1
|
Mahtani R, Collin SM, Tan Z, Shah CH, Adeyemi B, Davies S, John E, Vidal G. Human Epidermal Growth Factor Receptor 2-Positive (HER2+) Early Breast Cancer Treatment and Outcomes by Risk of Recurrence: A Retrospective US Electronic Health Records Study. Cancers (Basel) 2025; 17:1848. [PMID: 40507330 PMCID: PMC12153779 DOI: 10.3390/cancers17111848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/15/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
Background/Objectives: This study analyzed real-world data to assess patient characteristics, treatment patterns, and clinical outcomes in patients with human epidermal growth factor receptor 2-positive (HER2+) early breast cancer (eBC), as data for this patient group are limited. Methods: This was a retrospective observational cohort study of patients in a US electronic health record-derived deidentified database. Patient characteristics, treatment patterns, and clinical outcomes were analyzed overall and by risk of recurrence (high-risk versus non-high-risk). Results: Of 1290 patients, 366 (28.4%) were classified as high-risk and 924 (71.6%) as non-high-risk. At 5 years, high-risk versus non-high-risk eBC had a lower probability of invasive disease-free survival (72.3% [95% CI: 66.8-77.1%] vs. 80.7% [95% CI: 77.6-83.5%]), distant recurrence-free survival (78.7% [95% CI: 74.1-83.6%] vs. 89.3% [95% CI: 86.9-91.7%]), and overall survival (86.9% [95% CI: 82.3-90.4%] vs. 91.8% [95% CI: 89.4-93.7%]), and a shorter time to mBC diagnosis (22.6 vs. 34.1 months, respectively). Neoadjuvant therapy use increased from 18.2% in 2011-2013 to 67.3% in 2018-2021 in high-risk eBC and from 4.1% to 34.4% in non-high-risk eBC. However, 32.7% of eligible patients with high-risk eBC did not receive neoadjuvant therapy in 2018-2021. Use of post-neoadjuvant and adjuvant therapy increased in high-risk (but not non-high-risk) eBC from 81.8% in 2011-2013 to 91.8% in 2018-2021. Conclusions: High-risk HER2+ eBC manifested more aggressively than non-high-risk eBC. Utilization of neoadjuvant and (post-neo)adjuvant therapy increased over time. However, despite guideline recommendations, uptake of neoadjuvant therapy remains suboptimal.
Collapse
Affiliation(s)
- Reshma Mahtani
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA;
| | - Simon M. Collin
- Oncology Outcomes Research (O2R), Evidence Generation, Publications, and Partnerships, Global Medical Affairs, Oncology Business Unit, AstraZeneca, Cambridge CB2 0AA, UK;
| | - Ziyu Tan
- Oncology Outcomes Research (O2R), Evidence Generation, Publications, and Partnerships, Global Medical Affairs, Oncology Business Unit, AstraZeneca, Cambridge CB2 0AA, UK;
| | - Chintal H. Shah
- Oncology Outcomes Research (O2R), Evidence Generation, Publications, and Partnerships, Global Medical Affairs, Oncology Business Unit, AstraZeneca, Gaithersburg, MD 20878, USA;
| | - Basirat Adeyemi
- US Medical Affairs, Oncology Business Unit, AstraZeneca, Gaithersburg, MD 20878, USA;
| | - Sophie Davies
- Global Medical Affairs, Oncology Business Unit, AstraZeneca, Cambridge CB2 0AA, UK;
| | - Ellie John
- Global Medical Affairs Payer Biometrics, Statistics, Oncology Business Unit, AstraZeneca, Cambridge CB2 0AA, UK;
| | - Gregory Vidal
- West Cancer Center Research Institute and Lee S. Schwartzberg Research Center, Germantown, TN 38138, USA;
| |
Collapse
|
2
|
Cherny NI, Oosting SF, Dafni U, Latino NJ, Galotti M, Zygoura P, Dimopoulou G, Amaral T, Barriuso J, Calles A, Kiesewetter B, Gomez-Roca C, Gyawali B, Piccart M, Passaro A, Roitberg F, Tarazona N, Trapani D, Curigliano G, Wester R, Zarkavelis G, Zielinski C, de Vries EGE. ESMO-Magnitude of Clinical Benefit Scale version 2.0 (ESMO-MCBS v2.0). Ann Oncol 2025:S0923-7534(25)00166-8. [PMID: 40409995 DOI: 10.1016/j.annonc.2025.04.006] [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: 12/21/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND The ESMO-Magnitude of Clinical Benefit Scale (ESMO-MCBS) is a validated tool to assess the magnitude of clinical benefit from new cancer therapies, with planned updates based upon recognition of new needs and shortcomings. This paper describes the development of ESMO-MCBS v2.0. METHODOLOGY The revision process incorporates nine steps: (i) review of critiques and suggestions and identification of problems in the application of ESMO-MCBS v1.1; (ii) identification of shortcomings for revision in the upcoming version; (iii) drafting solutions addressing identified shortcomings; (iv) field-testing of solutions; (v) preparation of a near-final revised version for peer review for reasonableness by members of the ESMO Faculty and ESMO Guidelines Committee; (vi) amendments based on peer review for reasonableness; (vii) near-final review by members of the ESMO-MCBS Working Group; (viii) final amendments; (ix) final review and approval by members of the ESMO-MCBS Working Group and the ESMO Executive Board. RESULTS Seventeen issues for revision or amendment were considered, and 13 amendments were formulated to address identified shortcomings. In the curative setting, studies evaluated based on disease-free survival now credit improved time without treatment or disease even when overall survival is not significantly improved, and studies with small absolute gain in disease-free survival are credited more conservatively. Additionally, acute and persistent toxicity annotations are added. In the non-curative setting, the approach to crediting a difference in the tail of overall survival and progression-free survival curves is more statistically valid, and the toxicity evaluation has been revised. In peer review all amendments were found to be either reasonable or mostly reasonable. The amendments changed the scoring of 85/353 of evaluated studies. CONCLUSIONS The amendments incorporated into ESMO-MCBS v2.0 change the scores of 13.6% of evaluated studies (10.5% downgraded, 3.1% upgraded) and add toxicity annotations to 45.5% of the studies in the curative setting, and improve its discriminatory capacity and utility.
Collapse
Affiliation(s)
- N I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - S F Oosting
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - U Dafni
- Laboratory of Biostatistics, Division of Public Health, Department of Nursing, University of Athens, Athens; Frontier Science Foundation-Hellas, Athens, Greece
| | - N J Latino
- ESMO Head Office, European Society for Medical Oncology, Lugano, Switzerland
| | - M Galotti
- ESMO Head Office, European Society for Medical Oncology, Lugano, Switzerland
| | - P Zygoura
- Frontier Science Foundation-Hellas, Athens, Greece; Athens University of Economics and Business, Athens, Greece
| | - G Dimopoulou
- Frontier Science Foundation-Hellas, Athens, Greece
| | - T Amaral
- Skin Cancer Center, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - J Barriuso
- The Christie NHS Foundation Trust and Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A Calles
- Medical Oncology Department, Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | - B Kiesewetter
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - C Gomez-Roca
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - B Gyawali
- Department of Oncology, Queen's University, Kingston, Canada
| | - M Piccart
- Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - A Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - F Roitberg
- Rede Ebserh (Empresa Brasileira de Serviços Hospitalares), Brasilia, Brazil
| | - N Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - D Trapani
- European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - G Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - R Wester
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - G Zarkavelis
- University of Ioannina, Department of Medical Oncology, Ioannina, Greece
| | - C Zielinski
- Wiener Privat Klinik, Central European Academy Cancer Center, Vienna, Austria
| | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
3
|
Zheng F, Du F, Yang Z, Wang X, Yue J, Ling Y, Yuan P. Potential prognostic value of HER2/CEP17 FISH ratio in HER2-positive non-metastatic breast cancer: a real-world study. Discov Oncol 2025; 16:800. [PMID: 40382749 PMCID: PMC12086127 DOI: 10.1007/s12672-025-02495-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 04/24/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND HER2-positive breast cancer (BC) requires anti-HER2 therapy. We aimed to determine whether the expression of the HER2/centromeric probe for chromosome 17 (CEP17) ratio was associated with prognosis in patients with HER2-positive non-metastatic BC. METHODS 267 HER2-positive BC were enrolled between January 2010 and December 2011. Stabilized inverse probability treatment weighting (sIPTW) was used to balance baseline characteristics. Real-world disease-free survival (DFS) and overall survival (OS) was analyzed. RESULTS The median follow-up time was 10.3 years (interquartile range: 9.4-10.8 years). HER2/CEP17 ratio of > 7.0 was defined as the HER2 ultra-positive group; a HER2/CEP17 ratio of ≤ 7.0 was defined as the HER2 normal-positive group. After sIPTW adjustment, no differences were observed in DFS and OS when anti-HER2 therapy was unknown, and similarly in the patients who were recorded as not receive trastuzumab (all p > 0.05). Interestingly, HER2 ultra-positive group had a worse DFS than the normal-positive group (hazard ratio [HR] = 2.72, p = 0.02), but there was no difference in OS (p = 0.30) in patients did receive trastuzumab. The multivariate Cox models also showed that the HER2 ultra-positive had worse DFS than HER2 normal-positive patients (HR = 3.71; p < 0.01). CONCLUSION For non-metastatic HER2-positive BC with or without trastuzumab treatment, the HER2/CEP17 ratio did not predict DFS and OS. However, our study supported that HER2 ultra-positive group had a worse DFS than the normal-positive group among non-metastatic HER2-positive BC patients receiving trastuzumab; therefore, this could be a potential predictor of DFS in these patients.
Collapse
Affiliation(s)
- Fangchao Zheng
- Department of VIP Medical Services, Department of Medical Oncology, National Cancer Centre/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, 100021, China
- Department of Medical Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Feng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), The VIPII Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital and Institute, Beijing, 100021, China
| | - Zixuan Yang
- Department of VIP Medical Services, Department of Medical Oncology, National Cancer Centre/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, 100021, China
| | - Xue Wang
- Department of VIP Medical Services, Department of Medical Oncology, National Cancer Centre/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, 100021, China
| | - Jian Yue
- Department of VIP Medical Services, Department of Medical Oncology, National Cancer Centre/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, 100021, China
| | - Yun Ling
- Department of Pathology, National Cancer Centre/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Yuan
- Department of VIP Medical Services, Department of Medical Oncology, National Cancer Centre/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, 100021, China.
| |
Collapse
|
4
|
Miski H, Krupa K, Budzik MP, Deptała A, Badowska-Kozakiewicz A. HER2-Positive Breast Cancer-Current Treatment Management and New Therapeutic Methods for Brain Metastasis. Biomedicines 2025; 13:1153. [PMID: 40426980 PMCID: PMC12109299 DOI: 10.3390/biomedicines13051153] [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: 04/01/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Breast cancer can be classified based on the immunohistochemistry (IHC) phenotypes, defined by the presence or absence of the main IHC markers. IHC phenotyping is important as it determines the prognosis and guides treatment. For example, human epidermal growth factor receptor 2 (HER2) overexpression, which triggers cell growth and division, is observed in HER2-positive breast cancer. Methods: The standard treatment is based on trastuzumab plus pertuzumab in combination with taxane chemotherapy. The possibility of developing metastases depends on those phenotypes. Approximately 25-50% of patients with HER2-positive breast cancer experience brain metastases. This aspect is especially important, as 20% of those patients die as a result. Results: Through the years, many advanced therapies have been introduced to treat brain metastases, including whole brain radiotherapy, stereotactic radiosurgery, and a tyrosine kinase inhibitor (TKI), neratinib. Nonetheless, this still remains a therapeutic challenge. Conclusions: In this review, we focus on the treatment and efficiency of therapies targeting HER2-positive breast cancer, mainly concentrating on the current and newly developed treatment options for brain metastases, such as trastuzumab deruxtecan and tucatinib.
