tel +61 2 9355 5600, Fax +61 2 9355 5602, Email
| |
Collapse
|
32
|
O’Brien SR, Edmonds CE, Katz D, Mankoff DA, Pantel AR. 18F-Fluoroestradiol (FES) PET/CT: review of current practice and future directions. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00494-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
33
|
Ha MJ, Raghavendra AS, Kettner NM, Qiao W, Damodaran S, Layman RM, Kelly KH, Shen Y, Tripathy D, Keyomarsi K. Palbociclib plus endocrine therapy significantly enhances overall survival of HR+/HER2- metastatic breast cancer patients compared to endocrine therapy alone in the second-line setting-a large institutional study. Int J Cancer 2022; 150:2025-2037. [PMID: 35133007 PMCID: PMC9018572 DOI: 10.1002/ijc.33959] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/27/2021] [Accepted: 01/25/2022] [Indexed: 12/24/2022]
Abstract
Cyclin-dependent-kinase-4/6 inhibitor (CDKi) plus endocrine therapy (ET) is standard of care for patients with advanced hormone receptor (HR)-positive, HER2-negative breast cancer (BC). The Breast Medical Oncology database at MD Anderson Cancer Center (MDACC) was analyzed to assess effectiveness of the CDKi palbociclib plus ET compared to ET alone. From a total of 5402 advanced HR+ HER2- BC patients referred to MDACC between 1997 and 2020, we identified eligible patients who received palbociclib in combination with first- (n=778) and second-line (n=410) ET. We further identified "control" patients who received ET alone in the first- (n=2452) and second-line (n=1183) settings. Propensity score matching analysis was conducted to balance baseline demographic and clinical characteristics between palbociclib and control cohorts to assess the effect of palbociclib treatment on progression-free survival (PFS) and overall survival (OS). For propensity-matched-cohort in the first-line setting (n=708), palbociclib group had significantly longer median PFS (17.4 vs. 11.1 months; p<0.0001) compared to controls. Median OS (44.3 vs. 40.2 months) did not show a statistically significant benefit in the first line setting. However, in the second-line setting, with 380 propensity-matched-cohort, the palbociclib group had significantly longer PFS (10 vs 5 months, p<0.0001) as well as OS (33 vs 24 months; p < 0.022), compared to controls. We conclude that in this single center analysis of a large cohort of metastatic HR+ HER2- BC patients, palbociclib in combination with ET was associated with improved PFS in both first- and second-line settings and OS in the second-line setting compared with ET alone cohort.
Collapse
Affiliation(s)
- Min Jin Ha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Nicole M Kettner
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Qiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Senthil Damodaran
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rachel M Layman
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - K Hunt Kelly
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yu Shen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Khandan Keyomarsi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
34
|
Xia S, Lin Q. Estrogen Receptor Bio-Activities Determine Clinical Endocrine Treatment Options in Estrogen Receptor-Positive Breast Cancer. Technol Cancer Res Treat 2022; 21:15330338221090351. [PMID: 35450488 PMCID: PMC9036337 DOI: 10.1177/15330338221090351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In estrogen receptor positive (ER+) breast cancer therapy, estrogen receptors (ERs) are the major targeting molecules. ER-targeted therapy has provided clinical benefits for approximately 70% of all breast cancer patients through targeting the ERα subtype. In recent years, mechanisms underlying breast cancer occurrence and progression have been extensively studied and largely clarified. The PI3K/AKT/mTOR pathway, microRNA regulation, and other ER downstream signaling pathways are found to be the effective therapeutic targets in ER+ BC therapy. A number of the ER+ (ER+) breast cancer biomarkers have been established for diagnosis and prognosis. The ESR1 gene mutations that lead to endocrine therapy resistance in ER+ breast cancer had been identified. Mutations in the ligand-binding domain of ERα which encoded by ESR1 gene occur in most cases. The targeted drugs combined with endocrine therapy have been developed to improve the therapeutic efficacy of ER+ breast cancer, particularly the endocrine therapy resistance ER+ breast cancer. The combination therapy has been demonstrated to be superior to monotherapy in overall clinical evaluation. In this review, we focus on recent progress in studies on ERs and related clinical applications for targeted therapy and provide a perspective view for therapy of ER+ breast cancer.
