1
|
Tripathi A, Chhabra A, Rizvi S, Tyagi RK. Selective steroid receptor modulators, degraders and PROTACs: Therapeutic strategies in management of endocrine-related cancers. Mol Cell Endocrinol 2025; 605:112569. [PMID: 40339978 DOI: 10.1016/j.mce.2025.112569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/23/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
Endocrine-related disorders are highly prevalent globally, affecting millions of people. Such diseases are multifactorial in origin and are influenced by the complex interplay of genetics, lifestyle, and environmental factors. Recurring disruptions in the endocrine homeostasis can lead to a cascade of endocrine-related cancers. It is well known that nuclear receptors (NRs), particularly estrogen receptor and androgen receptor malfunctioning promote the oncogenesis of breast cancer and prostate cancer, respectively. However, existing therapeutics against these diseases, including aromatase inhibitors, (anti-) hormonal therapy, etc., often yield limited success, prompting to explore alternative methods of disease management. Additionally, drug resistance is prominent in cancer patients undergoing multidrug therapy. Currently, novel drug design strategies targeting NRs are being implemented for the discovery of a new generation of small molecule modulators, including selective NR modulators (SNuRMs) and degraders (SNuRDs). Moreover, proteolysis-targeting chimeras (PROTACs) as NR degraders, are also being developed primarily to overcome drug resistance, enhance protein selectivity, and mitigate off-target toxicity. This review highlights recent advancements in SNuRMs and SNuRDs for managing NRs-associated endocrine/metabolic disorders. Furthermore, we discuss the therapeutic potential of PROTAC degraders as a stand-alone strategy for receptor-mediated disease intervention, offering new avenues for precision medicine.
Collapse
Affiliation(s)
- Anjali Tripathi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India
| | - Ayushi Chhabra
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India
| | - Sheeba Rizvi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India
| | - Rakesh K Tyagi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067, India.
| |
Collapse
|
2
|
Tracy PD, Bopp E, Milner E, Garrido-Castro AC, Giordano A, Mayer EL, Tolaney SM, Tarantino P, Schlam I. Management of Metastatic Hormone Receptor-Positive Breast Cancer Beyond CDK4/6 Inhibitors. Curr Oncol Rep 2025:10.1007/s11912-025-01689-9. [PMID: 40434676 DOI: 10.1007/s11912-025-01689-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2025] [Indexed: 05/29/2025]
Abstract
PURPOSE OF REVIEW Since the introduction of cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) in combination with endocrine therapy (ET) as the first-line treatment for metastatic hormone receptor (HR) positive, human epidermal growth factor receptor 2 (HER2) negative (HR+/HER2-) breast cancer, there has been a significant expansion in the number of therapeutic options for subsequent lines of therapy. Many new agents are being studied, with potential for future regulatory approval. The increased number of therapeutic options raises questions about the optimal selection and sequencing of therapies for individual patients. These advances represent an important clinical challenge in this rapidly evolving field, given the introduction of new therapies targeting various pathways (alone or in combination) and new therapeutic classes being studied. RECENT FINDINGS Recently approved targeted therapies have demonstrated improvements in progression free survival (PFS) for patients whose cancer harbors mutations in the PI3K/AKT pathway, ESR1, BRCA1/2, and/or PALB2. Data to support continuation of CDK4/6 inhibition after progression on a prior CDK4/6i remains mixed, though some studies suggest a subset of patients may benefit from this approach. Several agents with unique mechanisms of action have shown promise in data from early phase trials, and have the potential to enter the treatment lexicon in the coming years. Examples include CDK2- and CDK4-selective inhibitors, complete estrogen receptor antagonists (CERANs), proteolysis targeting chimeras (PROTACs), and next-generation PI3K pathway inhibitors. In this narrative review, we summarize the current and upcoming treatments for metastatic HR+/HER2- breast cancer after progression on a CDK4/6i plus ET, with a focus on the following: an overview of first-line regimens of CDK4/6i plus ET and observed mechanisms of resistance; currently approved second-line therapy options; and upcoming options currently under exploration in clinical trials. We focus primarily on new therapy classes that may offer therapeutic options beyond currently available treatments.
Collapse
Affiliation(s)
- Philip D Tracy
- Department of Hematology/Oncology, Tufts Medical Center, Boston, MA, 02111, USA
- Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Emily Bopp
- Department of Medicine, Tufts Medical Center, Boston, MA, 02111, USA
| | - Emily Milner
- Department of Medicine, Tufts Medical Center, Boston, MA, 02111, USA
| | - Ana C Garrido-Castro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Antonio Giordano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Erica L Mayer
- 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, Milan, 20122, Italy.
| | - Ilana Schlam
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
3
|
Hamilton EP, Jeselsohn RM, Vahdat LT, Hurvitz SA. PROteolysis TArgeting Chimera (PROTAC) Estrogen Receptor Degraders for Treatment of Estrogen Receptor-Positive Advanced Breast Cancer. Target Oncol 2025; 20:431-444. [PMID: 40327300 DOI: 10.1007/s11523-025-01137-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 05/07/2025]
Abstract
The estrogen receptor (ER) signaling pathway is a key driver of breast cancer, primarily through the activation of genes that promote tumor cell survival and growth. The recommended first-line treatment for ER-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) advanced or metastatic breast cancer is endocrine therapy plus a cyclin-dependent kinase 4/6 (CDK4/6) inhibitor. However, most patients experience disease progression, and there is no clear standard of care in the second-line setting. Thus, novel treatments in the advanced setting are needed. In this narrative review, we describe the unique mechanisms of action of a new class of drugs called PROteolysis TArgeting Chimera (PROTAC) ER degraders. Unlike other ER-targeted therapies, these small molecules harness the body's primary intracellular natural protein disposal machinery, the ubiquitin-proteasome system, to directly induce ER degradation. Vepdegestrant (ARV-471) is the furthest advanced PROTAC ER degrader currently in clinical development. Preclinical data demonstrate increased tumor growth inhibition with vepdegestrant alone or in combination with CDK4/6 inhibitors compared with the selective ER degrader fulvestrant. In a first-in-human phase 1/2 clinical study, vepdegestrant administered orally as monotherapy or in combination with palbociclib showed promising clinical activity and a favorable safety profile in patients with heavily pretreated ER+/HER2- advanced breast cancer. Several other PROTAC ER degraders (AC699, ERD-3111, ERD-4001, and HP568) are in early development and have demonstrated activity in preclinical breast cancer models, with some recently entering clinical trials. The data highlight the potential for PROTAC ER degraders to be a new backbone therapy in breast cancer.
Collapse
Affiliation(s)
- Erika P Hamilton
- Breast Cancer Research Program, Sarah Cannon Research Institute, 335 24th Avenue North, Suite 300, Nashville, TN, 37203, USA.
| | | | - Linda T Vahdat
- Hematology/Oncology, Dartmouth Cancer Center, Lebanon, NH, USA
- Department of Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Sara A Hurvitz
- Division of Hematology Oncology, Department of Medicine, UW Medicine, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
4
|
Vidal M, Falato C, Pascual T, Sanchez-Bayona R, Muñoz-Mateu M, Cebrecos I, Gonzalez-Farré X, Cortadellas T, Margelí Vila M, Luna MA, Siso C, Amillano K, Galván P, Bergamino MA, Ferrero-Cafiero JM, Salvador F, Espinosa Guerrero A, Pare L, Sanfeliu E, Prat A, Bellet M. Elacestrant in Women with Estrogen Receptor-Positive and HER2-Negative Early Breast Cancer: Results from the Preoperative Window-of-Opportunity ELIPSE Trial. Clin Cancer Res 2025; 31:1223-1232. [PMID: 39820652 PMCID: PMC11959270 DOI: 10.1158/1078-0432.ccr-24-2460] [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: 07/30/2024] [Revised: 10/04/2024] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE Elacestrant has shown significantly prolonged progression-free survival compared with standard-of-care endocrine therapy in estrogen receptor-positive (ER-positive), HER2-negative metastatic breast cancer, whereas potential benefit in early-stage disease requires further exploration. The SOLTI-ELIPSE window-of-opportunity trial investigated the biological changes induced by a short course of preoperative elacestrant in postmenopausal women with early breast cancer. PATIENTS AND METHODS Eligible patients with untreated T1c (≥1.5 cm)-T3, N0, ER-positive/HER2-negative breast cancer with locally assessed Ki67 ≥10% received elacestrant at a daily dose of 345 mg for 4 weeks. The primary efficacy endpoint was complete cell cycle arrest, defined as Ki67 ≤2.7%, on day 28. RESULTS Overall, 22 patients were evaluable for the primary endpoint. Elacestrant was associated with a complete cell cycle arrest rate of 27.3% and a statistically significant Ki67 geometric mean change of -52.9% (P = 0.007; 95% confidence interval, -67.4 to -32.1). Notably, the treatment with elacestrant led to a shift toward a more endocrine-sensitive and less proliferative tumor phenotype based on PAM50-based gene signatures. Elacestrant increased the expression of immune-response genes (GZMB, CD4, and CD8A) and suppressed proliferation and estrogen-signaling genes (MKI67, ESR1, and AR). These biological changes were independent of the levels of Ki67 suppression on day 28. The most common adverse events were grade 1 anemia (21.7%), hot flushes (8.7%), constipation (8.7%), and abdominal pain (8.7%). One patient experienced a grade 3 cutaneous rash, leading to treatment discontinuation. No other serious adverse events were reported. CONCLUSIONS Preoperative treatment with elacestrant in early breast cancer demonstrated relevant biological and molecular responses and exhibited a manageable safety profile. These findings support further investigation of elacestrant in the early setting.
Collapse
Affiliation(s)
- Maria Vidal
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Claudette Falato
- SOLTI Cancer Research Group, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Tomás Pascual
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Rodrigo Sanchez-Bayona
- SOLTI Cancer Research Group, Barcelona, Spain
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Montserrat Muñoz-Mateu
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Isaac Cebrecos
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Tomás Cortadellas
- Breast Unit, Department of Obstetrics and Gynaecology, Hospital Universitari General de Catalunya, Barcelona, Spain
| | - Mireia Margelí Vila
- SOLTI Cancer Research Group, Barcelona, Spain
- B-ARGO Group, Medical Oncology Department, ICO Badalona, Germans Trias I Pujol Institute, Badalona, Spain
- Medicine Department, Autonomous University, Barcelona, Spain
| | - Miguel A. Luna
- B-ARGO Group, Medical Oncology Department, ICO Badalona, Germans Trias I Pujol Institute, Badalona, Spain
| | | | - Kepa Amillano
- Hospital Universitari Sant Joan de Reus, Barcelona, Spain
| | - Patricia Galván
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Milana A. Bergamino
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- B-ARGO Group, Medical Oncology Department, ICO Badalona, Germans Trias I Pujol Institute, Badalona, Spain
| | | | | | | | - Laia Pare
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Esther Sanfeliu
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Pathology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Aleix Prat
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Meritxell Bellet
- SOLTI Cancer Research Group, Barcelona, Spain
- Medicine Department, Autonomous University, Barcelona, Spain
- Vall d’Hebron University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
5
|
Fan Z, Xu Y, Guo S, Song B. Post-marketing safety of elacestrant in breast cancer: a pharmacovigilance investigation using the FDA adverse event reporting system. BMC Pharmacol Toxicol 2025; 26:56. [PMID: 40069893 PMCID: PMC11895172 DOI: 10.1186/s40360-025-00887-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Recently, the US Food and Drug Administration approved a new oral selective estrogen receptor downregulator for breast cancer, namely, elacestrant (Orserdu). This study aimed to analyze the signals of adverse events (AEs) within the introduction of elacestrant to the market using the FDA Adverse Event Reporting System (FAERS) database. METHODS Reports on the AEs of elacestrant after its marketing were obtained from the FAERS database. Disproportionality was analyzed using the reporting odds ratio to calculate the magnitude of the risk of the target drug and the AE combination, and the proportional reporting ratio to quantify the strength of the association between the drug and the AEs. RESULTS A total of 3132 reports on elacestrant-related AEs were obtained, with disease progression, drug ineffectiveness, product dose omission, arthralgia, asthenia, increased tumor marker levels, and bone pain (Number of reported cases (a) ≥ 3 and lower limit of 95% confidence interval >1) being the high-frequency events not mentioned on the drug label. The top three total frequencies at the system organ class level comprised general disorders and administration site conditions, gastrointestinal disorders, and musculoskeletal and connective tissue disorders. CONCLUSIONS FAERS data analyses were conducted to evaluate the safety of post-marketing clinical use of elacestrant and to ensure that physicians identify the risk factors for the AEs of this drug.