Collapse
Affiliation(s)
- Hanna Miski
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (H.M.); (K.K.)
| | - Kamila Krupa
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (H.M.); (K.K.)
| | - Michał Piotr Budzik
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (A.D.); (A.B.-K.)
| | - Andrzej Deptała
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (A.D.); (A.B.-K.)
| | - Anna Badowska-Kozakiewicz
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (A.D.); (A.B.-K.)
| |
Collapse
|
5
|
Qin H, Teng Y, Yu P, Ning Z, Liu J. Cardiovascular adverse events associated with EGFR and HER2 dual TKIs: a pharmacovigilance study based on the FAERS database. Expert Opin Drug Saf 2025:1-9. [PMID: 40195028 DOI: 10.1080/14740338.2025.2489529] [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: 12/28/2024] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND EGFR and HER2 dual TKIs have been approved for the treatment of advanced breast cancer in patients who are HER2-positive for second and/or third-line treatment. However, there is a lack of attention to the cardiovascular adverse events (AEs) caused by EGFR and HER2 dual TKIs. RESEARCH DESIGN AND METHODS We analyzed data spanning between March 2007 and June 2024 using the FAERS database and the reporting odds ratio, proportional reporting ratio, and Bayesian confidence propagation neural network to perform disproportionality analysis. RESULTS Compared with non-cardiovascular AEs, cardiovascular AEs were associated with more severe clinical outcomes, such as higher rates of hospitalization, life-threatening events, disability, and death. Our analysis revealed that lapatinib had a higher-than-expected reporting rate for three SMQs, including cardiac failure, embolic and thrombotic events, and cardiomyopathy. No significant cardiovascular signals were observed for neratinib. CONCLUSION Disproportionality analysis results revealed a positive signal for cardiac failure, embolic and thrombotic events, and cardiomyopathy of lapatinib, and no positive signal for neratinib. We should pay attention to high-risk signals in clinical practice and monitor them appropriately.
Collapse
Affiliation(s)
- Henan Qin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yibin Teng
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Peiyao Yu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhen Ning
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Ryspayeva D, Seyhan AA, MacDonald WJ, Purcell C, Roady TJ, Ghandali M, Verovkina N, El-Deiry WS, Taylor MS, Graff SL. Signaling pathway dysregulation in breast cancer. Oncotarget 2025; 16:168-201. [PMID: 40080721 PMCID: PMC11906143 DOI: 10.18632/oncotarget.28701] [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: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
This article provides a comprehensive analysis of the signaling pathways implicated in breast cancer (BC), the most prevalent malignancy among women and a leading cause of cancer-related mortality globally. Special emphasis is placed on the structural dynamics of protein complexes that are integral to the regulation of these signaling cascades. Dysregulation of cellular signaling is a fundamental aspect of BC pathophysiology, with both upstream and downstream signaling cascade activation contributing to cellular process aberrations that not only drive tumor growth, but also contribute to resistance against current treatments. The review explores alterations within these pathways across different BC subtypes and highlights potential therapeutic strategies targeting these pathways. Additionally, the influence of specific mutations on therapeutic decision-making is examined, underscoring their relevance to particular BC subtypes. The article also discusses both approved therapeutic modalities and ongoing clinical trials targeting disrupted signaling pathways. However, further investigation is necessary to fully elucidate the underlying mechanisms and optimize personalized treatment approaches.
Collapse
Affiliation(s)
- Dinara Ryspayeva
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Tyler J. Roady
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - Maryam Ghandali
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Nataliia Verovkina
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| | - Martin S. Taylor
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Brown Center on the Biology of Aging, Brown University, RI 02903, USA
| | - Stephanie L. Graff
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| |
Collapse
|
7
|
Xie J, Yang Z, Li Z, Zhang T, Chen H, Chen X, Dai Z, Chen T, Hou J. Triple-positive breast cancer: navigating heterogeneity and advancing multimodal therapies for improving patient outcomes. Cancer Cell Int 2025; 25:77. [PMID: 40045297 PMCID: PMC11881339 DOI: 10.1186/s12935-025-03680-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/07/2025] [Indexed: 03/09/2025] Open
Abstract
Triple-positive breast cancer (TPBC), a unique subtype of luminal breast cancer, is characterized by concurrent positivity for estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Owing to the crosstalk between the ER and HER2 signaling pathways, the standard of care and drug resistance of this particular subtype are difficult challenges. Recent research and clinical trials have indicated a shift in the treatment paradigm for TPBC from single-target therapies to multi-pathway, multitarget strategies aiming to comprehensively modulate intricate signaling networks, thereby overcoming resistance and enhancing therapeutic outcomes. Among multiple strategies, triple-drug therapy has emerged as a promising treatment modality, demonstrating potential efficacy in patients with TPBC. Moving forward, there is a critical need to perform in-depth analyses of specific mechanisms of cancer pathogenesis and metastasis, decipher the complex interactions between different genes or proteins, and identify concrete molecular targets, thus paving the way for the development of tailored therapeutic strategies to combat TPBC effectively.
Collapse
Affiliation(s)
- Jie Xie
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Zhihui Yang
- Zunyi Medical University, No.6 Xuefu West Road, Zunyi, 563006, Guizhou Province, China
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Zhuolin Li
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Tianyu Zhang
- Urology Department, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Huan Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Xueru Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Zehua Dai
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Tao Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Jing Hou
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China.
| |
Collapse
|
8
|
Li X, Zhang X, Yin S, Nie J. Challenges and prospects in HER2-positive breast cancer-targeted therapy. Crit Rev Oncol Hematol 2025; 207:104624. [PMID: 39826885 DOI: 10.1016/j.critrevonc.2025.104624] [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: 07/09/2024] [Revised: 12/29/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
Breast cancer remains the most prevalent malignancy among women globally and ranks as the leading cause of cancer-related mortality in this demographic. Approximately 13 %-15 % of all breast cancer cases are classified as HER2-positive, a subtype associated with a particularly unfavorable prognosis. A large number of patients with HER2-positive breast cancer continue to face disease progression after receiving standardized treatment. Given these challenges, a thorough exploration into the mechanisms underlying drug resistance in HER2-targeted therapy is imperative. This review focuses on the factors related to drug resistance in HER2-targeted therapy, including tumor heterogeneity, antibody-binding efficacy, variations in the tumor microenvironment, and abnormalities in signal activation and transmission. Additionally, corresponding strategies to counteract these resistance mechanisms are discussed, to advance therapeutic efficacy and clinical benefits in the management of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Xiyin Li
- Department of Breast Cancer, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, the Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China.
| | - Xueying Zhang
- Department of Breast Cancer, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, the Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China.
| | - Saige Yin
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650118, China.
| | - Jianyun Nie
- Department of Breast Cancer, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, the Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China.
| |
Collapse
|
9
|
Mahdi AF, Ashfield N, Crown J, Collins DM. Pre-Clinical Rationale for Amcenestrant Combinations in HER2+/ER+ Breast Cancer. Int J Mol Sci 2025; 26:460. [PMID: 39859174 PMCID: PMC11765389 DOI: 10.3390/ijms26020460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/19/2024] [Accepted: 12/27/2024] [Indexed: 01/30/2025] Open
Abstract
HER2-positive/oestrogen receptor-positive (HER2+/ER+) represents a unique breast cancer subtype. The use of individual HER2- or ER-targeting agents can lead to the acquisition of therapeutic resistance due to compensatory receptor crosstalk. New drug combinations targeting HER2 and ER could improve outcomes for patients with HER2+/ER+ breast cancer. In this study, the pre-clinical rationale is explored for combining amcenestrant (Amc), a selective oestrogen receptor degrader (SERD), with HER2-targeted therapies including trastuzumab, trastuzumab-emtansine (T-DM1) and tyrosine kinase inhibitors (TKIs). The combination of Amc and anti-HER2 therapies was investigated in a panel of four HER2+/ER+ cell lines: BT-474, MDA-MB-361, EFM-192a and a trastuzumab-resistant variant BT-474-T. Proliferation (IC50 and matrix combination assays) was determined using acid phosphatase assays. HER2/ER and intracellular signalling pathway protein levels/activity were investigated by western blot. Apoptosis was assessed using caspase 3/7 assays. Additivity and synergy were observed between Amc and the TKIs neratinib, lapatinib and tucatinib in all cell lines. Amc increased the anti-proliferative effect of trastuzumab in MDA-MB-361 and BT-474-T. Addition of Amc also increased anti-proliferative efficacy of T-DM1 in BT-474-T. TKI/Amc combinations reduced p-HER2 and ER levels and resulted in increased apoptosis. Higher ER expression in MDA-MB-361 and BT-474-T was associated with greater potential for synergy. In conclusion, the combination of Amc- and HER2-targeted treatments has potential as a therapeutic strategy for the treatment of HER2+/ER+ breast cancer and warrants further clinical investigation to validate safety and efficacy in patients.
Collapse
Affiliation(s)
- Amira F. Mahdi
- Cancer Biotherapeutics Research Group, Life Sciences Institute, School of Biotechnology, Dublin City University, Dublin 9, D09 NR58 Dublin, Ireland; (N.A.); (J.C.)
- Limerick Digital Cancer Research Centre, Health Research Institute, School of Medicine, University of Limerick, V94 T9PX Limerick, Ireland
| | - Niall Ashfield
- Cancer Biotherapeutics Research Group, Life Sciences Institute, School of Biotechnology, Dublin City University, Dublin 9, D09 NR58 Dublin, Ireland; (N.A.); (J.C.)
| | - John Crown
- Cancer Biotherapeutics Research Group, Life Sciences Institute, School of Biotechnology, Dublin City University, Dublin 9, D09 NR58 Dublin, Ireland; (N.A.); (J.C.)
- Department of Medical Oncology, St. Vincent’s University Hospital, Dublin 4, D04 T6F4 Dublin, Ireland
| | - Denis M. Collins
- Cancer Biotherapeutics Research Group, Life Sciences Institute, School of Biotechnology, Dublin City University, Dublin 9, D09 NR58 Dublin, Ireland; (N.A.); (J.C.)
| |
Collapse
|
10
|
Kidane RD, Ruddy KJ, Lin G, Sandhu NP. Cardiovascular Health Considerations for Primary Care Physicians Treating Breast Cancer Survivors. Mayo Clin Proc 2025; 100:124-140. [PMID: 39641716 DOI: 10.1016/j.mayocp.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/09/2024] [Accepted: 08/13/2024] [Indexed: 12/07/2024]
Abstract
Breast cancer (BC) survivors are at increased risk for cardiovascular disease (CVD) and require their primary care physicians to manage their long-term general medical care, including cardiovascular (CV) health. Yet, evidence exists that some primary care physicians possess insufficient knowledge about survivorship care. With the goal of bridging these knowledge gaps, a PubMed review was conducted from July 7, 2020, through October 2, 2020, with an updated PubMed review from January 3, 2024, through April 28, 2024, focusing on CV health considerations in the primary care of BC survivors. Search terms included variations of "breast cancer survivors" and "cardiovascular." In total, 152 publications were included. Breasts cancer survivors may have increased CVD risk because some anticancer therapies are cardiotoxic and risk factors for BC often also increase the risk for CVD. Multiple risk factors overlap for BC and CVD such as older age, Western diet, early menarche, physical inactivity, high body mass index, and smoking. In this review, results are summarized from studies that report the presence of CV risk factors and CVD in BC survivors. Also described are the CV effects of BC therapies (chemotherapy, hormonal agents, targeted therapies, and radiotherapy) and the type of CV evaluation (cardiac imaging and measurement of biomarkers) that these patients may need. Primary care physicians have an important role in managing the CV health of BC survivors from preventing, assessing, and managing CV risk factors to referring patients to appropriate specialists when needed.