Collapse
Affiliation(s)
- Song Xia
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiong Lin
- School of Medicine, Jiangsu University, Zhenjiang, China
- Qiong Lin, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, China.
| |
Collapse
|
35
|
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: 60] [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
|
36
|
Ohoka N, Yokoo H, Okuhira K, Demizu Y, Naito M. Molecular Design, Synthesis, and Evaluation of SNIPER (ER) that Induces Targeted Protein Degradation of ERα. Methods Mol Biol 2022; 2418:363-382. [PMID: 35119675 DOI: 10.1007/978-1-0716-1920-9_20] [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] [Indexed: 06/14/2023]
Abstract
Manipulation of protein stability using small molecules has a great potential for both basic research and clinical therapy. Based on our protein knockdown technology, we developed chimeric degrader molecules SNIPER(ER)s that target the estrogen receptor alpha (ERα) for degradation via the ubiquitin-proteasome system. This chapter describes the design and synthesis of SNIPER(ER) compounds and methods for the evaluation of their activity in cellular systems and in a tumor xenograft model.
Collapse
Affiliation(s)
- Nobumichi Ohoka
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Hidetomo Yokoo
- Division of Organic Chemistry, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Keiichiro Okuhira
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
- Department of Environment and Health Sciences, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Yosuke Demizu
- Division of Organic Chemistry, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Mikihiko Naito
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan.
- Division of Organic Chemistry, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan.
- Laboratory of Targeted Protein Degradation, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
37
|
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: 5.3] [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
|
38
|
McAndrew NP, Finn RS. Clinical Review on the Management of Hormone Receptor-Positive Metastatic Breast Cancer. JCO Oncol Pract 2021; 18:319-327. [PMID: 34637323 DOI: 10.1200/op.21.00384] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The natural history of hormone receptor-positive breast cancer tends to be more favorable than other subtypes such as human epidermal growth factor receptor 2-amplified and triple-negative. In addition, the natural dependence on steroid hormone signaling has allowed for therapeutic targeting of this pathway and significant improvements in survival while maintaining quality of life: the two main goals in management of the disease. The sequential use of endocrine agents including the selective estrogen receptor modulators (tamoxifen), aromatase inhibitors (letrozole, anastrozole, and exemestane) and the selective estrogen receptor degrader fulvestrant has been the backbone of management for years. In the past decade, the introduction of molecularly targeted agents against intracellular targets such as mammalian target of rapamycin (everolimus), cyclin-dependent kinases 4 and 6 (palbociclib, ribociclib, and abemaciclib), and phosphatidylinositol 3-kinase (alpelisib) has offered patients effective nonchemotherapy-based options, which are improving outcomes. Although knowledge gaps still exist in regard to the optimal sequencing of these new regimens, they have expanded our repertoire of options for patients and have shifted the need for cytotoxic chemotherapy and its associated complications to later lines. Still, formatting a plan for these patients includes taking into account traditional prognostic factors such as menopausal status, previous treatments, disease-free interval for those patients with early breast cancer that has recurred, and tumor burden. To assist in developing this treatment plan, we will review the current data with systemic agents in the management of these patients.
Collapse
Affiliation(s)
- Nicholas P McAndrew
- Division of Hematology Oncology, Department of Medicine, Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Richard S Finn
- Division of Hematology Oncology, Department of Medicine, Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
39
|
Bai C, Wu S, Ren S, Zhu M, Luo G, Xiang H. Benzothiophene derivatives as selective estrogen receptor covalent antagonists: Design, synthesis and anti-ERα activities. Bioorg Med Chem 2021; 47:116395. [PMID: 34509864 DOI: 10.1016/j.bmc.2021.116395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/22/2021] [Accepted: 08/31/2021] [Indexed: 01/11/2023]
Abstract
Estrogen receptor α emerged as a well validated therapeutic target of breast cancer for decades. However, approximately 50% of patients who initially responding to standard-of-care (SoC), such as undergo therapy of Tamoxifen, generally inevitably progress to an endocrine-resistance ER+ phenotype. Recently, selective estrogen receptor covalent antagonists (SERCAs) targeted to ERα have been demonstrated as a therapeutic alternative. In the present study, series of novel 6-OH-benzothiophene (BT) derivatives targeting ERα and deriving from Raloxifene were designed, synthesized, and biologically evaluated as covalent antagonists. Driven by the antiproliferative efficacy in ER+ breast cancer cells, our chemical optimization finally led to compound 19d that with potent antagonistic activity in ER+ tumor cells while without agonistic activity in endometrial cells. Moreover, the docking simulation was carried out to elucidate the binding mode, revealing 19d as an antagonist and covalently binding to the cysteine residue at the 530 position of ER helix H11.