Collapse
Affiliation(s)
- Ziyi Fan
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030032, China
| | - Yanan Xu
- Department of Laboratory, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Shuding Guo
- School of Graduate Studies, Lingnan University, Hong Kong, China
| | - Bin Song
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030032, China.
| |
Collapse
|
6
|
Mulligan M, Boudreau MW, Bouwens BA, Lee Y, Carrell HW, Zhu J, Mousses S, Shapiro DJ, Nelson ER, Fan TM, Hergenrother PJ. Single Dose of a Small Molecule Leads to Complete Regressions of Large Breast Tumors in Mice. ACS CENTRAL SCIENCE 2025; 11:228-238. [PMID: 40028352 PMCID: PMC11869136 DOI: 10.1021/acscentsci.4c01628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/25/2024] [Accepted: 12/19/2024] [Indexed: 03/05/2025]
Abstract
Patients with estrogen receptor α positive (ERα+) breast cancer typically undergo surgical resection, followed by 5-10 years of treatment with adjuvant endocrine therapy. This prolonged intervention is associated with a host of undesired side effects that reduce patient compliance, and ultimately therapeutic resistance and disease relapse/progression are common. An ideal anticancer therapy would be effective against recurrent and refractory disease with minimal dosing; however, there is little precedent for marked tumor regression with a single dose of a small molecule therapeutic. Herein we report ErSO-TFPy as a small molecule that induces quantitative or near-quantitative regression of tumors in multiple mouse models of breast cancer with a single dose. Importantly, this effect is robust and independent of tumor size with eradication of even very large tumors (500-1500 mm3) observed. Mechanistically, these tumor regressions are a consequence of rapid induction of necrotic cell death in the tumor and are immune cell independent. If successfully translated to human cancer patients, the benefits of such an anticancer drug that is effective with a single dose would be significant.
Collapse
Affiliation(s)
- Michael
P. Mulligan
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Matthew W. Boudreau
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Brooke A. Bouwens
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Department
of Biochemistry, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yoongyeong Lee
- Department
of Comparative Biosciences, University of
Illinois at Urbana-Champaign, Urbana, Illinois 61802, United States
| | - Hunter W. Carrell
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Junyao Zhu
- Department
of Biochemistry, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Spyro Mousses
- Systems
Oncology, Scottsdale, Arizona 85255, United States
| | - David J. Shapiro
- Department
of Biochemistry, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Erik R. Nelson
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department
of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Division
of Nutritional Sciences, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute
for Advanced Science and Technology, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department
of Veterinary Clinical Medicine, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61802, United States
| | - Paul J. Hergenrother
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
7
|
Sarfraz A, Sarfraz M, Javad F, Khalid M, Shah B, Gul A, Ganiyani MA, Ismail A, Cheema K. Elacestrant in hormone receptor-positive metastatic breast cancer: a post-hoc analysis. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002293. [PMID: 39991467 PMCID: PMC11847623 DOI: 10.37349/etat.2025.1002293] [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: 09/16/2024] [Accepted: 11/29/2024] [Indexed: 02/25/2025] Open
Abstract
Background Breast cancer is a leading cause of mortality in women. Hormone therapy plays a crucial role in treatment of hormone receptor-positive metastatic breast cancer. Elacestrant is a selective estrogen receptor degrader (SERD) that has shown promise in early-phase clinical trials. This post-hoc analysis systematically evaluates elacestrant's effectiveness in hormone receptor-positive metastatic breast cancer patients, providing insights into its efficacy, safety, and potential advantages over existing treatments. Methods We adhered to the PRISMA Statement 2020 guidelines and systematically searched the databases PubMed/MEDLINE, ClinicalTrials.gov, Web of Science, and Embase. We conducted the post-hoc analysis using R software (V 4.3.3), applying the inverse variance method and the DerSimonian-Laird estimator to pool effect estimates with a random-effects model. We assessed heterogeneity using the Cochran's Q test and the I 2 statistic. Results Our post-hoc analysis encompassed 3 clinical trials and a total of 835 participants. The mean age of all 835 participants across the three trials was 59.5 years (95% CI: 58.7-60.3). The pooled progression-free survival (PFS)-was estimated at 4.38 (95% CI: -7.58-16.35, P = 0.47), and the pooled objective response rate (ORR) was 7% (95% CI: 2-18%, P = 0.04), with significant heterogeneity observed among the studies. Discussion Elacestrant shows promise for improving outcomes in hormone receptor-positive metastatic breast cancer, but further research is needed to confirm its effectiveness. Future studies should include larger sample sizes, comprehensive phase II and III trials, and investigation of elacestrant in combination with other drugs or in preoperative settings.
Collapse
Affiliation(s)
- Azza Sarfraz
- Department of Pediatrics, The Aga Khan University, Karachi 74800, Pakistan
| | - Muzna Sarfraz
- Department of Research, King Edward Medical University, Lahore 54000, Pakistan
| | - Faheem Javad
- Department of Research, Al Nafees Medical College, Islamabad 45600, Pakistan
| | - Musfira Khalid
- Department of Research, Fatima Jinnah Medical University, Lahore 54000, Pakistan
| | - Bushra Shah
- Department of Research, Fatima Jinnah Medical University, Lahore 54000, Pakistan
| | - Amna Gul
- Department of Research, Liaquat National Medical College and Hospital, Karachi 74800, Pakistan
| | - Mohammad Arfat Ganiyani
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
| | - Areeba Ismail
- Department of Research, Jinnah Sindh Medical University, Karachi 75510, Pakistan
| | - Khadija Cheema
- Department of Medicine, Tucson Medical Center, Tucson, AZ 85712, USA
| |
Collapse
|
8
|
Das GM, Oturkar CC, Menon V. Interaction between Estrogen Receptors and p53: A Broader Role for Tamoxifen? Endocrinology 2025; 166:bqaf020. [PMID: 39891710 PMCID: PMC11837209 DOI: 10.1210/endocr/bqaf020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/20/2025] [Accepted: 01/30/2025] [Indexed: 02/03/2025]
Abstract
Tamoxifen is one of the most widely used anticancer drugs in the world. It is a safe drug with generally well-tolerated side effects and has been prescribed for the treatment of early-stage and advanced-stage or metastatic estrogen receptor α (ERα/ESR1)-positive breast cancer. Tamoxifen therapy also provides a 38% reduction of the risk of developing breast cancer in women at high risk. With the advent of newer medications targeting ERα-positive breast cancer, tamoxifen is now mainly used as adjuvant therapy for lower-risk premenopausal breast cancer and cancer prevention. It is widely accepted that tamoxifen as a selective estrogen receptor modulator exerts its therapeutic effect by competitively binding to ERα, leading to the recruitment of corepressors and inhibition of transcription of genes involved in the proliferation of breast cancer epithelium. As such, expression of ERα in breast tumors has been considered necessary for tumors to be responsive to tamoxifen therapy. However, ERα-independent effects of tamoxifen in various in vitro and in vivo contexts have been reported over the years. Importantly, the recent discovery that ERα and estrogen receptor β (ERβ/ESR2) can bind tumor suppressor protein p53 with functional consequences has provided new insights into the mechanisms underlying response to tamoxifen therapy and resistance. Furthermore, these findings have paved the way for broadening the use of tamoxifen by potentially repurposing it to treat triple negative (negative for ERα, human epidermal growth factor receptor 2, and progesterone receptor) breast cancer. Herein, we summarize these developments and discuss their mechanistic underpinnings and clinical implications.
Collapse
Affiliation(s)
- Gokul M Das
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Chetan C Oturkar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Vishnu Menon
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
9
|
Scafetta R, Zagami P, Del Re M, Criscitiello C, Marra A, Curigliano G. Oral selective estrogen receptor degraders for breast cancer treatment: focus on pharmacological differences. Breast Cancer Res Treat 2025; 209:455-465. [PMID: 39776334 DOI: 10.1007/s10549-024-07595-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE The management of hormone receptor-positive (HR +) breast cancer (BC) relies on endocrine therapy (ET), with a primary focus on disrupting estrogen receptor (ER) signaling due to its critical role in BC tumorigenesis and progression. While effective for both early-stage and advanced breast cancers, ET frequently encounters resistance mechanisms, including both ligand-dependent and ligand-independent trajectories, ultimately leading to disease progression. METHODS We searched PubMed, EMBASE and Scopus databases to review the current evidence on the use of novel oral selective estrogen receptor degraders (SERDs) for the treatment of HR+ BC. CONCLUSIONS Somatic activating mutations of the estrogen receptor 1 (ESR1) gene are known to sustain ER activity, boost ER-dependent gene transcription, and foster resistance to ET. The most significant gap remains after treatment failure with ET and cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors, where fulvestrant monotherapy typically results in a median progression-free survival of 2-3 months. Novel compounds, including oral SERDs, have been explored for their potential to overcome therapeutic resistance, both as monotherapy and in combination with other targeted therapies. Herein, we provide an overview on the latest findings on oral SERDs, examining their mechanism of action, safety data, and pharmacokinetics and pharmacodynamics profiles.
Collapse
Affiliation(s)
- Roberta Scafetta
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Medical Oncology Department, Campus Bio-Medico University of Rome, Rome, Italy
| | - Paola Zagami
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Marzia Del Re
- Saint Camillus International, University of Health and Medical Sciences, Rome, Italy
- Scientific Direction, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Antonio Marra
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy.
| |
Collapse
|
10
|
Howell SJ, Howell A. Targeting Oestrogen Receptor Signalling in Breast Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:449-474. [PMID: 39821038 DOI: 10.1007/978-3-031-70875-6_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
There has been over 130 years of research into the treatment of breast cancer using approaches that target oestrogen receptor signalling. Here, we summarise the development of the key pillars of such endocrine therapy, namely, oestrogen deprivation, achieved through ovarian suppression and/or aromatase inhibition, and oestrogen receptor blockade, through selective oestrogen receptor modulators, downregulators and novel compounds entering early phase development. The translation of these compounds from advanced to early breast cancer settings is discussed with a focus on the placebo-controlled breast cancer prevention studies to most accurately describe the side effect profiles of the main approaches.
Collapse
Affiliation(s)
- Sacha J Howell
- Division of Cancer Sciences, University of Manchester, Manchester, UK.
- Manchester University NHS Foundation Trust, Manchester, UK.