Collapse
Affiliation(s)
- Redet D Kidane
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Kathryn J Ruddy
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Grace Lin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nicole P Sandhu
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
11
|
Veeraraghavan J, De Angelis C, Gutierrez C, Liao FT, Sabotta C, Rimawi MF, Osborne CK, Schiff R. HER2-Positive Breast Cancer Treatment and Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:495-525. [PMID: 39821040 DOI: 10.1007/978-3-031-70875-6_24] [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
HER2-positive (+) breast cancer is an aggressive disease with poor prognosis, a narrative that changed drastically with the advent and approval of trastuzumab, the first humanized monoclonal antibody targeting HER2. In addition to another monoclonal antibody, more classes of HER2-targeted agents, including tyrosine kinase inhibitors, and antibody-drug conjugates were developed in the years that followed. While these potent therapies have substantially improved the outcome of patients with HER2+ breast cancer, resistance has prevailed as a clinical challenge ever since the arrival of targeted agents. Efforts to develop new treatment regimens to treat/overcome resistance is futile without a primary understanding of the mechanistic underpinnings of resistance. Resistance could be attributed to mechanisms that are either specific to the tumor epithelial cells or those that emerge through changes in the tumor microenvironment. Reactivation of the HER receptor layer due to incomplete blockade of the HER receptor layer or due to alterations in the HER receptors is one of the major mechanisms. In other instances, resistance may occur due to deregulations in key downstream signaling such as the PI3K/AKT or RAS/MEK/ERK pathways or due to the emergence of compensatory pathways such as ER, other RTKs, or metabolic pathways. Potent new targeted agents and approaches to target key actionable drivers of resistance have already been identified, many of which are in early clinical development or under preclinical evaluation. Ongoing and future translational research will continue to uncover additional therapeutic vulnerabilities, as well as new targeted agents and approaches to treat and/or overcome anti-HER2 treatment resistance.
Collapse
Affiliation(s)
- Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Carmine De Angelis
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Carolina Gutierrez
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Fu-Tien Liao
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Caroline Sabotta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Mothaffar F Rimawi
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - C Kent Osborne
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
12
|
Khalid T, Cutress RI, Remer M, Copson ER. Clinical Impact of Somatic Genomic Testing on Breast Cancer Care. Clin Oncol (R Coll Radiol) 2025; 37:103665. [PMID: 39541893 DOI: 10.1016/j.clon.2024.10.037] [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: 10/04/2023] [Revised: 08/12/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024]
Abstract
Developments in our understanding of the molecular biology of breast cancer have had a direct impact on the investigations needed to provide optimal breast cancer care. Somatic genomic tests are now used routinely to inform decisions regarding adjuvant chemotherapy use in selected early breast cancer patients, and to identify patients with advanced disease who can potentially benefit from novel targeted agents. In this overview, we describe the somatic genomic tests currently available within the National Health Service (NHS) for early and advanced breast cancer patients. We review the underlying biology and the evidence base for clinical utility of these tests in routine clinical practice. In addition, we identify the somatic genomic biomarkers currently in use in breast cancer clinical trials that are most likely to influence future breast cancer management. We also consider the challenges associated with tissue-based genomic testing in advanced breast cancer and the role of circulating tumour deoxyribonucleic acid (ctDNA) testing.
Collapse
Affiliation(s)
- T Khalid
- University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| | - R I Cutress
- University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK; Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - M Remer
- Basingstoke and North Hampshire Hospitals, Hampshire Hospitals NHS Foundation Trust, Aldermaston Road, Basingstoke RG24 9NA, UK
| | - E R Copson
- University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK; Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
13
|
Weller J, Potthoff A, Zeyen T, Schaub C, Duffy C, Schneider M, Herrlinger U. Current status of precision oncology in adult glioblastoma. Mol Oncol 2024; 18:2927-2950. [PMID: 38899374 PMCID: PMC11619805 DOI: 10.1002/1878-0261.13678] [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: 11/16/2023] [Revised: 04/05/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The concept of precision oncology, the application of targeted drugs based on comprehensive molecular profiling, has revolutionized treatment strategies in oncology. This review summarizes the current status of precision oncology in glioblastoma (GBM), the most common and aggressive primary brain tumor in adults with a median survival below 2 years. Targeted treatments without prior target verification have consistently failed. Patients with BRAF V600E-mutated GBM benefit from BRAF/MEK-inhibition, whereas targeting EGFR alterations was unsuccessful due to poor tumor penetration, tumor cell heterogeneity, and pathway redundancies. Systematic screening for actionable molecular alterations resulted in low rates (< 10%) of targeted treatments. Efficacy was observed in one-third and currently appears to be limited to BRAF-, VEGFR-, and mTOR-directed treatments. Advancing precision oncology for GBM requires consideration of pathways instead of single alterations, new trial concepts enabling rapid and adaptive drug evaluation, a focus on drugs with sufficient bioavailability in the CNS, and the extension of target discovery and validation to the tumor microenvironment, tumor cell networks, and their interaction with immune cells and neurons.
Collapse
Affiliation(s)
- Johannes Weller
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | | | - Thomas Zeyen
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | - Christina Schaub
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | - Cathrina Duffy
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | | | - Ulrich Herrlinger
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| |
Collapse
|
14
|
Nolan L, Davey MG, Calpin GG, Ryan ÉJ, Boland MR. Risk of locoregional recurrence after breast cancer surgery by molecular subtype-a systematic review and network meta-analysis. Ir J Med Sci 2024; 193:2965-2974. [PMID: 39331262 DOI: 10.1007/s11845-024-03809-z] [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: 08/02/2023] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND The prevention of locoregional recurrence (LRR) is crucial in breast cancer, as it translates directly into reduced breast cancer-related death. Breast cancer is subclassified into distinct intrinsic biological subtypes with varying clinical outcomes. AIMS To perform a systematic review and network meta-analysis (NMA) to determine the rate of LRR by breast cancer molecular subtype. METHODS A NMA was performed as per PRISMA-NMA guidelines. Molecular subtypes were classified by St Gallen expert consensus statement (2013). Analysis was performed using R and Shiny. RESULTS Five studies were included including 6731 patients whose molecular subtypes were available. Overall, 47.3% (3182/6731) were Luminal A (LABC: estrogen receptor (ER) + /human epidermal growth factor receptor-2 (HER2) - /progesterone receptor (PR) + or Ki-67 < 20%), 25.5% (1719/6731) were Luminal B (LBBC: ER + /HER2 - /PR - or Ki-67 ≥ 20%), 11.2% (753/6731) were Luminal B-HER2 + (LBBC-HER2: ER + /HER2 +), 6.9% (466/6731) were HER2 + (HER2 ER - /HER2 +), and finally 9.1% (611/6731) were triple-negative breast cancer (TNBC: ER - /HER2 -). The median follow-up was 74.0 months and the overall LRR rate was 4.0% (271/6731). The LRR was 1.7% for LABC (55/3182), 5.1% for LBBC (88/1719), 6.0% for LBBC-HER2 (45/753), 6.0% for HER2 (28/466), and 7.9% for TNBC (48/611). At NMA, patients with TNBC (odds ratio (OR) 3.73, 95% confidence interval (CI) 1.80-7.74), HER2 (OR 3.24, 95% CI 1.50-6.99), LBBC-HER2 (OR 2.38, 95% CI 1.09-5.20), and LBBC (OR 2.20, 95% CI 1.07-4.50) were significantly more likely to develop LRR compared to LABC. CONCLUSION TNBC and HER2 subtypes are associated with the highest risk of LRR. Multidisciplinary team discussions should consider these findings to optimize locoregional control following breast cancer surgery.
Collapse
Affiliation(s)
- Lily Nolan
- Discipline of Surgery, University of Galway, Newcastle Road, Galway, H91YR71, Ireland.
| | - Matthew G Davey
- Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Dublin, D02YN77, Ireland
| | - Gavin G Calpin
- Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Dublin, D02YN77, Ireland
| | - Éanna J Ryan
- Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Dublin, D02YN77, Ireland
| | - Michael R Boland
- Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Dublin, D02YN77, Ireland
| |
Collapse
|
15
|
Rediti M, Venet D, Joaquin Garcia A, Maetens M, Vincent D, Majjaj S, El-Abed S, Di Cosimo S, Ueno T, Izquierdo M, Piccart M, Pusztai L, Loi S, Salgado R, Viale G, Rothé F, Sotiriou C. Identification of HER2-positive breast cancer molecular subtypes with potential clinical implications in the ALTTO clinical trial. Nat Commun 2024; 15:10402. [PMID: 39613746 PMCID: PMC11607438 DOI: 10.1038/s41467-024-54621-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
In HER2-positive breast cancer, clinical outcome and sensitivity to HER2-targeted therapies are influenced by both tumor and microenvironment features. However, we are currently unable to depict the molecular heterogeneity of this disease with sufficient granularity. Here, by performing gene expression profiling in HER2-positive breast cancers from patients receiving adjuvant trastuzumab in the ALTTO clinical trial (NCT00490139), we identify and characterize five molecular subtypes associated with the risk of distant recurrence: immune-enriched, proliferative/metabolic-enriched, mesenchymal/stroma-enriched, luminal, and ERBB2-dependent. Additionally, we validate the biological profiles of the subtypes and explore their prognostic/predictive value in external cohorts, namely the NeoALTTO trial (NCT00553358), SCAN-B (NCT02306096), I-SPY2 (NCT01042379), METABRIC and TCGA. Immune-enriched tumors present better survival outcomes, in contrast to mesenchymal/stroma-enriched and proliferative/metabolic-enriched tumors, while luminal and ERBB2-dependent tumors are characterized by low and high rates of pathological complete response, respectively. Of note, these molecular subtypes provide the rationale for treatment approaches leveraging the heterogeneous biology of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Mattia Rediti
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - David Venet
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Andrea Joaquin Garcia
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marion Maetens
- Laboratory for Translational Breast Cancer Research, KU Leuven, Leuven, Belgium
| | - Delphine Vincent
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Samira Majjaj
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Serena Di Cosimo
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Takayuki Ueno
- Breast Surgical Oncology, Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | - Martine Piccart
- Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Lajos Pusztai
- Yale School of Medicine, Yale Cancer Center, New Haven, CT, USA
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Roberto Salgado
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
| | - Giuseppe Viale
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Françoise Rothé
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
16
|
Li YW, Dai LJ, Wu XR, Zhao S, Xu YZ, Jin X, Xiao Y, Wang Y, Lin CJ, Zhou YF, Fu T, Yang WT, Li M, Lv H, Chen S, Grigoriadis A, Jiang YZ, Ma D, Shao ZM. Molecular Characterization and Classification of HER2-Positive Breast Cancer Inform Tailored Therapeutic Strategies. Cancer Res 2024; 84:3669-3683. [PMID: 39186675 DOI: 10.1158/0008-5472.can-23-4066] [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: 12/21/2023] [Revised: 04/19/2024] [Accepted: 08/08/2024] [Indexed: 08/28/2024]
Abstract
HER2-positive breast cancer is an aggressive subtype that accounts for 15% to 20% of all breast cancers. Recent studies have suggested that HER2-positive breast cancer is a group of heterogeneous diseases with different sensitivities to standard treatment regimens. Revealing the molecular heterogeneity of HER2-positive breast cancer could potentially enable more precise treatment strategies. In this study, we performed multiomics profiling on a HER2-positive breast cancer cohort and identified four transcriptome-based subtypes. The classical HER2 (HER2-CLA) subtype comprised 28.3% of the samples and displayed high ERBB2 activation and significant benefit from anti-HER2 therapy. The immunomodulatory (HER2-IM) subtype (20%) featured an immune-activated microenvironment, potentially suitable for de-escalated treatment and immunotherapy. The luminal-like (HER2-LUM) subtype (30.6%) possessed similar molecular features of hormone receptor-positive HER2-negative breast cancer, suggesting endocrine therapy and CDK4/6 inhibitors as a potential therapeutic strategy. Lastly, the basal/mesenchymal-like (HER2-BM) subtype (21.1%) had a poor response to current dual HER2-targeted therapy and could potentially benefit from tyrosine kinase inhibitors. The molecular characteristics and clinical features of the subtypes were further explored across multiple cohorts, and the feasibility of the proposed treatment strategies was validated in patient-derived organoid and patient-derived tumor fragment models. This study elucidates the molecular heterogeneity of HER2-positive breast cancer and paves the way for a more tailored treatment. Significance: Illumination of the inherent heterogeneity within HER2-positive breast cancers through the delineation of distinct molecular subtypes lays the groundwork for developing more personalized treatment strategies based on specific patient characteristics.