Collapse
Affiliation(s)
- Chengfeng Bai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shuangjie Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shengnan Ren
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Meiqi Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Guoshun Luo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hua Xiang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
40
|
Mouabbi JA, Osborne CK, Schiff R, Rimawi MF. Management of hormone receptor-positive, human epidermal growth factor 2-negative metastatic breast cancer. Breast Cancer Res Treat 2021; 190:189-201. [PMID: 34515904 DOI: 10.1007/s10549-021-06383-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/06/2021] [Indexed: 11/29/2022]
Abstract
Estrogen receptor (ER) is the major driver of most metastatic breast cancers (mBCs). Endocrine therapy (ET) is the most effective treatment for ER + mBC, but its effectiveness is limited by high rates of de novo and acquired resistance. A growing understanding of the biological characteristics and complexity of the ER pathway and the mechanisms of ET resistance has led to the development of a new generation of targeted therapies. One such mechanism is the cell cycle signaling pathways, which lead to the development of cyclin-dependent kinase 4/6 inhibitors (CDK4/6is) that have, in turn, transformed the management of such tumors. Another important mechanism is the alteration of the phosphatidylinositol 3'-kinase/AKT/mammalian target of rapamycin pathway. Drugs targeting each component of these pathways are currently used in clinical practice, and several more are in development. As a result, a myriad of new targeted therapies are consistently being added to the clinical oncologist armamentarium. Navigating the evolving and highly complex treatment landscape of HR + /HER2- mBC remains both an art and a challenge. In this review, we discuss the biological features of HR + /HER2- mBC and the different mechanisms of resistance to ET. We also discuss the management of mBC as the disease changes from endocrine-sensitive to endocrine-resistant.
Collapse
Affiliation(s)
- Jason A Mouabbi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - C Kent Osborne
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, 7200 Cambridge St., Suite 7A, Houston, TX, 77030, USA
| | - Rachel Schiff
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, 7200 Cambridge St., Suite 7A, Houston, TX, 77030, USA
| | - Mothaffar F Rimawi
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, 7200 Cambridge St., Suite 7A, Houston, TX, 77030, USA
| |
Collapse
|
41
|
Burguin A, Diorio C, Durocher F. Breast Cancer Treatments: Updates and New Challenges. J Pers Med 2021; 11:808. [PMID: 34442452 PMCID: PMC8399130 DOI: 10.3390/jpm11080808] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 12/31/2022] Open
Abstract
Breast cancer (BC) is the most frequent cancer diagnosed in women worldwide. This heterogeneous disease can be classified into four molecular subtypes (luminal A, luminal B, HER2 and triple-negative breast cancer (TNBC)) according to the expression of the estrogen receptor (ER) and the progesterone receptor (PR), and the overexpression of the human epidermal growth factor receptor 2 (HER2). Current BC treatments target these receptors (endocrine and anti-HER2 therapies) as a personalized treatment. Along with chemotherapy and radiotherapy, these therapies can have severe adverse effects and patients can develop resistance to these agents. Moreover, TNBC do not have standardized treatments. Hence, a deeper understanding of the development of new treatments that are more specific and effective in treating each BC subgroup is key. New approaches have recently emerged such as immunotherapy, conjugated antibodies, and targeting other metabolic pathways. This review summarizes current BC treatments and explores the new treatment strategies from a personalized therapy perspective and the resulting challenges.