- The Christie NHS Foundation Trust, Manchester, UK.
| | - Anthony Howell
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Manchester University NHS Foundation Trust, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
11
|
Rej RK, Hu B, Chen Z, Acharyya RK, Wu D, Metwally H, McEachern D, Wang Y, Jiang W, Bai L, Nishimura LS, Gersch CL, Wang M, Wen B, Sun D, Carlson K, Katzenellenbogen JA, Xu G, Zhang W, Wu W, Priestley ES, Sui Z, Rae JM, Wang S. Discovery of ERD-12310A as an Exceptionally Potent and Orally Efficacious PROTAC Degrader of Estrogen Receptor α (ERα). J Med Chem 2024; 67:20933-20965. [PMID: 39585895 DOI: 10.1021/acs.jmedchem.4c01401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Inhibition of estrogen receptor alpha (ERα) signaling is an established therapeutic approach for the treatment of ER-positive (ER+) breast cancers, but new therapeutic strategies are urgently needed to overcome clinical resistance. In the present study, we describe the discovery and extensive evaluation of ERD-12310A as an exceptionally potent and orally efficacious PROTAC degrader of ERα. ERD-12310A achieved a DC50 value of 47 pM and is 10 times more potent than ARV-471. ERD-12310A displayed an improved pharmacokinetic profile in mice and rats over ARV-471. ERD-12310A attained tumor regression in the ER+, estrogen-dependent MCF-7 breast cancer xenograft model with wild-type ER and is more potent than ARV-471. Importantly, ERD-12310A achieved strong tumor growth inhibition in MCF-7 xenograft tumors harboring the clinically relevant ESR1Y537S mutation, which confers resistance to traditional antiestrogens. Our data position ERD-12310A as a promising candidate for further development as a potential therapy for ER+ breast cancer.
Collapse
Affiliation(s)
- Rohan Kalyan Rej
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Biao Hu
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Zhixiang Chen
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Ranjan Kumar Acharyya
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Dimin Wu
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Hoda Metwally
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Donna McEachern
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Yu Wang
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Wei Jiang
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Longchuan Bai
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Leticia S Nishimura
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Christina L Gersch
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
| | - Meilin Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor 48109, United States
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor 48109, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor 48109, United States
| | - Kathryn Carlson
- Department of Chemistry, University of Illinois, 600 South Mathews Avenue, Urbana 61801, United States
| | - John A Katzenellenbogen
- Department of Chemistry, University of Illinois, 600 South Mathews Avenue, Urbana 61801, United States
| | - Guozhang Xu
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia 09406, United States
| | - Weihong Zhang
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia 09406, United States
| | - Wenxue Wu
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia 09406, United States
| | - E Scott Priestley
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia 09406, United States
| | - Zhihua Sui
- SK Life Science Laboratories, 2500 Renaissance Boulevard, King of Prussia 09406, United States
| | - James M Rae
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor 48109, United States
| | - Shaomeng Wang
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor 48109, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor 48109, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor 48109, United States
| |
Collapse
|
12
|
Zhang D, Lu Z, He Y, Leng XY, Meng X, Lei X, Kong D, Sun L, Hu W, Yang Y. Discovery of Thiochroman Derivatives as Potent, Oral Selective Estrogen Receptor Degraders and Antagonists for the Treatment of Endocrine-Resistant Breast Cancer. J Med Chem 2024; 67:21545-21567. [PMID: 39610216 DOI: 10.1021/acs.jmedchem.4c02453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Selective estrogen receptor degraders (SERDs) deplete the ER signaling pathway via antagonism and degradation of ERα and represent a promising strategy to tackle endocrine resistance. Here, we report a new class of SERDs by pharmacological evolution of a selective estrogen receptor modulator, lasofoxifene. The structure-activity relationship study and efforts to circumvent the issue of human ether-a-go-go-related gene led to the identification of compounds 51. This bifunctional compound displayed broad activity across a vast array of cell backgrounds and was capable of effectively degrading and antagonizing wild-type ERα and clinically relevant ERα mutants. 51 exhibited favorable pharmacokinetic properties and good brain penetration, with a brain/plasma ratio of 3.05, and significantly suppressed the growth of tumor in a tamoxifen-resistant MCF-7 Tam1 xenograft model. Overall, the study demonstrates 51 as a highly potent, oral, and brain penetrant ER degrader and pure antagonist, showing a good potential in overcoming endocrine resistance.
Collapse
Affiliation(s)
- Dan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhengyu Lu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yongqi He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xin-Yu Leng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Meng
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Lei
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Deyu Kong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lulu Sun
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wenhao Hu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yushe Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
Lu Y, Zhao Z, Wang S, Zheng T, Chen X, Liu L, Zhu Y, Liu W. Discovery of non-antiproliferative selective estrogen receptor degraders (SERDs) based on scaffold optimization of elacestrant. Eur J Med Chem 2024; 279:116897. [PMID: 39353239 DOI: 10.1016/j.ejmech.2024.116897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
Elacestrant, the first oral selective estrogen receptor degrader (SERD), has been approved for ER positive breast cancer in 2023. Recent study showed that elacestrant has moderate pharmacokinetic property and the oral bioavailability is 11 %. In this study, we have performed docking analyses of elacestrant with different cytochrome P450 isoforms. The results indicated that tetrahydronaphthalene scaffold of elacestrant located closely to Heme iron center of P450s and might undergo rapid metabolism by CYP3A4. Therefore, we have changed the tertiary carbon atom to nitrogen atom of the scaffold to attenuate the metabolic effect. The most interesting finding is that compound B16 exhibited significant degradation of ERα at 5 μM but didn't show antiproliferative activity at high concentrations in MCF-7 and T47D cells. Compound B16 may serve as an ER probe to investigate ER status in ER positive breast cancer cells.
Collapse
Affiliation(s)
- Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Zhihao Zhao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shanglong Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tianpeng Zheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xuejie Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lijuan Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuqing Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
14
|
Dai R, Bao X, Liu C, Yin X, Zhu Z, Zheng Z, Wang B, Yang K, Wen H, Li W, Zhu H, Du Q, Liu J. Drug discovery of N-methyl-pyrazole derivatives as potent selective estrogen receptor degrader (SERD) for the treatment of breast cancer. Eur J Med Chem 2024; 279:116894. [PMID: 39357315 DOI: 10.1016/j.ejmech.2024.116894] [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: 08/01/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
Nowadays, ERα is considered to be a primary target for the treatment of breast cancer, and selective estrogen receptor degraders (SERDs) are emerging as promising antitumor agents. By analysing ERα-SERDs complexes, the pharmacophore features of SERDs and the crucial protein-ligand interactions were identified. Then, by utilizing the scaffold-hopping and bioisosteres strategy, 23 novel derivatives were designed, synthesized and biologically evaluated. Among these derivatives, A20 exhibited potent ERα binding affinity (IC50 = 24.0 nM), degradation ability (EC50 = 5.3 nM), excellent ER selectivity, and outstanding anti-proliferative effects on MCF-7 cells (IC50 = 0.28 nM). Further biological studies revealed that A20 could degrade ERα through proteasome-mediated pathway, suppress signal transduction of MCF-7 cells, and arrest the cell cycle in G1 phase. Moreover, A20 showed excellent antitumor effect (TGI = 92.98 %, 30 mg kg-1 day-1) in the MCF-7 xenograft model in vivo with good safety and favorable pharmacokinetics (F = 39.6 %), making it a promising candidate for the treatment of breast cancer.
Collapse
Affiliation(s)
- Rupeng Dai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xueting Bao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chao Liu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029, China; School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xunkai Yin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhenzhen Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhe Zheng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Bo Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Kundi Yang
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Hongmei Wen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Haohao Zhu
- The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China.
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China; Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Jian Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
15
|
Astore S, Oneda E, Zaniboni A. A therapeutic algorithm guiding subsequent therapy selection after CDK4/6 inhibitors' failure: A review of current and investigational treatment for HR+/Her2- breast cancer. Crit Rev Oncol Hematol 2024; 204:104535. [PMID: 39433229 DOI: 10.1016/j.critrevonc.2024.104535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 10/23/2024] Open
Abstract
The first-line combination therapies utilizing cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) with endocrine therapy (ET) have significantly impacted the course of hormone receptor positive (HR+)/Human Epidermal Growth Factor Receptor 2 negative (HER2-) advanced breast cancer (ABC). However, resistance often emerges, leading to a molecularly different disease. Estrogen receptor one (ESR1) gene mutations, driving resistance to aromatase inhibitors (AIs), may guide the use of fulvestrant or emerging oral selective estrogen receptor degraders (SERDs) like elacestrant. The dynamic nature of ESR1 mutations suggests potential guidance for continuing CDK4/6i therapy beyond progression. Targeting mutations like breast cancer gene 1 and 2 (BRCA 1/2) with Poly (ADP-ribose) polymerase (PARP) inhibitors or the PI3K/AKT/mTOR pathway provides therapeutic options. The advent of antibody-drug conjugates (ADCs) like trastuzumab deruxtecan (T-DXd) and novel agents targeting Trophoblast cell surface antigen-2 (Trop-2) introduces further complexity, underscoring the need for early intervention targeting specific genomic alterations in metastatic BC.
Collapse
Affiliation(s)
- Serena Astore
- Medical Oncology Dept. Fondazione Poliambulanza, Brescia, Italy.
| | - Ester Oneda
- Medical Oncology Dept. Fondazione Poliambulanza, Brescia, Italy
| | | |
Collapse
|
16
|
Palaniappan M. Current Therapeutic Opportunities for Estrogen Receptor Mutant Breast Cancer. Biomedicines 2024; 12:2700. [PMID: 39767607 PMCID: PMC11673253 DOI: 10.3390/biomedicines12122700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Estrogen receptor α (ERα) drives two out of three breast cancers and therefore ERα is a major therapeutic target for ER-positive breast cancer patients. Drugs that inhibit ERα activity or block estrogen synthesis in the body are currently being used in the clinic to treat ER-positive breast cancer and have been quite successful in controlling breast cancer progression for the majority of patients. However, ER-positive breast cancer often becomes resistant to these endocrine therapies, leading to endocrine-resistant metastatic breast cancer, a very aggressive cancer that leads to death. Recent large-scale genomic studies have revealed a series of activating somatic mutations in the ERα gene (ESR1) in endocrine-resistant metastatic breast cancer patients. Of these, Y537S and D538G mutations are found at a much higher rate in patients with metastatic breast cancer. Remarkably, these mutations produce an ERα with much higher transcriptional activity than wild type in the absence of estradiol, and traditional endocrine therapy has poor efficacy against ER mutants. Therefore, the development of new drugs that target ER mutants is an unmet clinical need for endocrine-resistant metastatic breast cancer. This review summarizes the recent preclinical and clinical trials targeting estrogen receptor mutant breast cancer.
Collapse
Affiliation(s)
- Murugesan Palaniappan
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA;
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
17
|
Bai YR, Seng DJ, Xu Y, Zhang YD, Zhou WJ, Jia YY, Song J, He ZX, Liu HM, Yuan S. A comprehensive review of small molecule drugs approved by the FDA in 2023: Advances and prospects. Eur J Med Chem 2024; 276:116706. [PMID: 39053188 DOI: 10.1016/j.ejmech.2024.116706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
In 2023, the U.S. Food and Drug Administration has approved 55 novel medications, consisting of 17 biologics license applications and 38 new molecular entities. Although the biologics license applications including antibody and enzyme replacement therapy set a historical record, the new molecular entities comprising small molecule drugs, diagnostic agent, RNA interference therapy and biomacromolecular peptide still account for over 50 % of the newly approved medications. The novel and privileged scaffolds derived from drugs, active molecules and natural products are consistently associated with the discovery of new mechanisms, the expansion of clinical indications and the reduction of side effects. Moreover, the structural modifications based on the promising scaffolds can provide the clinical candidates with the improved biological activities, bypass the patent protection and greatly shorten the period of new drug discovery. Therefore, conducting an appraisal of drug approval experience and related information will expedite the identification of more potent drug molecules. In this review, we comprehensively summarized the pertinent information encompassing the clinical application, mechanism, elegant design and development processes of 28 small molecule drugs, and expected to provide the promising structural basis and design inspiration for pharmaceutical chemists.