Collapse
Affiliation(s)
- Yu-Wei Li
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei-Jie Dai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiang-Rong Wu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shen Zhao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Zheng Xu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi Jin
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Xiao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Wang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cai-Jin Lin
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Fan Zhou
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tong Fu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Tao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ming Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hong Lv
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Siyuan Chen
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Anita Grigoriadis
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding Ma
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Conlon NT, Roche S, Mahdi AF, Browne A, Breen L, Gaubatz J, Meiller J, O'Neill F, O'Driscoll L, Cremona M, Hennessy BT, Eli LD, Crown J, Collins DM. Neratinib plus dasatinib is highly synergistic in HER2-positive breast cancer in vitro and in vivo. Transl Oncol 2024; 49:102073. [PMID: 39191139 PMCID: PMC11396364 DOI: 10.1016/j.tranon.2024.102073] [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: 02/19/2024] [Revised: 07/05/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND HER2-targeted therapies have revolutionised the treatment of HER2-positive breast cancer. However, de novo resistance or the emergence of acquired resistance is a persistent clinical problem. Here we report that neratinib, an irreversible pan-HER inhibitor, in combination with the multi-kinase inhibitor dasatinib, currently used to treat certain leukemias, has strong anti-proliferative effects against models of HER2-positive breast cancer that are innately resistant to trastuzumab or have acquired resistance to neratinib. METHODS Neratinib plus dasatinib was examined in a panel of 20 breast cancer cell lines, including HER2-positive, estrogen-receptor-positive, triple negative, and acquired HER2-targeted therapy resistant models. Drug effects on migration and apoptosis induction was evaluated and signaling alterations were determined by reverse phase protein array (RPPA). In vivo efficacy was examined using orthotopically-implanted HCC1954 cells. RESULTS Synergy was observed in cell lines innately resistant to trastuzumab, models with acquired resistance to neratinib, and in triple negative breast cancer cell lines. Further investigation showed that neratinib plus dasatinib induced apoptosis and inhibited cell migration to a greater degree than either drug alone. RPPA revealed that the combination caused suppression of key survival signaling through EGFR, Akt, and MAPK inhibition. In vivo, neratinib plus dasatinib was well tolerated and had a prolonged anti-tumor effect against HCC1954 xenografts. CONCLUSIONS This study provides a strong pre-clinical rationale for the clinical investigation neratinib and dasatinib in HER2+ breast cancer.
Collapse
Affiliation(s)
- Neil T Conlon
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Sandra Roche
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Amira F Mahdi
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Alacoque Browne
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Laura Breen
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Johanna Gaubatz
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Justine Meiller
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Fiona O'Neill
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Science & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mattia Cremona
- Molecular Medicine - Laboratory of Molecular Oncology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Bryan T Hennessy
- Molecular Medicine - Laboratory of Molecular Oncology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Lisa D Eli
- Puma Biotechnology, Inc., 10880 Wilshire Boulevard, Suite 2150, Los Angeles, CA, 90024, USA
| | - John Crown
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland; Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Denis M Collins
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| |
Collapse
|
18
|
Ming C, Liu Z, Wang Q, Liu L, Shen X. Oral solid dosage form using alternate crystalline neratinib maleate anhydrous form: Pharmaceutical, bioequivalent, and clinical perspectives. Biomed Chromatogr 2024; 38:e5988. [PMID: 39164837 DOI: 10.1002/bmc.5988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/18/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024]
Abstract
Neratinib (an active pharmaceutical ingredient [API]) is an irreversible pan-human epidermal growth factor receptor (HER) inhibitor used to treat HER2-positive breast cancer. The dosage form (marketed in the United States, China, Europe, and other regions) is a tablet for oral administration, and the brand product (NERLYNX) has patent protection for the crystalline neratinib maleate monohydrate form. This paper describes the product development using stable crystalline neratinib maleate anhydrous form, stability study, bioequivalence (BE) study of the products, and discussion of the adverse effects observed in the clinical study.
Collapse
Affiliation(s)
| | - Zhi Liu
- Convalife Pharmaceuticals, Shanghai, China
| | - Qiong Wang
- Convalife Pharmaceuticals, Shanghai, China
| | - Ling Liu
- Convalife Pharmaceuticals, Shanghai, China
| | | |
Collapse
|
19
|
de Azambuja E, Piccart-Gebhart M, Fielding S, Townend J, Hillman DW, Colleoni M, Roylance R, Kelly CM, Lombard J, El-Abed S, Choudhury A, Korde L, Vicente M, Chumsri S, Rodeheffer R, Ellard SL, Wolff AC, Holtschmidt J, Lang I, Untch M, Boyle F, Xu B, Werutsky G, Tujakowski J, Huang CS, Baruch NB, Bliss J, Ferro A, Gralow J, Kim SB, Kroep JR, Krop I, Kuemmel S, McConnell R, Moscetti L, Knop AS, van Duijnhoven F, Gomez H, Cameron D, Di Cosimo S, Gelber RD, Moreno-Aspitia A. Final analysis of the ALTTO trial: adjuvant trastuzumab in sequence or in combination with lapatinib in patients with HER2-positive early breast cancer [BIG 2-06/NCCTG N063D (Alliance)]. ESMO Open 2024; 9:103938. [PMID: 39418883 PMCID: PMC11532431 DOI: 10.1016/j.esmoop.2024.103938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Dual anti-human epidermal growth factor receptor 2 (HER2) blockade has improved the outcomes of patients with early and metastatic HER2-positive breast cancer. Here we present the final 10-year analysis of the ALTTO trial. PATIENTS AND METHODS The ALTTO trial (NCT00490139) is a prospective randomized, phase III, open-label, multicenter study that investigated the role of adjuvant chemotherapy and trastuzumab alone, in combination or sequentially with lapatinib. The primary endpoint was disease-free survival (DFS) and secondary endpoints included overall survival (OS), time to distant recurrence and safety. RESULTS Overall, 6281 patients with HER2-positive early breast cancer were included in the final efficacy analysis in three treatment groups: trastuzumab (T), lapatinib + trastuzumab (L + T) and trastuzumab followed by lapatinib (T→L). Baseline characteristics were well balanced between groups. At a median follow-up of 9.8 years, the addition of lapatinib to trastuzumab and chemotherapy did not significantly improve DFS nor OS. The 10-year DFS was 77% in T, 79% in L + T and 79% in T→L, and the 10-year OS was 87%, 89% and 89%, respectively. The incidence of any cardiac event was low and similar in the three treatment groups. CONCLUSIONS With a longer follow-up, no significant improvement was observed in DFS in patients treated with dual anti-HER2 blockade with lapatinib + trastuzumab compared to trastuzumab alone. The 10-year survival rates for the combination group are consistent with other studies that have explored dual anti-HER2 therapy.
Collapse
Affiliation(s)
- E de Azambuja
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Institut Jules Bordet, Service d'oncologie médicale, Brussels.
| | - M Piccart-Gebhart
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Institut Jules Bordet, Brussels, Belgium
| | - S Fielding
- Frontier Science (Scotland) Ltd., Kincraig, UK
| | - J Townend
- Frontier Science (Scotland) Ltd., Kincraig, UK
| | - D W Hillman
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, USA
| | - M Colleoni
- Division of Medical Senology, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - R Roylance
- Department of Oncology, UCLH Foundation Trust, London, UK
| | - C M Kelly
- Cancer Trials Ireland, Dublin, Ireland; Mater Private Hospital, Dublin, Ireland
| | - J Lombard
- Breast Cancer Trials (Australia New Zealand), Newcastle, Australia
| | - S El-Abed
- Breast International Group (BIG), Brussels, Belgium
| | - A Choudhury
- Novartis Healthcare Pvt Ltd, Hyderabad, India
| | - L Korde
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, USA
| | - M Vicente
- Institut Jules Bordet, Brussels, Belgium
| | | | - R Rodeheffer
- Mayo Clinic College of Medicine and Science, Rochester, USA
| | | | - A C Wolff
- The Johns Hopkins Kimmel Cancer Center, Baltimore, USA
| | - J Holtschmidt
- German Breast Group (GBG) Forschungs GmbH, Neu-Isenburg, Germany
| | - I Lang
- Clinexpert Research, Gyongyos, Hungary
| | - M Untch
- Helios Klinikum Berlin-Buch, Head of Clinic for Gynecology, Gynecologic Oncology and Obstetrics, Breast Cancer Center, Berlin, Germany; AGO B Study Group, Study Center of the Women's Clinic, Erlangen University Hospital, Erlangen, Germany
| | - F Boyle
- Patricia Ritchie Centre for Cancer Care and Research, Mater Hospital, North Sydney, Australia; University of Sydney, Sydney, Australia
| | - B Xu
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - G Werutsky
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil
| | - J Tujakowski
- Chemotherapy, Centre of Oncology, Bydgoszcz, Poland
| | - C-S Huang
- National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | | - J Bliss
- The Institute of Cancer Research, London, UK
| | - A Ferro
- Breast Unit, Department of Medical Oncology, APSS, Trento, Italy
| | - J Gralow
- American Society of Clinical Oncology, Alexandria, USA
| | - S-B Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - J R Kroep
- Leiden University Medical Center, Leiden, The Netherlands
| | - I Krop
- Yale Cancer Center, New Haven, Connecticut, USA
| | - S Kuemmel
- Breast Unit, Kliniken Essen-Mitte, Essen; Clinic for Gynecology with Breast Center, Charité-Universitätsmedizin, Berlin, Germany
| | - R McConnell
- Frontier Science (Scotland) Ltd., Kincraig, UK
| | - L Moscetti
- Department of Oncology-Hematology, University Hospital of Modena, Modena; GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), Parma, Italy
| | - A S Knop
- Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - F van Duijnhoven
- Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - H Gomez
- Instituto Nacional de Enfermedades Neoplasicas, Lima, Peru
| | - D Cameron
- Edinburgh University Cancer Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - S Di Cosimo
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - R D Gelber
- Department of Data Science, Division of Biostatistics, Dana-Farber Cancer Institute, Harvard Medical School, Harvard T.H. Chan School of Public Health, Boston; Frontier Science Foundation, Boston, USA
| | | |
Collapse
|
20
|
Blanter J, Baldwin E, Patel R, Sheng T, Tiersten A. Patterns in use and tolerance of adjuvant neratinib in patients with hormone receptor (HR)-positive, HER2-positive early-stage breast cancer. Breast Cancer Res Treat 2024; 208:461-466. [PMID: 39153126 DOI: 10.1007/s10549-024-07461-0] [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: 07/24/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
PURPOSE One year of neratinib therapy is known to derive a significant invasive disease-free survival (iDFS) benefit in early-stage, hormone receptor-positive (HR +), HER2 + , node-positive breast cancer after trastuzumab-based adjuvant therapy. Limitations to neratinib use include significant gastrointestinal side effects, which often result in treatment discontinuation. In this study, we aimed to identify clinicopathologic features associated with adjuvant neratinib use and factors impacting treatment completion. METHODS We performed a retrospective review of patients with early-stage HR + HER2 + breast cancer who were prescribed neratinib from 2017 to 2023 at our institution. We used the electronic medical record to extract information on patient characteristics, clinical features, and treatment information. Patients were identified as high risk based on definitions adapted from the standard high-risk definition in HR + HER2- breast cancer combined with studies correlating high Ki67 or high tumor grade with lower recurrence-free survival. Statistical analysis was performed using two-sided T-tests and chi-square tests. RESULTS We identified 62 eligible patients of whom 55% completed 1 year of neratinib and 45% did not. Sixty percent (N = 37) of patients offered neratinib were considered high risk at diagnosis. The most common reason for neratinib discontinuation was inability to tolerate side effects (54%) followed by pill burden (18%). The most common side effect experienced by patients was diarrhea despite anti-diarrheal prophylaxis (56%), followed by rash (8%). Patients who received an up-titration of neratinib were more likely to complete the full course of neratinib when compared to those who did not (76% vs. 40.5% p = 0.013). The median starting dose of those who completed neratinib treatment was 140 vs. 240 mg in those who did not (p = 0.016). Neither group experienced a statistically significant greater likelihood of treatment holds or dose reductions. In terms of outcomes, 10 patients had progression of disease of whom 7 did not complete neratinib treatment (p = 0.169). Interestingly, those 7 patients developed metastatic disease and 57% (N = 4) had central nervous system metastases. CONCLUSION Patients are more likely to complete 1 year of adjuvant neratinib with dose up-titration. Dose reductions and interruptions did not affect neratinib adherence in our patient population. Seven patients (11%) in our study developed metastatic disease, all of whom did not complete adjuvant neratinib treatment.