Collapse
Affiliation(s)
- Anna Burguin
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1T 1C2, Canada;
- Cancer Research Center, CHU de Québec-Université Laval, Quebec City, QC G1V 4G2, Canada;
| | - Caroline Diorio
- Cancer Research Center, CHU de Québec-Université Laval, Quebec City, QC G1V 4G2, Canada;
- Department of Preventive and Social Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1T 1C2, Canada
| | - Francine Durocher
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1T 1C2, Canada;
- Cancer Research Center, CHU de Québec-Université Laval, Quebec City, QC G1V 4G2, Canada;
| |
Collapse
|
42
|
Efficacy of Fulvestrant in Women with Hormone-Resistant Metastatic Breast Cancer (mBC): A Canadian Province Experience. Cancers (Basel) 2021; 13:cancers13164163. [PMID: 34439317 PMCID: PMC8391338 DOI: 10.3390/cancers13164163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/04/2021] [Accepted: 08/16/2021] [Indexed: 01/08/2023] Open
Abstract
Simple Summary Fulvestrant is a medication that is approved as first and second-line treatment in patients with hormone receptor positive advanced breast cancer. In clinical practice, fulvestrant is still used beyond the second line of treatment. This study investigated the use of fulvestrant in a Saskatchewan population of women with advanced breast cancer. We found that fulvestrant is effective when used in both the early and later lines of treatment, although the benefit is more pronounced in the earlier line of therapy. Women with disease affecting their visceral organs such as lung, liver or peritoneum had decreased disease control and survival on fulvestrant. Women who had received chemotherapy after fulvestrant and had a clinical response to fulvestrant had better survival. Abstract Introduction: Fulvestrant has demonstrated efficacy in hormone receptor positive (HR+) metastatic breast cancer (mBC), both in first-and second-line settings. In clinical practice, however, fulvestrant has been used as a later-line therapy. This study assessed the efficacy of fulvestrant in women with mBC in early-versus later-line therapy. Methods: This retrospective cohort study assessed Saskatchewan women with HR+ mBC who received fulvestrant between 2003–2019. A multivariate Cox proportional survival analysis was performed. Results: One hundred and eighty-six women with a median age of 63.5 years were identified—178 (95.6%) had hormone-resistant mBC, 57.5% had visceral disease, and 43.0% had received chemotherapy before fulvestrant. 102 (54.8%) women received ≤2-line-therapy, and 84 (45.2%) received ≥3 line-therapy before fulvestrant. The median time to progression (TTP) was 12 months in the early-treatment vs. 6 months in the later-treatment group, p = 0.015. Overall survival (OS) from the start of fulvestrant was 26 months in the early-treatment group vs. 16 months in the later-treatment group, p = 0.067. On multivariate analysis, absence of visceral metastasis, HR: 0.70 (0.50–0.99), was significantly correlated with better TTP, whereas post-fulvestrant chemotherapy, HR: 0.32 (0.23–0.47), clinical benefit from fulvestrant, HR: 0.44 (0.30–0.65), and absence of visceral metastasis, HR: 0.70 (0.50–0.97), were correlated with better OS. Conclusions: Fulvestrant has demonstrated efficacy as both early-and later-line therapy in hormone-resistant mBC. Our results show that women with clinical benefit from fulvestrant, who received post-fulvestrant chemotherapy, or had non-visceral disease, had better survival.