Collapse
Affiliation(s)
- Yi-Ru Bai
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China; School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou, 450001, China
| | - Dong-Jie Seng
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Ying Xu
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Yao-Dong Zhang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Wen-Juan Zhou
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Yang-Yang Jia
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhang-Xu He
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Hong-Min Liu
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China; School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou, 450001, China.
| | - Shuo Yuan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China; School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
18
|
Lloyd MR, Jhaveri K, Kalinsky K, Bardia A, Wander SA. Precision therapeutics and emerging strategies for HR-positive metastatic breast cancer. Nat Rev Clin Oncol 2024; 21:743-761. [PMID: 39179659 DOI: 10.1038/s41571-024-00935-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Anti-oestrogen-based therapies, often combined with a CDK4/6 inhibitor, are the current standard-of-care first-line therapy for patients with advanced-stage hormone receptor-positive (HR+) breast cancer. Resistance to anti-oestrogen agents inevitably occurs, mediated by oestrogen receptor (ER)-dependent or ER-independent mechanisms that drive tumour progression. Emerging endocrine therapies include, but are not limited to, next-generation oral ER degraders and proteolysis targeting chimeras, which might be particularly effective in patients with ESR1-mutant breast cancer. Furthermore, cancers harbouring driver alterations in oncogenic signalling pathways, including AKT and PI3K, might be susceptible to novel combination strategies involving targeted inhibitors. Next-generation CDK2/4 inhibitors are an area of active clinical investigation, and efforts are ongoing to evaluate the role of sequential CDK inhibition. Approved and emerging antibody-drug conjugates exploiting novel target antigens have also demonstrated promising clinical activity. These novel agents, as well as further identification and characterization of predictive biomarkers, will hopefully continue to improve clinical outcomes, reduce the incidence of toxicities, and limit the extent of overtreatment in this population. In this Review, we describe the evolving treatment paradigm for patients with metastatic HR+ breast cancer in light of the growing armamentarium of drugs and biomarkers that will help to shape the future therapeutic landscape. These strategies are expected to involve tumour molecular profiling to enable the delivery of precision medicine.
Collapse
Affiliation(s)
| | - Komal Jhaveri
- Memorial Sloan Kettering Evelyn H. Lauder Breast Center, New York, NY, USA
| | - Kevin Kalinsky
- Winship Cancer Institute at Emory University, Atlanta, GA, USA
| | - Aditya Bardia
- UCLA Health Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Seth A Wander
- Massachusetts General Hospital Cancer Center, Boston, MA, USA.
| |
Collapse
|
19
|
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: 0] [Impact Index Per Article: 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
|
20
|
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
|
21
|
Hamilton E, Oliveira M, Turner N, García-Corbacho J, Hernando C, Ciruelos EM, Kabos P, Ruiz-Borrego M, Armstrong A, Patel MR, Vaklavas C, Twelves C, Boni V, Incorvati J, Brier T, Gibbons L, Klinowska T, Lindemann JPO, Morrow CJ, Sykes A, Baird RD. A phase I dose escalation and expansion trial of the next-generation oral SERD camizestrant in women with ER-positive, HER2-negative advanced breast cancer: SERENA-1 monotherapy results. Ann Oncol 2024; 35:707-717. [PMID: 38729567 DOI: 10.1016/j.annonc.2024.04.012] [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: 01/22/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND SERENA-1 (NCT03616587) is a phase I, multi-part, open-label study of camizestrant in pre- and post-menopausal women with estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer. Parts A and B aim to determine the safety and tolerability of camizestrant monotherapy and define doses for clinical evaluation. PATIENTS AND METHODS Women aged ≥18 years with metastatic or recurrent ER+, HER2- breast cancer, refractory (or intolerant) to therapy, were assigned 25 mg up to 450 mg once daily (QD; escalation) or 75, 150, or 300 mg QD (expansion). Safety and tolerability, antitumor efficacy, pharmacokinetics, and impact on mutations in the estrogen receptor gene (ESR1m) circulating tumor (ct)DNA levels were assessed. RESULTS By 9 March 2021, 108 patients received camizestrant monotherapy at 25-450 mg doses. Of these, 93 (86.1%) experienced treatment-related adverse events (TRAEs), 82.4% of which were grade 1 or 2. The most common TRAEs were visual effects (56%), (sinus) bradycardia (44%), fatigue (26%), and nausea (15%). There were no TRAEs grade 3 or higher, or treatment-related serious adverse events at doses ≤150 mg. Median tmax was achieved ∼2-4 h post-dose at all doses investigated, with an estimated half-life of 20-23 h. Efficacy was observed at all doses investigated, including in patients with prior cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) and/or fulvestrant treatment, with and without baseline ESR1 mutations, and with visceral disease, including liver metastases. CONCLUSIONS Camizestrant is a next-generation oral selective ER antagonist and degrader (SERD) and pure ER antagonist with a tolerable safety profile. The pharmacokinetics profile supports once-daily dosing, with evidence of pharmacodynamic and clinical efficacy in heavily pre-treated patients, regardless of ESR1m. This study established 75-, 150-, and 300-mg QD doses for phase II testing (SERENA-2, NCT04214288 and SERENA-3, NCT04588298).
Collapse
Affiliation(s)
- E Hamilton
- Sarah Cannon Research Institute, Nashville, USA
| | - M Oliveira
- Medical Oncology Department, Vall d'Hebron University Hospital and Breast Cancer Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - N Turner
- Breast Cancer Now, Toby Robins Research Centre, Institute of Cancer Research, London, UK
| | | | - C Hernando
- Department of Medical Oncology, Hospital Clinico Universitario de Valencia, Biomedical Research Institute (INCLIVA), Valencia
| | - E M Ciruelos
- Medical Oncology Department, 12 de Octubre University Hospital, Madrid, Spain
| | - P Kabos
- Division of Medical Oncology, University of Colorado, Denver, USA
| | - M Ruiz-Borrego
- Department of Medical Oncology, H U Virgen del Rocio, Seville, Spain
| | - A Armstrong
- The Christie NHS Foundation Trust and the University of Manchester, Manchester, UK
| | - M R Patel
- Florida Cancer Specialists/Sarah Cannon Research Institute/Sarasota Memorial Hospital, Sarasota
| | - C Vaklavas
- Huntsman Cancer Institute, University of Utah, Salt Lake City, USA
| | - C Twelves
- Leeds Teaching Hospitals NHS Trust and University of Leeds, Leeds, UK
| | - V Boni
- START Madrid, CIOCC, Madrid, Spain
| | - J Incorvati
- Fox Chase Cancer Center, East Norriton-Hospital Outpatient Center, Philadelphia, USA
| | - T Brier
- Research and Early Development, Oncology R&D, AstraZeneca, Cambridge
| | - L Gibbons
- Research and Early Development, Oncology R&D, AstraZeneca, Cambridge
| | - T Klinowska
- Late Development, Oncology R&D, AstraZeneca, Cambridge
| | - J P O Lindemann
- Research and Early Development, Oncology R&D, AstraZeneca, Cambridge
| | - C J Morrow
- Research and Early Development, Oncology R&D, AstraZeneca, Cambridge
| | - A Sykes
- Research and Early Development, Oncology R&D, AstraZeneca, Cambridge
| | - R D Baird
- Cancer Research UK, Cambridge Centre, Cambridge, UK.
| |
Collapse
|
22
|
Hageman E, Lussier ME. Elacestrant for ER-Positive HER2-Negative Advanced Breast Cancer. Ann Pharmacother 2024; 58:849-856. [PMID: 37888769 DOI: 10.1177/10600280231206131] [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: 10/28/2023] Open
Abstract
OBJECTIVE This article aims to discuss elacestrant, an oral selective estrogen receptor downregulator approved by the Food and Drug Administration (FDA) in January 2023 for the treatment of hormone receptor positive (HR+) human epidermal growth factor receptor 2 negative (HER2-) advanced breast cancer. DATA SOURCES PubMed, Embase, Medline, Clinicaltrials.gov, and the National Comprehensive Cancer Network (NCCN) were searched from inception to August 31, 2023. STUDY SELECTION AND DATA EXTRACTION Clinical trials published in English were included and relevant information regarding methodology and results were extracted. DATA SYNTHESIS Phase 1 and 3 trials showed elacestrant was safe and improved progression-free survival in patients with endocrine receptor 1 (ESR1) mutations who failed cyclin-dependent kinase 4/6 inhibitor (CDK 4/6i) plus 1 prior endocrine therapy compared with standard of care (SOC) (fulvestrant, anastrozole, letrozole, or exemestane monotherapy). RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE IN COMPARISON TO EXISTING DRUGS Elacestrant maintains a comparable adverse event profile with other endocrine therapies and offers an alternative to typical sequential therapy which can delay the use of or be used after traditional chemotherapy. Elacestrant is currently being studied in CDK 4/6 inhibitor naïve patients and as a component of combination therapy for first-line use which could lead to future indications. CONCLUSIONS Elacestrant gained FDA approval in January 2023 and can be considered in patients with HR+ HER2- advanced breast cancer and ESR1 mutations who have progressed despite therapy with either CDK 4/6i plus aromatase inhibitors (AI) or fulvestrant or chemotherapy.
Collapse
Affiliation(s)
- Elizabeth Hageman
- Department of Pharmacy Practice, Binghamton University School of Pharmacy and Pharmaceutical Sciences, Johnson City, NY, USA
| | - Mia E Lussier
- Department of Pharmacy Practice, Binghamton University School of Pharmacy and Pharmaceutical Sciences, Johnson City, NY, USA
| |
Collapse
|
23
|
Keenan JC, Medford AJ, Dai CS, Wander SA, Spring LM, Bardia A. Novel oral selective estrogen receptor degraders (SERDs) to target hormone receptor positive breast cancer: elacestrant as the poster-child. Expert Rev Anticancer Ther 2024; 24:397-405. [PMID: 38642015 DOI: 10.1080/14737140.2024.2346188] [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: 10/11/2023] [Accepted: 04/18/2024] [Indexed: 04/22/2024]
Abstract
INTRODUCTION Estrogen receptor positive (ER+) breast cancer is the most common breast cancer subtype, and therapeutic management relies primarily on inhibiting ER signaling. In the metastatic setting, ER signaling is typically targeted by selective estrogen receptor degraders (SERDs) or aromatase inhibitors (AIs), the latter of which prevent estrogen production. Activating ESR1 mutations are among the most common emergent breast cancer mutations and confer resistance to AIs. AREAS COVERED Until 2023, fulvestrant was the only approved SERD; fulvestrant is administered intramuscularly, and in some cases may also have limited efficacy in the setting of certain ESR1 mutations. In 2023, the first oral SERD, elacestrant, was approved for use in ESR1-mutated, ER+/HER2- advanced breast cancer and represents a new class of therapeutic options. While the initial approval was as monotherapy, ongoing studies are evaluating elacestrant (as well as other oral SERDs) in combination with other therapies including CDK4/6 inhibitors and PI3K inhibitors, which parallels the current combination uses of fulvestrant. EXPERT OPINION Elacestrant's recent approval sheds light on the use of biomarkers such as ESR1 to gauge a tumor's endocrine sensitivity. Ongoing therapeutic and correlative biomarker studies will offer new insight and expanding treatment options for patients with advanced breast cancer.