Collapse
Affiliation(s)
- Julia Blanter
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA.
| | - Elena Baldwin
- Division of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rima Patel
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA
| | - Tianxiang Sheng
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA
| | - Amy Tiersten
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA
| |
Collapse
|
21
|
Manna M, Gelmon KA, Boileau JF, Brezden-Masley C, Cao JQ, Jerzak KJ, Prakash I, Sehdev S, Simmons C, Bouganim N, Brackstone M, Cescon DW, Chia S, Dayes IS, Edwards S, Hilton J, Joy AA, Laing K, Webster M, Henning JW. Guidance for Canadian Breast Cancer Practice: National Consensus Recommendations for the Systemic Treatment of Patients with HER2+ Breast Cancer in Both the Early and Metastatic Setting. Curr Oncol 2024; 31:6536-6567. [PMID: 39590115 PMCID: PMC11593131 DOI: 10.3390/curroncol31110484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024] Open
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancer is an aggressive subtype of breast cancer associated with a poor prognosis when sub-optimally treated. Recent advances include new and effective targeted therapies that have significantly improved outcomes for patients. Despite these advances, there are significant gaps across Canada, underscoring the need for evidence-based consensus guidance to inform treatment decisions. Addressing these gaps is crucial to ensuring that effective therapies are integrated into clinical practice, so as to improve the lives of patients affected by this aggressive form of breast cancer. The Research Excellence, Active Leadership (REAL) Canadian Breast Cancer Alliance is a standing nucleus committee of clinical-academic oncologists across Canada and Breast Cancer Canada, a patient organization. The mandate of this group is to provide evidence-based guidance on best practices in the management of patients with breast cancer. These consensus recommendations were developed using a modified Delphi process with up to three rounds of anonymous voting. Consensus was defined a priori as ≥75% of voters agreeing with the recommendation as written. There are 9 recommendations in the early setting; 7 recommendations in the metastatic setting; and 10 recommendations for patients with brain metastases.
Collapse
Affiliation(s)
- Mita Manna
- Saskatchewan Cancer Agency, Regina, SK S4W 0G3, Canada;
| | - Karen A. Gelmon
- BC Cancer—Vancouver, Vancouver, BC V5Z 4E6, Canada; (K.A.G.); (C.S.); (S.C.)
| | | | | | - Jeffrey Q. Cao
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (J.Q.C.); (M.W.)
| | | | - Ipshita Prakash
- Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (J.-F.B.); (I.P.)
| | - Sandeep Sehdev
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada; (S.S.); (J.H.)
| | - Christine Simmons
- BC Cancer—Vancouver, Vancouver, BC V5Z 4E6, Canada; (K.A.G.); (C.S.); (S.C.)
| | | | | | - David W. Cescon
- Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada;
| | - Stephen Chia
- BC Cancer—Vancouver, Vancouver, BC V5Z 4E6, Canada; (K.A.G.); (C.S.); (S.C.)
| | - Ian S. Dayes
- Juravinski Cancer Centre, Hamilton, ON L8V 5C2, Canada;
| | - Scott Edwards
- Dr. H. Bliss Murphy Cancer Center, St. John’s, NL A1B 3V6, Canada; (S.E.); (K.L.)
| | - John Hilton
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada; (S.S.); (J.H.)
| | | | - Kara Laing
- Dr. H. Bliss Murphy Cancer Center, St. John’s, NL A1B 3V6, Canada; (S.E.); (K.L.)
| | - Marc Webster
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (J.Q.C.); (M.W.)
| | | |
Collapse
|
22
|
Nieto-Coronel T, Alette OG, Yacab R, Fernández-Figueroa EA, Lopez-Camarillo C, Marchat L, Astudillo-de la Vega H, Ruiz-Garcia E. PI3K Mutation Profiles on Exons 9 (E545K and E542K) and 20 (H1047R) in Mexican Patients With HER-2 Overexpressed Breast Cancer and Its Relevance on Clinical-Pathological and Survival Biological Effects. Int J Breast Cancer 2024; 2024:9058033. [PMID: 39444377 PMCID: PMC11496583 DOI: 10.1155/2024/9058033] [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: 10/01/2023] [Revised: 06/30/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024] Open
Abstract
Background: Trastuzumab resistance is associated with overexpressing the human epidermal growth factor receptor 2 (HER-2), which results from the altered phosphoinositide 3-kinase (PI3K) pathway in breast cancer patients. Objective: We quantified the frequency of PI3K enzyme single and double-point mutations in Mexican patients with HER-2 overexpressing breast cancer and its association with clinical-pathological variables. Methods: We embedded HER-2 breast samples in paraffin from 60 patients, extracted their DNA, and evaluated PI3K mutations in 49 HER-2-positive breast tumors. We focused on mutations for one exon 20 (H1047R) and two exon 9 PI3K (E545K, E542K) hotspots and characterized them as single and double-point mutations. The mean patient follow-up was 86 months. Results: Of 49 patients who tested positive for HER-2 breast cancer, 14.28% showed mutations in PI3K, 71.42% single-point, and 28.56% double-point mutations. We found single-point mutations in H1047R (42.85%) and E545K (28.57%). Only two patients exhibited double-point mutations: one in E542K/E545K and another in H1047R/E545K (14.28% each). Although we observed lower survival in patients with mutations in PI3K, we did not find a significant association between these factors (p = 0.191). However, single and double-point mutations in PI3K were significantly associated with the clinical stages of diagnosis and tumor size (p = 0.027 and p = 0.04, respectively). Conclusion: Single and double-point mutations in PI3K are related to tumor size and advanced clinical-pathological traits in Mexican patients with HER-2 overexpression, and future molecular studies are necessary to understand these findings.
Collapse
Affiliation(s)
- T. Nieto-Coronel
- Medical Oncology Unit, MyA Medic–Oncopalia Center, La Paz, Bolivia
| | - Ortega-Gómez Alette
- Translational Medicine Laboratory, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - R. Yacab
- Translational Medicine Laboratory, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - E. A. Fernández-Figueroa
- Core B of Innovation in Precision Medicine, National Institute of Genomic Medicine, Mexico City, Mexico
| | - C. Lopez-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| | - L. Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Mexico City, Mexico
| | - H. Astudillo-de la Vega
- Translational Research Laboratory in Cancer & Cellular Therapy, Hospital de Oncologia, Siglo XXI, IMSS-Instituto Mexicano del Seguridad Social, Mexico City, Mexico
| | - E. Ruiz-Garcia
- Translational Medicine Laboratory, Instituto Nacional de Cancerología, Mexico City, Mexico
| |
Collapse
|
23
|
Olivier T, Haslam A, Ochoa D, Fernandez E, Prasad V. Bedside implications of the use of surrogate endpoints in solid and haematological cancers: implications for our reliance on PFS, DFS, ORR, MRD and more. BMJ ONCOLOGY 2024; 3:e000364. [PMID: 39886154 PMCID: PMC11557723 DOI: 10.1136/bmjonc-2024-000364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 09/12/2024] [Indexed: 02/01/2025]
Abstract
Clinical endpoints, such as overall survival, directly measure relevant outcomes. Surrogate endpoints, in contrast, are intermediate, stand-in measures of various tumour-related metrics and include tumour growth, tumour shrinkage, blood results, etc. Surrogates may be a time point measurement, that is, tumour shrinkage at some point (eg, response rate) or biomarker-assessed disease status, measured at given time points (eg, circulating tumour DNA, ctDNA). They can also be measured over time, as with progression-free survival, which is the time until a patient presents with either disease progression or death. Surrogates are increasingly used in trials supporting the marketing authorisation of novel oncology drugs. Yet, the trial-level correlation between surrogates and clinical endpoints-meaning to which extent an improvement in the surrogate predicts an improvement in the direct endpoint-is often moderate to low. Here, we provide a comprehensive classification of surrogate endpoints: time point measurements and time-to-event endpoints in solid and haematological malignancies. Also, we discuss an overlooked aspect of the use of surrogates: the limitations of surrogates outside trial settings, at the bedside. Surrogates can result in the inappropriate stopping or switching of therapy. Surrogates can be used to usher in new strategies (eg, ctDNA in adjuvant treatment of colon cancer), which may erode patient outcomes. In liquid malignancies, surrogates can mislead us to use novel drugs and replace proven standards of care with costly medications. Surrogates can lead one to intensify treatment without clear improvement and possibly worsening quality of life. Clinicians should be aware of the role of surrogates in the development and regulation of drugs and how their use can carry real-world, bedside implications.
Collapse
Affiliation(s)
- Timothée Olivier
- Department of Oncology, Geneva University Hospitals, Geneve, Switzerland
| | - Alyson Haslam
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Dagney Ochoa
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Eduardo Fernandez
- Jane Anne Nohl Division of Hematology and Center for the Study of Blood Diseases, USC Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
24
|
Nguyen TPM, Woods SL, Secombe KR, Tang S, Elz AS, Ayton S, Finnie J, Nagpal A, Pouliot N, Bowen JM. Ferroptosis - a potential feature underlying neratinib-induced colonic epithelial injury. Cancer Chemother Pharmacol 2024; 94:493-505. [PMID: 39002022 PMCID: PMC11438713 DOI: 10.1007/s00280-024-04699-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE Neratinib, a small-molecule tyrosine kinase inhibitor (TKI) that irreversibly binds to human epidermal growth factor receptors 1, 2 and 4 (HER1/2/4), is an approved extended adjuvant therapy for patients with HER2-amplified or -overexpressed (HER2-positive) breast cancers. Patients receiving neratinib may experience mild-to-severe symptoms of gut toxicity including abdominal pain and diarrhoea. Despite being a highly prevalent complication in gut health, the biological processes underlying neratinib-induced gut injury, especially in the colon, remains unclear. METHODS Real-time quantitative polymerase chain reaction (RT-qPCR) and histology were integrated to study the effect of, and type of cell death induced by neratinib on colonic tissues collected from female Albino Wistar rats dosed with neratinib (50 mg/kg) daily for 28 days. Additionally, previously published bulk RNA-sequencing and CRISPR-screening datasets on human glioblastoma SF268 cell line and glioblastoma T895 xenograft, and mouse TBCP1 breast cancer cell line were leveraged to elucidate potential mechanisms of neratinib-induced cell death. RESULTS The severity of colonic epithelial injury, especially degeneration of surface lining colonocytes and infiltration of immune cells, was more pronounced in the distal colon than the proximal colon. Sequencing showed that apoptotic gene signature was enriched in neratinib-treated SF268 cells while ferroptotic gene signature was enriched in neratinib-treated TBCP1 cells and T895 xenograft. However, we found that ferroptosis, but less likely apoptosis, was a potential histopathological feature underlying colonic injury in rats treated with neratinib. CONCLUSION Ferroptosis is a potential feature of neratinib-induced colonic injury and that targeting molecular machinery governing neratinib-induced ferroptosis may represent an attractive therapeutic approach to ameliorate symptoms of gut toxicity.
Collapse
Affiliation(s)
- Triet P M Nguyen
- School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia.