Collapse
|
43
|
Dawood S, Konstantionva M, Dent R, Perazzo F, Kim SB, Villarreal-Garza C, Franco S, Dai MS, Simon S. Optimizing treatment selection, and sequencing decisions for Management of HR-Positive, HER2-Negative advanced breast cancer - Proceedings from breast cancer expert group meeting. BMC Proc 2021; 15:15. [PMID: 34372853 PMCID: PMC8351081 DOI: 10.1186/s12919-021-00224-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The therapeutic landscape of hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (mBC) has evolved considerably with the introduction of newer targeted agents and their combinations with endocrine therapies. In this scenario, optimizing treatment selection and sequencing is daunting for clinicians. The purpose of this review is to provide evidence-based answers to key clinical questions on treatment selection and sequencing for the management of HR + HER2 - mBC. DESIGN A panel of nine key opinion leaders from Argentina, Brazil, Colombia, Mexico, Moscow, Singapore, South Korea, Taiwan, and UAE convened in October 2018. They reviewed the literature and formulated answers to clinical questions on optimizing the management of HR + HER2 - mBC. RESULTS Evidence-based answers were formulated for: (1) optimal initial treatment choice; (2) ovarian function suppression, optimal endocrine partner, and role of cyclin-dependent kinase 4/6 (CDK4/6) inhibitors (in premenopausal women); (3) better first-line standard of care than aromatase inhibitors; (4) preferred second-line treatment; (5) treatment of oligometastatic disease; (6) factors influencing first-line single-agent endocrine therapy choice; (7) influence of endocrine resistance on treatment selection; (8) optimal maintenance regimen in visceral crisis; and (9) need for a breast cancer registry for patients with HR + HER2 - mBC. The panel also proposed a treatment-sequencing algorithm for the management of HR + HER2 - mBC. CONCLUSION The current article will serve as a comprehensive guide for optimizing the management of HR + HER2 - mBC. The proposed breast cancer registry will help identify unmet needs and develop strategic regional policies to help improve access to optimized care for HR + HER2 - mBC.
Collapse
Affiliation(s)
- Shaheenah Dawood
- Dubai Health Care City, Consultant Medical Oncologist, Mediclinic City Hospital - North Wing, Dubai, UAE.
| | - Maria Konstantionva
- Head of the Department of antitumor drug therapy, F. VladimirskIy Moscow Regional Research Clinical Institute (MONIKI), Moscow, Russia
| | - Rebecca Dent
- Head, Breast Medical Oncology Team, National Cancer Center Singapore, Singapore, Singapore
| | - Florencia Perazzo
- Department of Oncology, Centro de Educación Médicae Investigaciones Clínicas (CEMIC), Ciudad de Buenos Aires, Argentina
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, South Korea
| | - Cynthia Villarreal-Garza
- Centro de Cancer de Mama, Hospital Zambrano Hellion, Tecnologico de Monterrey, San Pedro Garza García, NL, Mexico
- Depto. de Investigacion, Instituto Nacional de Cancerologia, Mexico city, Mexico
| | - Sandra Franco
- Head of Oncology, Clínica del Country, Bogotá, Colombia
| | - Ming-Shen Dai
- Department of Hematology/Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan
| | - Sergio Simon
- Centro Paulista de Oncologia (CPO), Sao Paulo, Brazil
| |
Collapse
|
44
|
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: 20] [Impact Index Per Article: 5.0] [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
|
45
|
Fulvestrant-3-Boronic Acid (ZB716) Demonstrates Oral Bioavailability and Favorable Pharmacokinetic Profile in Preclinical ADME Studies. Pharmaceuticals (Basel) 2021; 14:ph14080719. [PMID: 34451816 PMCID: PMC8400955 DOI: 10.3390/ph14080719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 11/23/2022] Open
Abstract
Fulvestrant-3-boronic acid (ZB716), an oral selective estrogen receptor degrader (SERD) under clinical development, has been investigated in ADME studies to characterize its absorption, metabolism, and pharmacokinetics. ZB716 was found to have high plasma protein binding in human and animal plasma, and low intestinal mucosal permeability. ZB716 had high clearance in hepatocytes of all species tested. ZB716 was metabolized primarily by CYP2D6 and CYP3A. In human liver microsomes, ZB716 demonstrated relatively low inhibition of CYP1A2, 2C8, 2C9, 2C19, 2D6, and 3A4 (when testosterone was used as the substrate), and no inhibition of CYP2B6 and 3A4 (when midazolam was used as the substrate). In assays for enzyme activity, ZB716 induced CYP1A2, 2B6, and 3A4 in a concentration-dependent manner. Single-dose and repeated-dose pharmacokinetic studies in rats and dogs showed oral bioavailability, dose-proportional drug exposure, and drug accumulation as measured by maximum concentration and area under the concentration–time curve (AUC).