Collapse
Affiliation(s)
- Jennifer C Keenan
- Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Arielle J Medford
- Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Charles S Dai
- Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Seth A Wander
- Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Laura M Spring
- Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Aditya Bardia
- Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| |
Collapse
|
24
|
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
|
25
|
Guglielmi G, Del Re M, Gol LS, Bengala C, Danesi R, Fogli S. Pharmacological insights on novel oral selective estrogen receptor degraders in breast cancer. Eur J Pharmacol 2024; 969:176424. [PMID: 38402929 DOI: 10.1016/j.ejphar.2024.176424] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 02/27/2024]
Abstract
The therapeutic landscape of estrogen receptor (ER)-positive breast cancer includes endocrine treatments with aromatase inhibitors (AIs), selective estrogen receptor modulators (SERMs), and selective estrogen receptor degraders (SERDs). Fulvestrant is the first approved SERD with proven efficacy and good tolerability in clinical practice. However, drug resistance, low receptor affinity, and parental administration stimulated the search for new oral SERDs opening a new therapeutic era in ER + breast cancer. Elacestrant is an orally bioavailable SERD that has been recently approved by the FDA for postmenopausal women with ER+, human epidermal growth factor receptor 2-negative (HER2-), estrogen receptor 1 (ESR1)-mutated advanced or metastatic breast cancer with disease progression following at least one line of endocrine therapy. Other molecules of the same class currently tested in clinical trials are amcenestrant, giredestrant, camizestrant, and imlunestrant. The current review article offers a detailed pharmacological perspective of this emerging drug class, which may help with their possible future clinical applications.
Collapse
Affiliation(s)
- Giorgio Guglielmi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Leila Sadeghi Gol
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Carmelo Bengala
- Clinical Oncology Unit 1, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy; Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy.
| | - Stefano Fogli
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
26
|
Hu J, Zhu BY, Niu ZX. Catalysts of Healing: A Symphony of Synthesis and Clinical Artistry in Small-Molecule Agents for Breast Cancer Alleviation. Molecules 2024; 29:1166. [PMID: 38474678 DOI: 10.3390/molecules29051166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Breast cancer, characterized by its molecular intricacy, has witnessed a surge in targeted therapeutics owing to the rise of small-molecule drugs. These entities, derived from cutting-edge synthetic routes, often encompassing multistage reactions and chiral synthesis, target a spectrum of oncogenic pathways. Their mechanisms of action range from modulating hormone receptor signaling and inhibiting kinase activity, to impeding DNA damage repair mechanisms. Clinical applications of these drugs have resulted in enhanced patient survival rates, reduction in disease recurrence, and improved overall therapeutic indices. Notably, certain molecules have showcased efficacy in drug-resistant breast cancer phenotypes, highlighting their potential in addressing treatment challenges. The evolution and approval of small-molecule drugs have ushered in a new era for breast cancer therapeutics. Their tailored synthetic pathways and defined mechanisms of action have augmented the precision and efficacy of treatment regimens, paving the way for improved patient outcomes in the face of this pervasive malignancy. The present review embarks on a detailed exploration of small-molecule drugs that have secured regulatory approval for breast cancer treatment, emphasizing their clinical applications, synthetic pathways, and distinct mechanisms of action.
Collapse
Affiliation(s)
- Jing Hu
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Bi-Yue Zhu
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, Chongqing 400015, China
| | - Zhen-Xi Niu
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| |
Collapse
|
27
|
Min J, Liu X, Peng R, Chen CC, Wang W, Guo RT. New generation estrogen receptor-targeted agents in breast cancer: present situation and future prospectives. ACTA MATERIA MEDICA 2024; 3:57-71. [PMID: 39373009 PMCID: PMC11450757 DOI: 10.15212/amm-2024-0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Endocrine therapy which blocking the signaling of estrogen receptor, has long been effective for decades as a primary treatment choice for breast cancer patients expressing ER. However, the issue of drug resistance poses a significant clinical challenge. It's critically important to create new therapeutic agents that can suppress ERα activity, particularly in cases of ESR1 mutations. This review highlights recent efforts in drug development of next generation ER-targeted agents, including oral selective ER degraders (SERDs), proteolysis targeting chimera (PROTAC) ER degraders, other innovative molecules such as complete estrogen receptor antagonists (CERANs) and selective estrogen receptor covalent antagonists (SERCAs). The drug design, efficacy and clinical trials for each compound were detailed.
Collapse
Affiliation(s)
- Jian Min
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Xin Liu
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Rouming Peng
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Chun-Chi Chen
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, School of Life Sciences, Hubei University, Wuhan, 430062, China
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Wei Wang
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Rey-Ting Guo
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, School of Life Sciences, Hubei University, Wuhan, 430062, China
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Venetis K, Pepe F, Pescia C, Cursano G, Criscitiello C, Frascarelli C, Mane E, Russo G, Taurelli Salimbeni B, Troncone G, Guerini Rocco E, Curigliano G, Fusco N, Malapelle U. ESR1 mutations in HR+/HER2-metastatic breast cancer: Enhancing the accuracy of ctDNA testing. Cancer Treat Rev 2023; 121:102642. [PMID: 37864956 DOI: 10.1016/j.ctrv.2023.102642] [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: 08/19/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 10/23/2023]
Abstract
Activating mutations of the estrogen receptor alpha gene (ESR1) are common mechanisms of endocrine therapy (ET) resistance in hormone receptor-positive (HR + )/Human Epidermal Growth Factor Receptor 2 (HER2)-negative metastatic breast cancer (MBC). Recent clinical findings emphasize that both old and new generations of selective ER degraders (SERDs) demonstrate enhanced clinical effectiveness in patients with MBC who have detectable ESR1 mutations via liquid biopsy. This stands in contrast to individuals with MBC carrying these mutations and undergoing conventional endocrine monotherapies like aromatase inhibitors (AIs). Liquid biopsy, particularly the analysis of circulating tumor DNA (ctDNA), has emerged as a promising, minimally invasive alternative to conventional tissue-based testing for identifying ESR1 mutations. Within the context of the PADA-1 and EMERALD trials, distinct molecular methodologies and assays, specifically digital droplet PCR (ddPCR) and next-generation sequencing (NGS), have been employed to evaluate the mutational status of ESR1 within ctDNA. This manuscript critically examines the advantages and indications of various ctDNA testing methods on liquid biopsy for HR+/HER2-negative MBC. Specifically, we delve into the capabilities of ddPCR and NGS in identifying ESR1 mutations. Each methodology boasts unique strengths and limitations: ddPCR excels in its analytical sensitivity for pinpointing hotspot mutations, while NGS offers comprehensive coverage of the spectrum of ESR1 mutations. The significance of meticulous sample handling and timely analysis is emphasized, acknowledging the transient nature of cfDNA. Furthermore, we underscore the importance of detecting sub-clonal ESR1 mutations, as these variants can exert a pivotal influence on predicting both endocrine therapy resistance and responsiveness to SERDs. In essence, this work discusses the role of ctDNA analysis for detecting ESR1 mutations and their implications in tailoring effective therapeutic strategies for HR+/HER2- MBC.
Collapse
Affiliation(s)
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | - Carlo Pescia
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy; School of Pathology, University of Milan, Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Eltjona Mane
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | | | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | - Elena Guerini Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Naples, Italy
| |
Collapse
|
31
|
Nemati F, de Koning L, Gentien D, Assayag F, Henry E, Ait Rais K, Pierron G, Mariani O, Nijnikoff M, Champenois G, Nicolas A, Meseure D, Gardrat S, Servant N, Hupé P, Kamal M, Le Tourneau C, Piperno-Neumann S, Rodrigues M, Roman-Roman S, Decaudin D, Mariani P, Cassoux N. Patient Derived Xenografts (PDX) Models as an Avatar to Assess Personalized Therapy Options in Uveal Melanoma: A Feasibility Study. Curr Oncol 2023; 30:9090-9103. [PMID: 37887557 PMCID: PMC10604955 DOI: 10.3390/curroncol30100657] [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: 08/14/2023] [Revised: 09/13/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Uveal melanoma is the most common primary intraocular malignancy in adults. Up to 50% of UM patients develop metastatic disease, usually in the liver. When metastatic, the prognosis is poor, and few treatment options exist. Here, we investigated the feasibility of establishing patient-derived xenografts (PDXs) from a patient's tumor in order to screen for therapies that the patient could benefit from. Samples obtained from 29 primary tumors and liver metastases of uveal melanoma were grafted into SCID mice. PDX models were successfully established for 35% of primary patient tumors and 67% of liver metastases. The tumor take rate was proportional to the risk of metastases. PDXs showed the same morphology, the same GNAQ/11, BAP1, and SF3B1 mutations, and the same chromosome 3 and 8q status as the corresponding patient samples. Six PDX models were challenged with two compounds for 4 weeks. We show that, for 31% of patients with high or intermediate risk of metastasis, the timing to obtain efficacy results on PDX models derived from their primary tumors was compatible with the selection of the therapy to treat the patient after relapse. PDXs could thus be a valid tool ("avatar") to select the best personalized therapy for one third of patients that are most at risk of relapse.
Collapse
Affiliation(s)
- Fariba Nemati
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University Paris, 26 rue d’Ulm, CEDEX 05, 75248 Paris, France
| | - Leanne de Koning
- Translational Research Department, Institut Curie, PSL University Paris, 75248 Paris, France; (L.d.K.)
| | - David Gentien
- Genomics Platform, Translational Research Department, Institut Curie, PSL Research University, 75248 Paris, France
| | - Franck Assayag
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University Paris, 26 rue d’Ulm, CEDEX 05, 75248 Paris, France
| | - Emilie Henry
- Genomics Platform, Translational Research Department, Institut Curie, PSL Research University, 75248 Paris, France
| | - Khadija Ait Rais
- Department of Genetics, Institut Curie, PSL Research University, 75248 Paris, France
| | - Gaelle Pierron
- Department of Genetics, Institut Curie, PSL Research University, 75248 Paris, France
| | - Odette Mariani
- Biological Resource Center, Department of Pathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Michèle Nijnikoff
- Biological Resource Center, Department of Pathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Gabriel Champenois
- Department of Biopathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - André Nicolas
- Department of Biopathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Didier Meseure
- Department of Biopathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Sophie Gardrat
- Department of Biopathology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Nicolas Servant
- Institut Curie, INSERM U900, CBIO-Centre for Computational Biology, Mines Paris Tech, PSL-Research University, 75248 Paris, France
| | - Philippe Hupé
- Institut Curie, INSERM U900, CBIO-Centre for Computational Biology, Mines Paris Tech, PSL-Research University, 75248 Paris, France
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, 75248 Paris, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, 75248 Paris, France
- INSERM U900 Research Unit, Institut Curie, 92064 Saint-Cloud, France
- Paris-Saclay University, 75248 Paris, France
| | - Sophie Piperno-Neumann
- Department of Medical Oncology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Manuel Rodrigues
- Department of Medical Oncology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Sergio Roman-Roman
- Translational Research Department, Institut Curie, PSL University Paris, 75248 Paris, France; (L.d.K.)