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia.
| | - Susan L Woods
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Kate R Secombe
- School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Simon Tang
- School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Aurelia S Elz
- School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - John Finnie
- Discipline of Anatomy and Pathology, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Aadya Nagpal
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3000, Australia
| | - Normand Pouliot
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Joanne M Bowen
- School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
25
|
Xu L, Xie Y, Gou Q, Cai R, Bao R, Huang Y, Tang R. HER2-targeted therapies for HER2-positive early-stage breast cancer: present and future. Front Pharmacol 2024; 15:1446414. [PMID: 39351085 PMCID: PMC11439691 DOI: 10.3389/fphar.2024.1446414] [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: 06/09/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Breast cancer (BC) has the second highest incidence among cancers and is the leading cause of death among women worldwide. The human epidermal growth factor receptor 2 (HER2) is overexpressed in approximately 20%-30% of BC patients. The development of HER2-targeted drugs, including monoclonal antibodies (mAbs), tyrosine kinase inhibitors (TKIs) and antibody-drug conjugates (ADCs), has improved the operation rate and pathological remission rate and reduced the risk of postoperative recurrence for HER2-positive early-stage BC (HER2+ EBC) patients. This review systematically summarizes the mechanisms, resistance, therapeutic modalities and safety of HER2-targeted drugs and helps us further understand these drugs and their use in clinical practice for patients with HER2+ EBC.
Collapse
Affiliation(s)
- Luying Xu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxin Xie
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiheng Gou
- Department of Radiation Oncology and Department of Head & Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Cai
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Bao
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yucheng Huang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ruisi Tang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Disease Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Jung YJ, Lee S, Kang SK, Kim JY, Choo KS, Nam KJ, Joo JH, Kim JJ, Kim HY. Clinicopathological Factors Predicting Pathological Complete Response to Neoadjuvant Anti-HER2 Therapy in HER2-Positive Breast Cancer. Oncology 2024; 103:351-359. [PMID: 39250898 PMCID: PMC12048099 DOI: 10.1159/000541019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Human epidermal growth factor receptor 2 (HER2)-targeted therapies have shown effectiveness against HER2-positive breast cancer. This makes neoadjuvant chemotherapy (NAC) a valuable option for treating both early and advanced stages of the disease. The tumor's response to HER2-targeted NAC provides crucial prognostic information. Additionally, it allows for tailoring adjuvant treatment strategies for HER2+ breast cancer based on pathological responses. This study aimed to investigate the clinicopathological factors that influence tumor response. METHODS We retrospectively analyzed 122 patients diagnosed with HER2+ breast cancer. These patients received NAC and HER2-directed therapy between January 2018 and December 2022 at the Pusan National University Yangsan Hospital. Following surgery, tumor response was evaluated, categorizing patients into two groups: pathological complete response (pCR) and non-pCR groups. We analyzed data on various factors, including age, NAC regimen, type of breast and axillary surgery, clinical stage (cTNM), historical grade, and preoperative levels of carcinoembryonic antigen, cancer antigen 15-3 (CA 15-3), estrogen receptor (ER), progesterone receptor (PR), HER2, p53, and KI-67. RESULTS Out of the 122 patients, 75 achieved pCR, while 47 did not. Most clinicopathological factors showed no significant difference between the pCR and non-pCR groups. However, several factors were associated with a higher pCR rate: normal preoperative CA 15-3 levels (odds ratio [OR]: 3.74, confidence interval [CI]: 1.19-11.72, p = 0.02), preoperative ER positivity (OR: 2.65, CI: 1.25-5.59, p = 0.01), PR negativity (OR: 3.92, CI: 1.82-8.45, p < 0.05), and strong preoperative HER2 immunohistochemistry (IHC) 3+ staining. Multivariate analysis confirmed that PR negativity (OR: 2.8, CI: 1.23-6.42, p = 0.01) and strong preoperative HER2 IHC 3+ staining (OR: 0.18, CI: 0.03-0.84, p = 0.04) were independent predictors of a higher pCR rate. CONCLUSIONS A pCR after NAC impacts patient prognosis and influences the choice of adjuvant treatment for HER2+ breast cancer. Clinicopathological factors can help predict responses to HER2-targeted NAC. In our study, pre-ER/PR negativity, high pre-HER2 levels, and normal CA 15-3 levels were found to be potential predictors of pCR. These findings may contribute to developing more effective treatment strategies for HER2+ breast cancer.
Collapse
Affiliation(s)
- Youn Joo Jung
- Department of Surgery, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| | - Seungju Lee
- Department of Surgery, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| | - Seok Kyeong Kang
- Department of Surgery, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| | - Jee Yeon Kim
- Department of Pathology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| | - Ki Seok Choo
- Department of Radiology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| | - Kyung Jin Nam
- Department of Radiology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| | - Ji Hyeon Joo
- Department of Radiation Oncology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| | - Jae Joon Kim
- Department of Hematology and Oncology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| | - Hyun Yul Kim
- Department of Surgery, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| |
Collapse
|
27
|
Steadman JA, Hieken TJ. Advances from targeted therapy for non-metastatic HER2-positive inflammatory breast cancer. J Surg Oncol 2024; 130:366-370. [PMID: 39101322 DOI: 10.1002/jso.27797] [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: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/06/2024]
Abstract
Among inflammatory breast cancer (IBC) patients, over one-third have HER2-overexpressing (HER2+) tumors. Pathologic complete response (pCR) rates to neoadjuvant targeted and chemotherapy for patients with HER2+ non-metastatic IBC now apporach 60% and favorable long-term survival rates are being reported for those with a pCR. Immune mechanisms contributing to this phenomenon include antibody-mediated immune activation and induction of memory T-cell reponses which may explain the sustained antitumor response seen after discontinuation of targeted therapies.
Collapse
Affiliation(s)
- Jessica A Steadman
- Department of Surgery, Division of Breast and Melanoma Surgical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Tina J Hieken
- Department of Surgery, Division of Breast and Melanoma Surgical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
28
|
Zheng H, Mahmood SS, Khalique OK, Zhan H. Trastuzumab-Induced Cardiotoxicity: When and How Much Should We Worry? JCO Oncol Pract 2024; 20:1055-1063. [PMID: 38662969 DOI: 10.1200/op.23.00816] [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: 12/21/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 08/15/2024] Open
Abstract
This review critically analyzes the incidence of trastuzumab-induced left ventricular systolic dysfunction and congestive heart failure (CHF), distinguishing between cases with and without prior anthracycline exposure. It highlights the fact that the elevated risk of trastuzumab-induced cardiotoxicity is closely associated with prior anthracycline exposure. In the absence of prior anthracycline exposure, the incidence rates of trastuzumab-induced cardiotoxicity, particularly CHF (ranging from 0% to 0.5%), are largely comparable with those reported in the general population, especially when reversibility is taken into account. Current cardiac surveillance recommendations during trastuzumab treatment have not yet adapted to the increasing adoption of nonanthracycline treatment strategies and the associated low risk of cardiotoxicity. We propose a refined monitoring protocol to reduce the frequency of cardiac evaluations for low-risk to moderate-risk patients, especially those receiving nonanthracycline treatments. By focusing on patients at high risk or those with prior anthracycline exposure, this strategy seeks to optimize the cost-effectiveness of cardiac care in oncology.
Collapse
Affiliation(s)
- Haoyi Zheng
- CardioOncology Service, Saint Francis Hospital & Heart Center, Roslyn, NY
- Division of Cardiovascular Imaging, Saint Francis Hospital & Heart Center, Roslyn, NY
| | - Syed S Mahmood
- CardioOncology Service, Saint Francis Hospital & Heart Center, Roslyn, NY
| | - Omar K Khalique
- Division of Cardiovascular Imaging, Saint Francis Hospital & Heart Center, Roslyn, NY
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY
- Medical Service, Northport Veterans Affairs Medical Center, Northport, NY
| |
Collapse
|
29
|
Ketch PW, Zaharias RS, Leath CA. Pharmacotherapy for cervical cancer: current standard of care and new perspectives. Expert Opin Pharmacother 2024; 25:1591-1603. [PMID: 39164924 PMCID: PMC11453679 DOI: 10.1080/14656566.2024.2395379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/19/2024] [Indexed: 08/22/2024]
Abstract
INTRODUCTION Cervical cancer, while highly preventable, remains an international public health challenge especially in under resourced regions. Although early-stage cervix confined cancers are often amenable to surgical resection, larger tumors deemed locally advanced cervical cancer (LACC) necessitate systemic therapy as part of chemoradiation therapy. Moreover, systemic therapy is the standard therapeutic approach for those presenting with primary metastasis or recurrence. AREAS COVERED While several agents have been approved to treat recurrent cervical cancer including checkpoint inhibitors as well as both biomarker agnostic and specific antibody drug conjugates, the development of agents added to chemoradiation has been less fruitful. Until recently, the addition of novel therapies to chemoradiation has been negative in terms of improving outcomes; however, results of a recent Phase III clinical trial (NCT04221945) in LACC demonstrated that the addition of pembrolizumab to standard of care chemoradiation was associated with an improvement in progression-free survival and resulted in an FDA approval for this therapy. This observation led to the first change in treating LACC since the early 2000s. EXPERT OPINION Improvements in systemic therapy both alone and in combination with chemoradiation for cervical cancer have been realized. Ongoing research is needed for therapeutic options following immunotherapy.
Collapse
Affiliation(s)
- Peter W. Ketch
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rennan S. Zaharias
- Division of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Charles A. Leath
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
30
|
Bowen J, Braga S, Zotto VD, Finnie J, DiPrimeo D, Cooke B, Bischof GF, Wong A, Di Palma JA. Preclinical and clinical evaluation through serial colonoscopic evaluation of neratinib-induced diarrhea in HER2-positive breast cancer-A pilot study. Physiol Rep 2024; 12:e70008. [PMID: 39187401 PMCID: PMC11347019 DOI: 10.14814/phy2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024] Open
Abstract
The irreversible pan-HER tyrosine kinase inhibitor neratinib is approved for patients with HER2-positive, early-stage and metastatic breast cancer (BC). Neratinib-associated diarrhea is the most common reason for early discontinuation. Preclinical studies identified mechanisms of neratinib-induced diarrhea and rationale for prophylactic and preventive measures. We studied effects of neratinib on rat intestines and conducted a phase 2 study of colon pathogenesis in patients with HER2-positive BC treated with neratinib (NCT04366713). Colon samples from female albino Wistar rats receiving neratinib or vehicle were examined for histopathological changes. Patients with HER2-positive BC received neratinib 240 mg once daily for up to 1 year. Colonoscopy biopsies were collected at baseline and at Day 28 to identify changes consistent with rat pathologies. Rat colons were markedly altered in appearance, with similar short circuit currents (Isc) and responses to carbachol and forskolin. Mucosal barrier loss and/or significant increase in secretory propensity in neratinib- versus control-treated animals were not seen. Two of four endpoint-evaluable patients presented with mild pathological changes, largely comparable with the rat model. Preclinical evidence supports an inflammatory component of neratinib-induced diarrhea without mucosal barrier function loss. Colonoscopy findings in patients with BC indicate mild or no pathological changes in the colon due to neratinib treatment.