Collapse
|
46
|
Lu P, Santa-Maria CA, Ballinger TJ, Sheng JY. Landmark trials in the medical oncology management of metastatic breast cancer. Semin Oncol 2021; 48:246-258. [PMID: 34364700 PMCID: PMC8578298 DOI: 10.1053/j.seminoncol.2021.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022]
Abstract
Significant advances in the management of metastatic breast cancer (MBC) have guided more personalized treatment according to disease biology and led to improved survival outcomes and quality of life for patients. In this review, we discuss landmark clinical trials in medical oncology that have shaped the current standard of care for MBC. Combinations of endocrine therapy with cyclin-dependent kinase 4/6 inhibitors have led to substantial improvements in overall survival, thus becoming standard first-line treatment for patients with HR-positive MBC. Inhibition of the PI3K and mTOR pathway is another promising strategy to overcome resistance to endocrine therapy. HER2-targeted therapies have also evolved with the addition of pertuzumab to trastuzumab plus a taxane demonstrating remarkable overall survival advantage in patient with HER2-positive MBC. In second or later line therapies, novel anti-HER2 antibody-drug conjugates and TKIs have durable antitumor activity, survival benefit, and encouraging efficacy in the subgroup of patients with brain metastases. Triple negative breast cancer remains the most challenging subtype due to lack of druggable targets. Immunotherapy for patients with PDL-1 expression on tumor infiltrating immune cells and poly (ADP-ribose) polymerase inhibitors for those with germline BRCA1/2 mutations are the latest approved targeted strategies in this population. Numerous obstacles still exist in treating MBC, especially for patients whose disease develops resistance to available agents. Future research is eagerly awaited to address the optimal sequence or combination of therapies and to identify better biomarkers to guide precision medicine.
Collapse
Affiliation(s)
- Pei Lu
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | - Cesar A Santa-Maria
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore MD
| | - Tarah J Ballinger
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | - Jennifer Y Sheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore MD.
| |
Collapse
|
47
|
Hormone Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33983590 DOI: 10.1007/978-981-32-9620-6_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Hormone therapy is a major therapy for hormone receptor-positive breast cancer that improves survival. However, despite these hormone treatments, there are de novo or acquired resistance of breast cancer. Many studies revealed these resistance mechanisms, which are related to hormonal receptors including low expression or mutation of estrogen receptor alpha(ERα), co-factors and progesterone receptor, and with activation of growth signaling pathways such as PIK3A/Akt/mTOR pathway or cell cycle pathway. To overcome endocrine resistance based on these mechanisms, there have been many efforts in clinical studies of new agents which are representative of steroidal selective estrogen receptor down-regulator, cyclin-dependent kinase (CDK) 4/6 inhibitors, inhibitors of the PI3K/AKT/mTOR Pathway and histone deacetylase (HDAC) Inhibitors. Our studies at LBCB focused the endocrine resistance in young age and showed that age under 35 years is poor prognostic factor on not only single-center data but also Korean Breast Cancer Registry Data and that women with hormone receptor-positive breast cancer who were younger than 35 years of age had less response to anti-hormonal therapy. Also, a study for gene expression in hormone receptor-positive breast cancer at a very young age (<35) revealed that expression of cell cycle-related genes increased higher than that of premenopausal women in their forties. There have been a lot of studies and clinical trials to investigate the mechanisms of resistance to endocrine treatment and to overcome them with new drugs. However, many still do not know the precise mechanism of recurrence of breast cancer after endocrine treatment. In particular, the identification of the mechanism of endocrine resistance in young women, and the combination of drugs and clinical trials to overcome this require much effort.