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University Paris, 26 rue d’Ulm, CEDEX 05, 75248 Paris, France
- Department of Medical Oncology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Pascale Mariani
- Department of Surgical Oncology, Institut Curie, PSL Research University, 75248 Paris, France
| | - Nathalie Cassoux
- Department of Oncological Ophthalmology, Institut Curie, Université Paris Cité, 75248 Paris, France
| |
Collapse
|
32
|
Will M, Liang J, Metcalfe C, Chandarlapaty S. Therapeutic resistance to anti-oestrogen therapy in breast cancer. Nat Rev Cancer 2023; 23:673-685. [PMID: 37500767 PMCID: PMC10529099 DOI: 10.1038/s41568-023-00604-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 07/29/2023]
Abstract
The hormone receptor oestrogen receptor-α (ER) orchestrates physiological mammary gland development, breast carcinogenesis and the progression of breast tumours into lethal, treatment-refractory systemic disease. Selective antagonism of ER signalling has been one of the most successful therapeutic approaches in oncology, benefiting patients as both a cancer preventative measure and a cancer treatment strategy. However, resistance to anti-oestrogen therapy is a major clinical challenge. Over the past decade, we have gained an understanding of how breast cancers evolve under the pressure of anti-oestrogen therapy. This is best depicted by the case of oestrogen-independent mutations in the gene encoding ER (ESR1), which are virtually absent in primary breast cancer but highly prevalent (20-40%) in anti-oestrogen-treated metastatic disease. These and other findings highlight the 'evolvability' of ER+ breast cancer and the need to understand molecular processes by which this evolution occurs. Recent development and approval of next-generation ER antagonists to target ESR1-mutant breast cancer underscores the clinical importance of this evolvability and sets a new paradigm for the treatment of ER+ breast cancers.
Collapse
Affiliation(s)
- Marie Will
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jackson Liang
- Department of Oncology Biomarker Development, Genentech, South San Francisco, CA, USA
| | - Ciara Metcalfe
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA.
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
33
|
Miziak P, Baran M, Błaszczak E, Przybyszewska-Podstawka A, Kałafut J, Smok-Kalwat J, Dmoszyńska-Graniczka M, Kiełbus M, Stepulak A. Estrogen Receptor Signaling in Breast Cancer. Cancers (Basel) 2023; 15:4689. [PMID: 37835383 PMCID: PMC10572081 DOI: 10.3390/cancers15194689] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Estrogen receptor (ER) signaling is a critical regulator of cell proliferation, differentiation, and survival in breast cancer (BC) and other hormone-sensitive cancers. In this review, we explore the mechanism of ER-dependent downstream signaling in BC and the role of estrogens as growth factors necessary for cancer invasion and dissemination. The significance of the clinical implications of ER signaling in BC, including the potential of endocrine therapies that target estrogens' synthesis and ER-dependent signal transmission, such as aromatase inhibitors or selective estrogen receptor modulators, is discussed. As a consequence, the challenges associated with the resistance to these therapies resulting from acquired ER mutations and potential strategies to overcome them are the critical point for the new treatment strategies' development.
Collapse
Affiliation(s)
- Paulina Miziak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Marzena Baran
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Ewa Błaszczak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Alicja Przybyszewska-Podstawka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Joanna Kałafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Jolanta Smok-Kalwat
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland;
| | - Magdalena Dmoszyńska-Graniczka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Michał Kiełbus
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| |
Collapse
|
34
|
Beddok A, Cottu P, Fourquet A, Kirova Y. [Radiotherapy and targeted therapy for the management of breast cancer: A review]. Cancer Radiother 2023; 27:447-454. [PMID: 37173174 DOI: 10.1016/j.canrad.2023.02.002] [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: 08/21/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 05/15/2023]
Abstract
The purpose of this study was to review the current knowledge regarding combinations of the most commonly used targeted therapies or those under development for the management of breast cancer with radiation therapy. Several studies have shown that the combination of radiation therapy and tamoxifen increased the risk of radiation-induced lung toxicity; therefore, the two modalities are generally not given concurrently. The combination of HER2 inhibitors (trastuzumab, pertuzumab) and radiation therapy appeared to be safe. However, trastuzumab emtansine (T-DM1) should not be given concomitantly with brain radiation therapy because this combination may increase the risk of brain radionecrosis. The combination of radiation therapy with other new targeted therapies such as new selective estrogen receptor modulators (SERDs), lapatinib, cell cycle inhibitors, immune checkpoint inhibitors, or molecules acting on DNA damage repair seems feasible but has been mainly evaluated on retrospective or prospective studies with small numbers of patients. Moreover, there is a great heterogeneity between these studies regarding the dose and fractionation used in radiotherapy, the dosage of systemic treatments and the sequence of treatments used. Therefore, the combination of these new molecules with radiotherapy should be proposed sparingly, under close monitoring, pending the ongoing prospective studies cited in this review.
Collapse
Affiliation(s)
- A Beddok
- Laboratoire d'imagerie translationnelle en oncologie (Lito), Institut Curie, université PSL, université Paris Saclay, Inserm, 91898 Orsay, France; Département de radiothérapie oncologique, institut Curie, université PSL, Centre de protonthérapie, centre universitaire, 91898 Orsay, France.
| | - P Cottu
- Département d'oncologie médicale, institut Curie, Paris, France
| | - A Fourquet
- Département de radiothérapie oncologique, institut Curie, université PSL, Paris, France
| | - Y Kirova
- Département de radiothérapie oncologique, institut Curie, université PSL, Paris, France
| |
Collapse
|
35
|
Dubash TD, Bardia A, Chirn B, Reeves BA, LiCausi JA, Burr R, Wittner BS, Rai S, Patel H, Bihani T, Arlt H, Bidard FC, Kaklamani VG, Aftimos P, Cortés J, Scartoni S, Fiascarelli A, Binaschi M, Habboubi N, Iafrate AJ, Toner M, Haber DA, Maheswaran S. Modeling the novel SERD elacestrant in cultured fulvestrant-refractory HR-positive breast circulating tumor cells. Breast Cancer Res Treat 2023; 201:43-56. [PMID: 37318638 PMCID: PMC10300156 DOI: 10.1007/s10549-023-06998-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/26/2023] [Indexed: 06/16/2023]
Abstract
PURPOSE Metastatic hormone receptor-positive (HR+) breast cancer initially responds to serial courses of endocrine therapy, but ultimately becomes refractory. Elacestrant, a new generation FDA-approved oral selective estrogen receptor degrader (SERD) and antagonist, has demonstrated efficacy in a subset of women with advanced HR+breast cancer, but there are few patient-derived models to characterize its effect in advanced cancers with diverse treatment histories and acquired mutations. METHODS We analyzed clinical outcomes with elacestrant, compared with endocrine therapy, among women who had previously been treated with a fulvestrant-containing regimen from the recent phase 3 EMERALD Study. We further modeled sensitivity to elacestrant, compared with the currently approved SERD, fulvestrant in patient-derived xenograft (PDX) models and cultured circulating tumor cells (CTCs). RESULTS Analysis of the subset of breast cancer patients enrolled in the EMERALD study who had previously received a fulvestrant-containing regimen indicates that they had better progression-free survival with elacestrant than with standard-of-care endocrine therapy, a finding that was independent estrogen receptor (ESR1) gene mutations. We modeled elacestrant responsiveness using patient-derived xenograft (PDX) models and in ex vivo cultured CTCs derived from patients with HR+breast cancer extensively treated with multiple endocrine therapies, including fulvestrant. Both CTCs and PDX models are refractory to fulvestrant but sensitive to elacestrant, independent of mutations in ESR1 and Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha (PIK3CA) genes. CONCLUSION Elacestrant retains efficacy in breast cancer cells that have acquired resistance to currently available ER targeting therapies. Elacestrant may be an option for patients with HR+/HER2- breast cancer whose disease progressed on fulvestrant in the metastatic setting. TRANSLATIONAL RELEVANCE Serial endocrine therapy is the mainstay of management for metastatic HR+breast cancer, but acquisition of drug resistance highlights the need for better therapies. Elacestrant is a recently FDA-approved novel oral selective estrogen receptor degrader (SERD), with demonstrated efficacy in the EMERALD phase 3 clinical trial of refractory HR+breast cancer. Subgroup analysis of the EMERALD clinical trial identifies clinical benefit with elacestrant in patients who had received prior fulvestrant independent of the mutational status of the ESR1 gene, supporting its potential utility in treating refractory HR+breast cancer. Here, we use pre-clinical models, including ex vivo cultures of circulating tumor cells and patient-derived xenografts, to demonstrate the efficacy of elacestrant in breast cancer cells with acquired resistance to fulvestrant.
Collapse
Affiliation(s)
- Taronish D Dubash
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Brian Chirn
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Brittany A Reeves
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Joseph A LiCausi
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Risa Burr
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Ben S Wittner
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Sumit Rai
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | | | | | - Heike Arlt
- Radius Health, Inc, Waltham, MA, 02451, USA
| | | | | | - Philippe Aftimos
- Institut Jules Bordet-Université Libre de Bruxelles, Brussels, Belgium
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Quiron Group, Barcelona, Spain
| | | | | | | | - Nassir Habboubi
- Stemline Therapeutics/Menarini Group, New York, NY, 10022, USA
| | - A John Iafrate
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Mehmet Toner
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA.
- Howard Hughes Medical Institute, Bethesda, MD, 20810, USA.
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
36
|
Beumer JH, Foldi J. Pharmacology and pharmacokinetics of elacestrant. Cancer Chemother Pharmacol 2023; 92:157-163. [PMID: 37314500 PMCID: PMC10713198 DOI: 10.1007/s00280-023-04550-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023]
Abstract
Elacestrant, a novel oral selective estrogen receptor (ER) degrader (SERD), was approved by the Food and Drug Administration (FDA) on January 27, 2023, for use in patients with ER and/or progesterone receptor (PR)-positive and HER2-negative metastatic breast cancer whose tumors harbor an ESR1 missense mutation (ESR1-mut), after at least one line of endocrine therapy (ET). The FDA made its decision based on the randomized phase 3 EMERALD trial, which met its primary endpoint of improved median progression-free survival (mPFS) with elacestrant monotherapy versus standard-of-care endocrine monotherapy in the overall intention to treat population; however, this benefit was largely driven by the ESR1-mut cohort. Elacestrant is a dose-dependent mixed ER agonist/antagonist, which at high doses acts as a direct ER antagonist as well as selective downregulator of ER. It is 11% bioavailable, primarily metabolized by CYP3A4 in the liver and excreted in feces. This leads to drug-drug interactions with strong CYP3A4 inhibitors and inducers, such as itraconazole and rifampin, respectively. In accordance with its clearance route, dose reduction is recommended in patients with moderate hepatic dysfunction but not in renal dysfunction. Studies evaluating elacestrant in severe hepatic dysfunction as well as in patients from racial and ethnic minority groups are ongoing. Overall, elacestrant is the first orally bioavailable SERD approved by the FDA for use in patients with metastatic breast cancer. Current clinical trials are ongoing evaluating it in the adjuvant setting in patients with early stage ER-positive breast cancers.
Collapse
Affiliation(s)
- Jan H Beumer
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Hillman Research Pavilion, Room G27E, 5117 Centre Avenue, Pittsburgh, PA, 15213-1863, USA.