Collapse
Affiliation(s)
- Joanne Bowen
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
| | - Sofia Braga
- Medical OncologyHospital Prof. Doutor Fernando Fonseca, EPEAmadoraPortugal
| | - Valeria Dal Zotto
- Department of PathologyThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - John Finnie
- Division of Research and InnovationUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | | | - Blaire Cooke
- Puma Biotechnology, Inc.Los AngelesCaliforniaUSA
| | | | - Alvin Wong
- Puma Biotechnology, Inc.Los AngelesCaliforniaUSA
| | - Jack A. Di Palma
- Division of GastroenterologyUniversity of South Alabama, College of MedicineMobileAlabamaUSA
| |
Collapse
|
31
|
Cassoli JPB, Fernandes Í, Carvalho L, Fernandes M, Centrone AF, Taniwaki L, Lima RDC, Junior UDR, Dias IWR, Taranto P, Beal J, de Lima FT, Moura F, Cendoroglo M, Araújo SEA, Uson Junior PLS. Frequency of Deleterious Germline Variants in HER2-Low Breast Cancer Patients Using a Hereditary Multipanel Gene Testing. Curr Issues Mol Biol 2024; 46:7976-7985. [PMID: 39194688 DOI: 10.3390/cimb46080471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
HER2-Low is defined as low levels of HER2 expression, based on a score of 1+ on immunohistochemical (IHC) assay or as an IHC score of 2+ and negative results on in situ hybridization (ISH or FISH). They are a heterogeneous population of breast cancers that vary in prognosis and sensitivity to systemic treatments. The frequency and clinical characteristics of pathogenic germline variants (PGVs) in HER2-Low breast cancer (BC) patients is not defined. We analyzed results from patients with BC who underwent multi-gene panel testing (MGPT) (maximum 145 genes) between 2018-2019. We reclassified HER-2 status accordingly. Relationships between the variables of interest were assessed by adopting the proportional regression Cox models. Of a total of 167 BC patients who underwent MGPT, half were hormone-receptor-positive. The median age was 45 years. About two thirds of the patients were in the earlier stage of BC. A total of 57% of the cases were reclassified as HER-2-negative or -Low. PGVs were found in 19% of the patients overall, as follows: seven BRCA1, four BRCA2, two ATM, one ATR, two CFTR, three CHEK2, one FANCA, one MERTK, one MLH1, three MUTYH, one RAD50, three RAD51C, one RECQL4, and two TP53 mutations. In HER2-Low, 26.5% of the patients had PGVs, and in the overall cohort, this was 19.8%. In conclusion, differences in the prevalence of deleterious germline mutations in HER2-Low BC patients compared to non-HER2-Low BC patients were identified. Similar alterations in BRCA were observed in this group of patients compared to the overall cohort. Germline genetic tests should be evaluated in larger cohorts of patients with HER2-Low status to better address the findings.
Collapse
Affiliation(s)
- Janaina Pontes Batista Cassoli
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Ítalo Fernandes
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Leonardo Carvalho
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Milena Fernandes
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Ana Fernanda Centrone
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Letícia Taniwaki
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Rita de Cássia Lima
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | | | - Igor Wanderley Reis Dias
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Patrícia Taranto
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Juliana Beal
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Fernanda Teresa de Lima
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Fernando Moura
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Miguel Cendoroglo
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Sergio Eduardo Alonso Araújo
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Pedro Luiz Serrano Uson Junior
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| |
Collapse
|
32
|
Jiang M, Chai Y, Liu J, He M, Wang Y, Yang X, Xing Z, Zhang M, Zhou S, Ma F, Wang J, Yuan P, Xu B, Li Q. Neoadjuvant inetetamab and pertuzumab with taxanes and carboplatin (TCbIP) In locally advanced HER2-positive breast cancer: a prospective cohort study with propensity-matched analysis. BMC Cancer 2024; 24:877. [PMID: 39039516 PMCID: PMC11265051 DOI: 10.1186/s12885-024-12654-3] [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: 04/24/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Inetetamab is the first domestically developed innovative anti-HER2 monoclonal antibody in China, proven effective and safe in HER2-positive advanced breast cancer. However, its efficacy and safety in neoadjuvant treatment of HER2-positive locally advanced breast cancer (LABC) remain to be validated. METHODS This prospective cohort study aimed to evaluate the efficacy and safety of inetetamab combined with pertuzumab, taxanes, and carboplatin (TCbIP) in neoadjuvant therapy for HER2-positive LABC, comparing it to data from patients treated with the TCbHP regimen (trastuzumab combined with pertuzumab, taxanes, and carboplatin) using propensity score matching (PSM). The primary endpoint was total pathological complete response (tpCR). Adverse events (AEs), objective response rate (ORR), and near-pCR were key secondary endpoints. RESULTS Forty-four patients with clinical stage IIA-IIIC HER2-positive LABC were prospectively enrolled and treated with the TCbIP regimen. The tpCR rate among 28 patients who completed surgery was 60.7%, comparable to and slightly higher than the TCbHP group in PSM (60.7% vs. 53.6%, P = 0.510). The ORR was 96.4%, and the DCR reached 100.0%. The most common ≥ grade 3 AE was neutropenia (21.4% vs. 11.9%, P = 0.350). No significant reduction in left ventricular ejection fraction was observed, and no patient withdrew from treatment due to AEs. CONCLUSION Neoadjuvant therapy with TCbIP showed good efficacy and safety in patients with HER2-positive LABC and might be another promising option for neoadjuvant treatment. TRIAL REGISTRATION NCT05749016 (registration date: Nov 01, 2021).
Collapse
Affiliation(s)
- Mingxia Jiang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Chai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaxuan Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Maiyue He
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yipeng Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Yang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zeyu Xing
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengqi Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shihan Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiayu Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Qiao Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
33
|
Gupta M, Akhtar OS, Bahl B, Mier-Hicks A, Attwood K, Catalfamo K, Gyawali B, Torka P. Health-related quality of life outcomes reporting associated with FDA approvals in haematology and oncology. BMJ ONCOLOGY 2024; 3:e000369. [PMID: 39886148 PMCID: PMC11256025 DOI: 10.1136/bmjonc-2024-000369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/21/2024] [Indexed: 02/01/2025]
Abstract
Objective Health-related quality of life (HRQoL) outcomes are important in making clinical and policy decisions. This study aimed to examine the HRQoL reporting in cancer drug trials leading to Food and Drug Administration (FDA) approvals. Methods and analysis This retrospective cohort study analysed HRQoL data for trials leading to FDA approvals between July 2015 and May 2020. Proportion of included trials that reported HRQoL, latency between FDA approval and first report of HRQoL data, HRQoL outcomes, and their correlation with OS (overall survival) and PFS (progression-free survival) were analysed. Results Of the 233 trials associated with 207 FDA approvals, HRQoL was reported in 50% of trials, of which only 42% had the data reported by the time of FDA approval. There were no changes in frequency of HRQoL reporting between 2015 and 2020. HRQoL data were first reported in the primary publication in only 30% trials. Of the 115 trials with HRQoL data available, HRQoL improved in 43%, remained stable in 53% and worsened in 4% of trials. Among the trials that led to FDA approvals based on surrogate endpoints (79%), HRQoL was reported in 45% and improved only in 18% trials. There was no association between OS and PFS benefit and HRQoL outcomes. Conclusion Rates of HRQoL reporting were suboptimal in trials that led to FDA approvals with no improvements seen between 2015 and 2020. HRQoL reporting was often delayed and not presented in the primary publication. HRQoL reporting was further sparse in trials with approvals based on surrogate endpoints and HRQoL improved in only a minority of them.
Collapse
Affiliation(s)
- Medhavi Gupta
- Program in Women's Oncology, Brown University, Providence, Rhode Island, USA
| | | | - Bhavyaa Bahl
- University of Colorado System, Denver, Colorado, USA
| | | | | | - Kayla Catalfamo
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | | | - Pallawi Torka
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
34
|
Jasim SA, Farber IM, Noraldeen SAM, Bansal P, Alsaab HO, Abdullaev B, Alkhafaji AT, Alawadi AH, Hamzah HF, Mohammed BA. Incorporation of immunotherapies and nanomedicine to better normalize angiogenesis-based cancer treatment. Microvasc Res 2024; 154:104691. [PMID: 38703993 DOI: 10.1016/j.mvr.2024.104691] [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: 02/19/2024] [Revised: 04/11/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
Neoadjuvant targeting of tumor angiogenesis has been developed and approved for the treatment of malignant tumors. However, vascular disruption leads to tumor hypoxia, which exacerbates the treatment process and causes drug resistance. In addition, successful delivery of therapeutic agents and efficacy of radiotherapy require normal vascular networks and sufficient oxygen, which complete tumor vasculopathy hinders their efficacy. In view of this controversy, an optimal dose of FDA-approved anti-angiogenic agents and combination with other therapies, such as immunotherapy and the use of nanocarrier-mediated targeted therapy, could improve therapeutic regimens, reduce the need for administration of high doses of chemotherapeutic agents and subsequently reduce side effects. Here, we review the mechanism of anti-angiogenic agents, highlight the challenges of existing therapies, and present how the combination of immunotherapies and nanomedicine could improve angiogenesis-based tumor treatment.
Collapse
Affiliation(s)
| | - Irina M Farber
- Department of children's diseases of the F. Filatov clinical institute of children's health, I. M. Sechenov First Moscow State Medical University of Health of Russian Federation (Sechenov University), Moscow, Russia
| | | | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| | - Bekhzod Abdullaev
- Research Department of Biotechnology, New Uzbekistan University, Mustaqillik Avenue 54, Tashkent 100007, Uzbekistan; Department of Oncology, School of Medicine, Central Asian University, Milliy Bog Street 264, Tashkent 111221, Uzbekistan..
| | | | - Ahmed Hussien Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Qadisiyyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | |
Collapse
|
35
|
Khan S, Araji G, Yetiskul E, Keesari PR, Haddadin F, Khamis Z, Chowdhry V, Niazi M, Afif S, Dhar M, El-Sayegh S. Systemic oncological therapy in breast cancer patients on dialysis. World J Clin Oncol 2024; 15:730-744. [PMID: 38946836 PMCID: PMC11212603 DOI: 10.5306/wjco.v15.i6.730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 06/24/2024] Open
Abstract
The advancement of renal replacement therapy has significantly enhanced the survival rates of patients with end-stage renal disease (ESRD) over time. However, this prolonged survival has also been associated with a higher likelihood of cancer diagnoses among these patients including breast cancer. Breast cancer treatment typically involves surgery, radiation, and systemic therapies, with approaches tailored to cancer type, stage, and patient preferences. However, renal replacement therapy complicates systemic therapy due to altered drug clearance and the necessity for dialysis sessions. This review emphasizes the need for optimized dosing and administration strategies for systemic breast cancer treatments in dialysis patients, aiming to ensure both efficacy and safety. Additionally, challenges in breast cancer screening and diagnosis in this population, including soft-tissue calcifications, are highlighted.
Collapse
Affiliation(s)
- Salman Khan
- Department of Internal Medicine, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| | - Ghada Araji
- Department of Internal Medicine, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| | - Ekrem Yetiskul
- Department of Internal Medicine, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| | - Praneeth Reddy Keesari
- Department of Internal Medicine, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| | - Fadi Haddadin
- Department of Internal Medicine, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| | - Zaid Khamis
- Department of Internal Medicine, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| | - Varun Chowdhry
- Department of Internal Medicine, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| | - Muhammad Niazi
- Department of Hematology and Oncology, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| | - Sarah Afif
- Department of Internal Medicine, CUNY School of Medicine, New York, NY 10031, United States
| | - Meekoo Dhar
- Department of Hematology and Oncology, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| | - Suzanne El-Sayegh
- Department of Internal Medicine, Northwell Health - Staten Island University Hospital, Staten Island, NY 10305, United States
| |
Collapse
|
36
|
Moyer CL, Lanier A, Qian J, Coleman D, Hill J, Vuligonda V, Sanders ME, Mazumdar A, Brown PH. IRX4204 Induces Senescence and Cell Death in HER2-positive Breast Cancer and Synergizes with Anti-HER2 Therapy. Clin Cancer Res 2024; 30:2558-2570. [PMID: 38578278 PMCID: PMC11145169 DOI: 10.1158/1078-0432.ccr-23-3839] [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: 12/08/2023] [Revised: 03/04/2024] [Accepted: 04/03/2024] [Indexed: 04/06/2024]
Abstract
PURPOSE Rexinoids, agonists of nuclear retinoid X receptor (RXR), have been used for the treatment of cancers and are well tolerated in both animals and humans. However, the usefulness of rexinoids in treatment of breast cancer remains unknown. This study examines the efficacy of IRX4204, a highly specific rexinoid, in breast cancer cell lines and preclinical models to identify a biomarker for response and potential mechanism of action. EXPERIMENTAL DESIGN IRX4204 effects on breast cancer cell growth and viability were determined using cell lines, syngeneic mouse models, and primary patient-derived xenograft (PDX) tumors. In vitro assays of cell cycle, apoptosis, senescence, and lipid metabolism were used to uncover a potential mechanism of action. Standard anti-HER2 therapies were screened in combination with IRX4204 on a panel of breast cancer cell lines to determine drug synergy. RESULTS IRX4204 significantly inhibits the growth of HER2-positive breast cancer cell lines, including trastuzumab and lapatinib-resistant JIMT-1 and HCC1954. Treatment with IRX4204 reduced tumor growth rate in the MMTV-ErbB2 mouse and HER2-positive PDX model by 49% and 44%, respectively. Mechanistic studies revealed IRX4204 modulates lipid metabolism and induces senescence of HER2-positive cells. In addition, IRX4204 demonstrates additivity and synergy with HER2-targeted mAbs, tyrosine kinase inhibitors, and antibody-drug conjugates. CONCLUSIONS These findings identify HER2 as a biomarker for IRX4204 treatment response and demonstrate a novel use of RXR agonists to synergize with current anti-HER2 therapies. Furthermore, our results suggest that RXR agonists can be useful for the treatment of anti-HER2 resistant and metastatic HER2-positive breast cancer.