Collapse
|
48
|
Andrahennadi S, Sami A, Manna M, Pauls M, Ahmed S. Current Landscape of Targeted Therapy in Hormone Receptor-Positive and HER2-Negative Breast Cancer. Curr Oncol 2021; 28:1803-1822. [PMID: 34064867 PMCID: PMC8161804 DOI: 10.3390/curroncol28030168] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/21/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Hormone receptor-positive and HER2-negative breast cancer (HR + BC) is the most prevalent breast cancer. Endocrine therapy is the mainstay of treatment, however, due to the heterogeneous nature of the disease, resistance to endocrine therapy is not uncommon. Over the past decades, the emergence of novel targeted therapy in combination with endocrine therapy has shown improvement in outcomes of HR + BC. This paper reviews available data of targeted therapy and the results of pivotal clinical trials in the management of HR + BC. Methods: A literature search in PubMed and Google Scholar was performed using keywords related to HR + BC and targeted therapy. Major relevant studies that were presented in international cancer research conferences were also included. Results: Endocrine therapy with tamoxifen and aromatase inhibitors are backbone treatments for women with early-stage HR + BC leading to a significant reduction in mortality. They can also be used for primary prevention in women with a high risk of breast cancer. Preliminary data has shown the efficacy of adjuvant cyclin-dependent kinase (CDK) 4/6 inhibitor, abemaciclib, in high-risk disease in combination with aromatase inhibitors. For most women with advanced HR + BC, endocrine therapy is the primary treatment. Recent evidence has shown that the use of CKD 4/6 inhibitors, mTOR inhibitors, and PI3K inhibitors in combination with endocrine therapy has been associated with better outcomes and delays initiation of chemotherapy. Several novel agents are under study for HR + BC. Discussion: Targeted treatment options for HR + BC have evolved. The future of overcoming resistance to targeted therapy, novel compounds, and predictive markers are key to improving HR + BC outcomes.
Collapse
Affiliation(s)
- Samitha Andrahennadi
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.A.); (A.S.); (M.M.); (M.P.)
| | - Amer Sami
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.A.); (A.S.); (M.M.); (M.P.)
- Saskatoon Cancer Center, Saskatchewan Cancer Agency, University of Saskatchewan, 20 Campus Drive, Saskatoon, SK S7N 4H4, Canada
| | - Mita Manna
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.A.); (A.S.); (M.M.); (M.P.)
- Saskatoon Cancer Center, Saskatchewan Cancer Agency, University of Saskatchewan, 20 Campus Drive, Saskatoon, SK S7N 4H4, Canada
| | - Mehrnoosh Pauls
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.A.); (A.S.); (M.M.); (M.P.)
- Saskatoon Cancer Center, Saskatchewan Cancer Agency, University of Saskatchewan, 20 Campus Drive, Saskatoon, SK S7N 4H4, Canada
| | - Shahid Ahmed
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (S.A.); (A.S.); (M.M.); (M.P.)
- Saskatoon Cancer Center, Saskatchewan Cancer Agency, University of Saskatchewan, 20 Campus Drive, Saskatoon, SK S7N 4H4, Canada
| |
Collapse
|
49
|
Zhou CM, Xue Q, Wang Y, Tong J, Ji M, Yang JJ. Machine learning to predict the cancer-specific mortality of patients with primary non-metastatic invasive breast cancer. Surg Today 2021; 51:756-763. [DOI: 10.1007/s00595-020-02170-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/03/2020] [Indexed: 03/28/2023]
|
50
|
Cardiovascular toxicity of breast cancer treatment: an update. Cancer Chemother Pharmacol 2021; 88:15-24. [PMID: 33864486 DOI: 10.1007/s00280-021-04254-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/28/2021] [Indexed: 10/21/2022]
Abstract
Novel chemotherapeutic agents have marked a new era in oncology during the past decade, prolonging significantly the overall survival of breast cancer patients. Nevertheless, contemporary antineoplastic treatments can frequently cause adverse cardiovascular side effects. Common manifestations of chemotherapy-induced cardiotoxicity include cardiomyopathy, ischemia, conduction disturbances, hypertension and thromboembolic events, while the type of the treatment regimen administered crucially determines clinical outcome. The aim of this literature review is to analyze the incidence and the underlying mechanisms of cardiovascular toxicity caused by agents approved for breast cancer, as well as to describe ways of monitoring and treating the cardiotoxic effects in breast cancer patients. Moreover, our work intends to provide an easy-to-grasp synopsis of recent and clinically meaningful advances in the field.
Collapse
|