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Julia Foldi
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Hillman Research Pavilion, Room G27E, 5117 Centre Avenue, Pittsburgh, PA, 15213-1863, USA
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
37
|
Shastry M, Hamilton E. Novel Estrogen Receptor-Targeted Agents for Breast Cancer. Curr Treat Options Oncol 2023; 24:821-844. [PMID: 37129836 DOI: 10.1007/s11864-023-01079-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2023] [Indexed: 05/03/2023]
Abstract
OPINION STATEMENT It has become clear that patients whose cancers have progressed post-CDK4/6 inhibitor therapy (CDK4/6i) are not deriving the same magnitude of benefit to subsequent standard endocrine therapy as historical studies would suggest. For example, anticipated duration of benefit to fulvestrant prior to CDK4/6i historically was ~ 5-6 months, and data from the VERONICA and EMERALD trials report less than 2 months. This has magnified our need for novel endocrine agents. Some have argued that patients post-CDK4/6i may just have more endocrine-resistant tumors and perhaps should just receive chemotherapy. While this may be appropriate for some, we do not currently have an assay that reliably predicts whose cancers remain endocrine sensitive and whose are endocrine resistant. ESR1 mutations can enrich for patients whose tumors are more likely to be heavily dependent on estrogen, but this is certainly not the whole answer and many patients without ESR1 mutations continue to derive benefit from subsequent endocrine agents. Most patients would strongly prefer the side effect profile of endocrine agents compared to chemotherapy, and thus, premature use of cytotoxic agents when subsequent ER targeting can control disease is not preferred. These novel ER targeting agents (PROTAC, SERD, SERCA, CERAN) hold great promise to not only outperform standard agents like fulvestrant, but also offer the promise of agents with a different side effect profile that may be more advantageous when compared to menopausal symptoms, hot flashes, arthralgias, and sexual side effects so commonly seen with AIs. We also are likely to see these novel agents move to earlier lines, whether that be 1st line in combination with CDK4/6i or even adjuvant disease.
Collapse
Affiliation(s)
| | - Erika Hamilton
- Sarah Cannon Research Institute, Nashville, TN, USA.
- Tennessee Oncology, 335 24th Ave North, Suite 300, Nashville, TN, 37203, USA.
| |
Collapse
|
38
|
Rej RK, Thomas JE, Acharyya RK, Rae JM, Wang S. Targeting the Estrogen Receptor for the Treatment of Breast Cancer: Recent Advances and Challenges. J Med Chem 2023. [PMID: 37377342 DOI: 10.1021/acs.jmedchem.3c00136] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Estrogen receptor alpha (ERα) is a well-established therapeutic target for the treatment of ER-positive (ER+) breast cancers. Despite the tremendous successes achieved with tamoxifen, a selective ER modulator, and aromatase inhibitors (AIs), resistance to these therapies is a major clinical problem. Therefore, induced protein degradation and covalent inhibition have been pursued as new therapeutic approaches to target ERα. This Perspective summarizes recent progress in the discovery and development of oral selective ER degraders (SERDs), complete estrogen receptor antagonists (CERANs), selective estrogen receptor covalent antagonists (SERCAs), and proteolysis targeting chimera (PROTAC) ER degraders. We focus on those compounds which have been advanced into clinical development.
Collapse
Affiliation(s)
- Rohan Kalyan Rej
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Junius Eugene Thomas
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ranjan Kumar Acharyya
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - James Michael Rae
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
39
|
Ferro A, Generali D, Caffo O, Caldara A, De Lisi D, Dipasquale M, Lorenzi M, Monteverdi S, Fedele P, Ciribilli Y. Oral selective estrogen receptor degraders (SERDs): The new emperors in breast cancer clinical practice? Semin Oncol 2023; 50:90-101. [PMID: 37673696 DOI: 10.1053/j.seminoncol.2023.08.002] [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: 05/03/2023] [Revised: 07/24/2023] [Accepted: 08/10/2023] [Indexed: 09/08/2023]
Abstract
Endocrine therapy (ET) targeting estrogen receptor (ER) signaling is still the mainstay treatment option for early or advanced ER-positive breast cancer (BC) and may involve suppressing estrogen production by means of aromatase inhibitors or directly blocking the ER pathway through selective estrogen receptor modulators such as tamoxifen or selective estrogen receptor degraders such as fulvestrant. However, despite the availability of this armamentarium in clinical practice, de novo or acquired resistance to ET is the main cause of endocrine-based treatment failure leading to the progression of the BC. Recent advances in targeting, modulating, and degrading ERs have led to the development of new drugs capable of overcoming intrinsic or acquired ET resistance related to alterations in the ESR1 gene. The new oral selective estrogen receptor degraders, which are capable of reducing ER protein expression and blocking estrogen-dependent and -independent ER signaling, have a broader spectrum of activity against ESR1 mutations and seem to be a promising means of overcoming the failure of standard ET. The aim of this review is to summarize the development of oral selective estrogen receptor degraders, their current status, and their future perspectives.
Collapse
Affiliation(s)
- Antonella Ferro
- Medical Oncology, Breast Unit Santa Chiara Hospital, APSS Trento, Largo Medaglie D'Oro, Trento, Italy.
| | - Daniele Generali
- UO Patologia Mammaria, Cremona Hospital, ASST Cremona, Italy; Department of Medicine, Surgery and Health Sciences, University of Trieste, Italy
| | - Orazio Caffo
- Medical Oncology Unit, Santa Chiara Hospital, APSS Trento, Italy
| | - Alessia Caldara
- Medical Oncology, Breast Unit Santa Chiara Hospital, APSS Trento, Largo Medaglie D'Oro, Trento, Italy
| | - Delia De Lisi
- Medical Oncology, Breast Unit Santa Chiara Hospital, APSS Trento, Largo Medaglie D'Oro, Trento, Italy
| | - Mariachiara Dipasquale
- Medical Oncology, Breast Unit Santa Chiara Hospital, APSS Trento, Largo Medaglie D'Oro, Trento, Italy
| | - Martina Lorenzi
- Medical Oncology, Breast Unit Santa Chiara Hospital, APSS Trento, Largo Medaglie D'Oro, Trento, Italy
| | - Sara Monteverdi
- Medical Oncology, Breast Unit Santa Chiara Hospital, APSS Trento, Largo Medaglie D'Oro, Trento, Italy
| | - Palma Fedele
- Oncology Unit, Dario Camberlingo Hospital, ASL Brindisi, Francavilla Fontana, Italy
| | - Yari Ciribilli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Povo, Italy.
| |
Collapse
|
40
|
Li X, Huang Y, Zheng K, Yu G, Wang Q, Gu L, Li J, Wang H, Zhang W, Sun Y, Li C. Integrated proteomic and phosphoproteomic data-independent acquisition data evaluate the personalized drug responses of primary and metastatic tumors in colorectal cancer. BIOPHYSICS REPORTS 2023; 9:67-81. [PMID: 37753059 PMCID: PMC10518519 DOI: 10.52601/bpr.2022.210048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 11/18/2022] [Indexed: 02/19/2023] Open
Abstract
Mass spectrometry (MS)-based proteomics and phosphoproteomics are powerful methods to study the biological mechanisms, diagnostic biomarkers, prognostic analysis, and drug therapy of tumors. Data-independent acquisition (DIA) mode is considered to perform better than data-dependent acquisition (DDA) mode in terms of quantitative reproducibility, specificity, accuracy, and identification of low-abundance proteins. Mini patient derived xenograft (MiniPDX) model is an effective model to assess the response to antineoplastic drugs in vivo and is helpful for the precise treatment of cancer patients. Kinases are favorable spots for tumor-targeted drugs, and their functional completion relies on signaling pathways through phosphorylating downstream substrates. Kinase-phosphorylation networks or edge interactions are considered more credible and permanent for characterizing complex diseases. Here, we provide a workflow for personalized drug response assessment in primary and metastatic colorectal cancer (CRC) tumors using DIA proteomic data, DIA phosphoproteomic data, and MiniPDX models. Three kinase inhibitors, afatinib, gefitinib, and regorafenib, are tested pharmacologically. The process mainly includes the following steps: clinical tissue collection, sample preparation, hybrid spectral libraries establishment, MS data acquisition, kinase-substrate network construction, in vivo drug test, and elastic regression modeling. Our protocol gives a more direct data basis for individual drug responses, and will improve the selection of treatment strategies for patients without the druggable mutation.
Collapse
Affiliation(s)
- Xumiao Li
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yiming Huang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kuo Zheng
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Guanyu Yu
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Qinqin Wang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Gu
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jingquan Li
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hui Wang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei Zhang
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yidi Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chen Li
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
41
|
Corti C, De Angelis C, Bianchini G, Malorni L, Giuliano M, Hamilton E, Jeselsohn R, Jhaveri K, Curigliano G, Criscitiello C. Novel endocrine therapies: What is next in estrogen receptor positive, HER2 negative breast cancer? Cancer Treat Rev 2023; 117:102569. [PMID: 37146385 DOI: 10.1016/j.ctrv.2023.102569] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 05/07/2023]
Abstract
Endocrine therapy (ET) is the cornerstone of management in hormone receptor (HR)+ breast cancer (BC). Indeed, targeting the estrogen receptor (ER) signaling at different levels is a successful strategy, since BC largely relies on the ER signaling as a driver of tumorigenesis and progression. In metastatic BC, progression of disease typically occurs due to either ligand-independent ER signaling, which favors tumor proliferation and survival in the absence of hormonal stimuli, or an ER-independent signaling, which exploits alternative transcription pathways. For instance, estrogen receptor 1 (ESR1) mutations induce constitutive ER activity, in turn upregulating ER-dependent gene transcription and causing resistance to estrogen depleting therapies. The largest unmet need lies after progression on ET + cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors, where fulvestrant alone provides an average 2-3-month PFS. In this context, novel oral selective estrogen receptor degraders (SERDs) and other next-generation ETs are being investigated, both as single agents and in combination with targeted therapies. Elacestrant, the next generation ET in most advanced clinical development and the first to be FDA approved, demonstrated improved outcomes compared to standard ETs in ET pre-treated HR+/HER2- metastatic BC in the phase 3 EMERALD clinical trial. Additionally, other agents are showing promising results in both preclinical and early phase clinical settings. In this review, emerging data related to oral SERDs and other novel ETs in managing HR+/HER2- BC are presented. Major challenges and future perspectives related to the optimal sequence of therapeutic options and the molecular landscape of endocrine resistance are also provided.
Collapse
Affiliation(s)
- Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato - Oncology (DIPO), University of Milan, Milan, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy; Laster and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Giampaolo Bianchini
- Department of Medical Oncology, Ospedale San Raffaele, IRCCS, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Luca Malorni
- "Sandro Pitigliani" Department of Medical Oncology, Hospital of Prato, Azienda USL Toscana Centro, Prato, Italy; "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, Azienda USL Toscana Centro, Prato, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy; Laster and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Erika Hamilton
- Sarah Cannon Research Institute, Nashville, TN, USA; Tennessee Oncology, PLLC, Nashville, TN, USA
| | - Rinath Jeselsohn
- Breast Oncology Center, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA; Division of Molecular and Cellular Oncology, Dana Farber Cancer Institute, Boston, MA, USA; Center for Functional Cancer Epigenetics, Dana Farber Cancer Institute, Boston, MA, USA
| | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato - Oncology (DIPO), University of Milan, Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato - Oncology (DIPO), University of Milan, Milan, Italy.
| |
Collapse
|
42
|
Patel R, Klein P, Tiersten A, Sparano JA. An emerging generation of endocrine therapies in breast cancer: a clinical perspective. NPJ Breast Cancer 2023; 9:20. [PMID: 37019913 PMCID: PMC10076370 DOI: 10.1038/s41523-023-00523-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/10/2023] [Indexed: 04/07/2023] Open
Abstract
Anti-estrogen therapy is a key component of the treatment of both early and advanced-stage hormone receptor (HR)-positive breast cancer. This review discusses the recent emergence of several anti-estrogen therapies, some of which were designed to overcome common mechanisms of endocrine resistance. The new generation of drugs includes selective estrogen receptor modulators (SERMs), orally administered selective estrogen receptor degraders (SERDs), as well as more unique agents such as complete estrogen receptor antagonists (CERANs), proteolysis targeting chimeric (PROTACs), and selective estrogen receptor covalent antagonists (SERCAs). These drugs are at various stages of development and are being evaluated in both early and metastatic settings. We discuss the efficacy, toxicity profile, and completed and ongoing clinical trials for each drug and highlight key differences in their activity and study population that have ultimately influenced their advancement.
Collapse
Affiliation(s)
- Rima Patel
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA.
| | - Paula Klein
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Amy Tiersten
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Joseph A Sparano
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| |
Collapse
|
43
|
Abstract
Elacestrant (ORSERDU™) is an orally available selective estrogen receptor degrader (SERD) being developed by Stemline Therapeutics, a subsidiary of Menarini Group, for the treatment of estrogen receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer. In January 2023, elacestrant received its first approval for the treatment of postmenopausal women or adult men with ER-positive, HER2-negative, estrogen receptor 1 (ESR1)-mutated (as determined by a US FDA-approved test) advanced or metastatic breast cancer with disease progression following ≥ 1 line of endocrine therapy in the USA. A regulatory assessment of elacestrant for the treatment of ER-positive, HER2-negative advanced or metastatic breast cancer is currently underway in the EU. Development of elacestrant for the treatment of vasomotor symptoms has been discontinued. This article summarizes the milestones in the development of elacestrant leading to this first approval for this indication.
Collapse
Affiliation(s)
- Sheridan M Hoy
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand.
| |
Collapse
|
44
|
El-Tanani M, Al Khatib AO, Al-Najjar BO, Shakya AK, El-Tanani Y, Lee YF, Serrano-Aroca Á, Mishra V, Mishra Y, Aljabali AA, Goyal R, Negi P, Farani MR, Binabaj MM, Gholami A, Binabaj MM, Charbe NB, Tambuwala MM. Cellular and molecular basis of therapeutic approaches to breast cancer. Cell Signal 2023; 101:110492. [PMID: 36241056 DOI: 10.1016/j.cellsig.2022.110492] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022]
Abstract
In recent decades, there has been a significant amount of research into breast cancer, with some important breakthroughs in the treatment of both primary and metastatic breast cancers. It's a well-known fact that treating breast cancer is still a challenging endeavour even though physicians have a fantastic toolset of the latest treatment options at their disposal. Due to limitations of current clinical treatment options, traditional chemotherapeutic drugs, and surgical options are still required to address this condition. In recent years, there have been several developments resulting in a wide range of treatment options. This review article discusses the cellular and molecular foundation of chemotherapeutic drugs, endocrine system-based treatments, biological therapies, gene therapy, and innovative techniques for treating breast cancer.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan; Centre for Cancer Research and Cell Biology, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, Northern Ireland, UK; Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK.
| | - Arwa Omar Al Khatib
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Belal O Al-Najjar
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Ashok K Shakya
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Yahia El-Tanani
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, Northern Ireland, UK; Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Yin-Fai Lee
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; School of Life Sciences, Faculty of Science and Engineering, Anglia Ruskin University, Cambridge CB1 1PT, UK; Neuroscience, Psychology & Behaviour, College of Life Sciences, University of Leicester, Leicester LE1 9HN, UK
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Yachana Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 566, Jordan
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173229, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173229, India
| | - Marzieh Ramezani Farani
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), the Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), 1417614411 Tehran, Iran.
| | - Maryam Moradi Binabaj
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Amir Gholami
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Maryam Moradi Binabaj
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Nitin B Charbe
- Center for pharmacometrics and system pharmacology, department of pharmaceutics, college of pharmacy, University of Florida, FL, USA
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; Neuroscience, Psychology & Behaviour, College of Life Sciences, University of Leicester, Leicester LE1 9HN, UK.
| |
Collapse
|
45
|
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
|
46
|
Hancock GR, Young KS, Hosfield DJ, Joiner C, Sullivan EA, Yildiz Y, Lainé M, Greene GL, Fanning SW. Unconventional isoquinoline-based SERMs elicit fulvestrant-like transcriptional programs in ER+ breast cancer cells. NPJ Breast Cancer 2022; 8:130. [PMID: 36517522 PMCID: PMC9748900 DOI: 10.1038/s41523-022-00497-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022] Open
Abstract
Estrogen receptor alpha (ERα) is a ligand-dependent master transcriptional regulator and key driver of breast cancer pathology. Small molecule hormones and competitive antagonists favor unique ERα conformational ensembles that elicit ligand-specific transcriptional programs in breast cancer and other hormone-responsive tissues. By affecting disparate ligand binding domain structural features, unconventional ligand scaffolds can redirect ERα genomic binding patterns to engage novel therapeutic transcriptional programs. To improve our understanding of these ERα structure-transcriptional relationships, we develop a series of chemically unconventional antagonists based on the antiestrogens elacestrant and lasofoxifene. High-resolution x-ray co-crystal structures show that these molecules affect both classical and unique structural motifs within the ERα ligand binding pocket. They show moderately reduced antagonistic potencies on ERα genomic activities but are effective anti-proliferative agents in luminal breast cancer cells. Interestingly, they favor a 4-hydroxytamoxifen-like accumulation of ERα in breast cancer cells but lack uterotrophic activities in an endometrial cell line. Importantly, RNA sequencing shows that the lead molecules engage transcriptional pathways similar to the selective estrogen receptor degrader fulvestrant. This advance shows that fulvestrant-like genomic activities can be achieved without affecting ERα accumulation in breast cancer cells.
Collapse
Affiliation(s)
- G R Hancock
- Department of Cancer Biology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - K S Young
- Department of Cancer Biology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - D J Hosfield
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, 60637, USA
| | - C Joiner
- Department of Cancer Biology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - E A Sullivan
- Department of Cancer Biology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Y Yildiz
- Department of Cancer Biology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - M Lainé
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, 60637, USA
| | - G L Greene
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, 60637, USA
| | - S W Fanning
- Department of Cancer Biology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA.
| |
Collapse
|
47
|
Wang Y, Tang SC. The race to develop oral SERDs and other novel estrogen receptor inhibitors: recent clinical trial results and impact on treatment options. Cancer Metastasis Rev 2022; 41:975-990. [PMID: 36229710 PMCID: PMC9560885 DOI: 10.1007/s10555-022-10066-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/29/2022] [Indexed: 01/25/2023]
Abstract
Hormonal therapy plays a vital part in the treatment of estrogen receptor-positive (ER +) breast cancer. ER can be activated in a ligand-dependent and independent manner. Currently available ER-targeting agents include selective estrogen receptor modulators (SERMs), selective estrogen receptor degraders (SERDs), and aromatase inhibitors (AIs). Estrogen receptor mutation (ESR1 mutation) is one of the common mechanisms by which breast cancer becomes resistant to additional therapies from SERMs or AIs. These tumors remain sensitive to SERDs such as fulvestrant. Fulvestrant is limited in clinical utilization by its intramuscular formulation and once-monthly injection in large volumes. Oral SERDs are being rapidly developed to replace fulvestrant with the potential of higher efficacy and lower toxicities. Elacestrant is the first oral SERD that went through a randomized phase III trial showing increased efficacy, especially in tumors bearing ESR1 mutation, and good tolerability. Two other oral SERDs recently failed to achieve the primary endpoints of longer progression-free survival (PFS). They targeted tumors previously treated with several lines of prior therapies untested for ESR1 mutation. Initial clinical trial data demonstrated that tumors without the ESR1 mutation are less likely to benefit from the SERDs and may still respond to SERMs or AIs, including tumors previously exposed to hormonal therapy. Testing for ESR1 mutation in ongoing clinical trials and in hormonal therapy for breast cancer is highly recommended. Novel protein degradation technologies such as proteolysis-targeting chimera (PROTACS), molecular glue degrader (MGD), and lysosome-targeting chimeras (LYTACS) may result in more efficient ER degradation, while ribonuclease-targeting chimeras (RIBOTAC) and small interfering RNA (siRNA) may inhibit the production of ER protein.
Collapse
Affiliation(s)
- Yating Wang
- Ascension Providence Hospital, Southfield, MI, USA
| | - Shou-Ching Tang
- Cancer Center and Research Institute, University of Mississippi Medical Center, Guyton Research Building, G-651-07, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
48
|
Pagliuca M, Donato M, D’Amato AL, Rosanova M, Russo AOM, Scafetta R, De Angelis C, Trivedi MV, André F, Arpino G, Del Mastro L, De Laurentiis M, Puglisi F, Giuliano M. New steps on an old path: Novel estrogen receptor inhibitors in breast cancer. Crit Rev Oncol Hematol 2022; 180:103861. [DOI: 10.1016/j.critrevonc.2022.103861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
|
49
|
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
|
50
|
Donahue K, Xie H, Li M, Gao A, Ma M, Wang Y, Tipton R, Semanik N, Primeau T, Li S, Li L, Tang W, Xu W. Diptoindonesin G is a middle domain HSP90 modulator for cancer treatment. J Biol Chem 2022; 298:102700. [PMID: 36395883 PMCID: PMC9771721 DOI: 10.1016/j.jbc.2022.102700] [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: 07/11/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022] Open
Abstract
HSP90 inhibitors can target many oncoproteins simultaneously, but none have made it through clinical trials due to dose-limiting toxicity and induction of heat shock response, leading to clinical resistance. We identified diptoindonesin G (dip G) as an HSP90 modulator that can promote degradation of HSP90 clients by binding to the middle domain of HSP90 (Kd = 0.13 ± 0.02 μM) without inducing heat shock response. This is likely because dip G does not interfere with the HSP90-HSF1 interaction like N-terminal inhibitors, maintaining HSF1 in a transcriptionally silent state. We found that binding of dip G to HSP90 promotes degradation of HSP90 client protein estrogen receptor α (ER), a major oncogenic driver protein in most breast cancers. Mutations in the ER ligand-binding domain (LBD) are an established mechanism of endocrine resistance and decrease the binding affinity of mainstay endocrine therapies targeting ER, reducing their ability to promote ER degradation or transcriptionally silence ER. Because dip G binds to HSP90 and does not bind to the LBD of ER, unlike endocrine therapies, it is insensitive to ER LBD mutations that drive endocrine resistance. Additionally, we determined that dip G promoted degradation of WT and mutant ER with similar efficacy, downregulated ER- and mutant ER-regulated gene expression, and inhibited WT and mutant cell proliferation. Our data suggest that dip G is not only a molecular probe to study HSP90 biology and the HSP90 conformation cycle, but also a new therapeutic avenue for various cancers, particularly endocrine-resistant breast cancer harboring ER LBD mutations.
Collapse
Affiliation(s)
- Kristine Donahue
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Haibo Xie
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Miyang Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ang Gao
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Min Ma
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yidan Wang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Rose Tipton
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Nicole Semanik
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Tina Primeau
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Shunqiang Li
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Weiping Tang
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA,For correspondence: Wei Xu; Weiping Tang
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA,For correspondence: Wei Xu; Weiping Tang
| |
Collapse
|