Collapse
Affiliation(s)
- Cassandra L. Moyer
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amanda Lanier
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Qian
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Darian Coleman
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jamal Hill
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
37
|
Arshad M, Azad A, Chan PYK, Vigneswara V, Feldinger K, Nafi SNM, Laporte-Maguire E, De Santo C, Zuo J, Shaaban AM, Kong A. Neratinib could be effective as monotherapy or in combination with trastuzumab in HER2-low breast cancer cells and organoid models. Br J Cancer 2024; 130:1990-2002. [PMID: 38600326 PMCID: PMC11182766 DOI: 10.1038/s41416-024-02665-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 02/23/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Previous studies have suggested that patients with HER2-low breast cancers do not benefit from trastuzumab treatment although the reasons remain unclear. METHODS We investigated the effect of trastuzumab monotherapy and its combination with different HER2 targeting treatments in a panel of breast cancer cell lines and patient-derived organoids (PDOs) using biochemical methods and cell viability assays. RESULTS Compared to sensitive HER2 over-expressing (IHC3 + ) breast cancer cells, increasing doses of trastuzumab could not achieve IC50 in MDA-MB-361 (IHC 2 + FISH + ) and MDA-MB-453 (IHC 2 + FISH-) cells which showed an intermediate response to trastuzumab. Trastuzumab treatment induced upregulation of HER ligand release, resulting in the activation of HER receptors in these cells, which could account for their trastuzumab insensitivity. Adding a dual ADAM10/17 inhibitor to inhibit the shedding of HER ligands in combination with trastuzumab only showed a modest decrease in the cell viability of HER2-low breast cancer cells and PDOs. However, the panHER inhibitor neratinib was an effective monotherapy in HER2-low breast cancer cells and PDOs, and showed additive effects when combined with trastuzumab. CONCLUSION This study demonstrates that neratinib in combination with trastuzumab may be effective in a subset of HER2-low breast cancers although further validation is required in a larger panel of PDOs and in future clinical studies.
Collapse
Affiliation(s)
- Maryam Arshad
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - Abul Azad
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK
| | - Phoebe Yuen Ka Chan
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK
| | - Vasanthy Vigneswara
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - Katharina Feldinger
- Previous association, Department of Molecular Oncology, The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Siti Norasikin Mohd Nafi
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kota Bharu, Kelantan, Malaysia
| | - Eloise Laporte-Maguire
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK
| | - Carmela De Santo
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, B15 2TT, UK
| | - Abeer M Shaaban
- Department of cellular pathology, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Anthony Kong
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK.
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT, UK.
- Previous association, Department of Molecular Oncology, The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| |
Collapse
|
38
|
Bartsch R, Rinnerthaler G, Petru E, Egle D, Gnant M, Balic M, Sliwa T, Singer C. Updated Austrian treatment algorithm for metastatic triple-negative breast cancer. Wien Klin Wochenschr 2024; 136:347-361. [PMID: 37682349 PMCID: PMC11156740 DOI: 10.1007/s00508-023-02254-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 09/09/2023]
Abstract
Approximately 15% of newly diagnosed breast cancer patients have neither hormone receptors expression nor HER2 overexpression and/or HER2/neu gene amplification. This subtype of breast cancer is known as Triple Negative Breast Cancer (TNBC), and carries a significantly elevated risk of local and distant recurrence. In comparison with other breast cancer subtypes, there is a higher rate of visceral and brain metastases. The majority of metastases of TNBC are diagnosed within three years after initial breast cancer diagnosis. While there have been major advances in hormone-receptor- positive and in human epidermal growth factor receptor 2 (HER2)-positive disease over the past two decades, only limited improvements in outcomes for patients with triple negative breast cancer (TNBC) have been observed. A group of Austrian breast cancer specialists therefore convened an expert meeting to establish a comprehensive clinical risk-benefit profile of available mTNBC therapies and discuss the role sacituzumab govitecan may play in the treatment algorithm of the triple-negative breast cancer patients.
Collapse
Affiliation(s)
- Rupert Bartsch
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Gabriel Rinnerthaler
- Third Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, 5020, Salzburg, Austria
| | - Edgar Petru
- Department of Gynecology and Obstetrics, Division of Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036, Graz, Austria
| | - Daniel Egle
- Department of Gynecology, Breast Cancer Center Tirol, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Michael Gnant
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Marija Balic
- Department of Internal Medicine, Division of Clinical Oncology, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Thamer Sliwa
- 3rd Medical Department, Hematology and Oncology, Hanusch Hospital, Heinrich-Collin-Straße 30, 1140, Vienna, Austria
| | - Christian Singer
- Department of Gynecology, Breast Cancer Center Vienna, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
39
|
Rotem O, Geiger KR, Hanovich E, Moskovitz M, Kurman N, Reinhorn D, Peretz I, Yerushalmi R, Stemmer SM. Seeing the Trees From the Forest: Challenges in Subgroup Analysis-Based Guidelines in Oncology. Oncol Rev 2024; 18:1355256. [PMID: 38855534 PMCID: PMC11162106 DOI: 10.3389/or.2024.1355256] [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: 12/13/2023] [Accepted: 05/03/2024] [Indexed: 06/11/2024] Open
Abstract
As clinical trials in oncology require substantial efforts, maximizing the insights gained from them by conducting subgroup analyses is often attempted. The goal of these analyses is to identify subgroups of patients who are likely to benefit, as well as the subgroups of patients who are unlikely to benefit from the studied intervention. International guidelines occasionally include or exclude novel medications and technologies for specific subpopulations based on such analyses of pivotal trials without requiring confirmatory trials. This Perspective discusses the importance of providing a complete dataset of clinical information when reporting subgroup analyses and explains why such transparency is key for better clinical interpretation of the results and the appropriate application to clinical care, by providing examples of transparent reporting of clinical studies and examples of incomplete reporting of clinical studies.
Collapse
Affiliation(s)
- Ofer Rotem
- Davidoff Center, Rabin Medical Center, Petah Tikva, Israel
| | - Karyn Revital Geiger
- Leumit Health Services, Tel Aviv, Israel
- Coller School of Management, Tel Aviv University, Tel Aviv, Israel
| | | | - Mor Moskovitz
- Davidoff Center, Rabin Medical Center, Petah Tikva, Israel
| | - Noga Kurman
- Davidoff Center, Rabin Medical Center, Petah Tikva, Israel
| | - Daniel Reinhorn
- Davidoff Center, Rabin Medical Center, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Idit Peretz
- Davidoff Center, Rabin Medical Center, Petah Tikva, Israel
| | - Rinat Yerushalmi
- Davidoff Center, Rabin Medical Center, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Salomon M. Stemmer
- Davidoff Center, Rabin Medical Center, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
He M, Zhao W, Wang P, Li W, Chen H, Yuan Z, Pan G, Gao H, Sun L, Chu J, Li L, Hu Y. Efficacy and safety of Trastuzumab Emtansine in treating human epidermal growth factor receptor 2-positive metastatic breast cancer in Chinese population: a real-world multicenter study. Front Med (Lausanne) 2024; 11:1383279. [PMID: 38741766 PMCID: PMC11089149 DOI: 10.3389/fmed.2024.1383279] [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: 02/07/2024] [Accepted: 04/04/2024] [Indexed: 05/16/2024] Open
Abstract
Background Trastuzumab emtansine (T-DM1) has been approved worldwide for treating metastatic breast cancer (mBC) in patients who have received first-line therapy, shown disease progression, and are human epidermal growth factor receptor 2 (HER2)-positive. T-DM1 received approval in China to treat early-stage breast cancer (BC) in 2020 and for mBC in 2021. In March 2023, T-DM1 was included in medical insurance coverage, significantly expanding the eligible population. Materials and methods This post-marketing observational study aimed to assess the safety and effectiveness of T-DM1 in real-world clinical practice in China. This study enrolled 31 individuals with HER2-positive early-stage BC and 70 individuals with HER2-positive advanced BC from 8 study centers in Shandong Province, China. The T-DM1 dosage was 3.6 mg/kg injected intravenously every 3 weeks until the disease advanced or the drug toxicity became uncontrollable, whichever occurred earlier. Additionally, efficacy and safety information on T-DM1 were collected. Results During the 7-month follow-up period, no recurrence or metastases were observed in patients who had early-stage BC. The disease control rate was 31.43% (22/70) in patients with advanced BC. The most common adverse effect of T-DM1 was thrombocytopenia, with an incidence of 69.31% (70/101), and the probability of Grade ≥ 3 thrombocytopenia was 11.88% (12/101). Conclusion This real-world study demonstrated that T-DM1 had good efficacy and was well tolerated by both HER2-positive early-stage BC and mBC patients.
Collapse
Affiliation(s)
- Miao He
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
- Department of Medical Oncology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Wen Zhao
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Peng Wang
- Department of Medical Oncology, Qingdao Shibei Changqing Hospital, Qingdao, Shandong Province, China
| | - Wenhuan Li
- Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Hanhan Chen
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zonghuai Yuan
- Department of General Surgery, People’s Hospital of Rizhao, Rizhao, Shandong Province, China
| | - Guangye Pan
- Department of General Surgery, People’s Hospital of Rizhao, Rizhao, Shandong Province, China
| | - Hong Gao
- Department of Breast and Thyroid Surgery, Rizhao Traditional Chinese Medical Hospital, Rizhao, Shandong Province, China
| | - Lijun Sun
- Department of Breast and Thyroid Surgery, People’s Hospital of Juxian, Rizhao, Shandong Province, China
| | - Jiahui Chu
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Li Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Yu Hu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
41
|
Wang J, Li B, Luo M, Huang J, Zhang K, Zheng S, Zhang S, Zhou J. Progression from ductal carcinoma in situ to invasive breast cancer: molecular features and clinical significance. Signal Transduct Target Ther 2024; 9:83. [PMID: 38570490 PMCID: PMC10991592 DOI: 10.1038/s41392-024-01779-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Ductal carcinoma in situ (DCIS) represents pre-invasive breast carcinoma. In untreated cases, 25-60% DCIS progress to invasive ductal carcinoma (IDC). The challenge lies in distinguishing between non-progressive and progressive DCIS, often resulting in over- or under-treatment in many cases. With increasing screen-detected DCIS in these years, the nature of DCIS has aroused worldwide attention. A deeper understanding of the biological nature of DCIS and the molecular journey of the DCIS-IDC transition is crucial for more effective clinical management. Here, we reviewed the key signaling pathways in breast cancer that may contribute to DCIS initiation and progression. We also explored the molecular features of DCIS and IDC, shedding light on the progression of DCIS through both inherent changes within tumor cells and alterations in the tumor microenvironment. In addition, valuable research tools utilized in studying DCIS including preclinical models and newer advanced technologies such as single-cell sequencing, spatial transcriptomics and artificial intelligence, have been systematically summarized. Further, we thoroughly discussed the clinical advancements in DCIS and IDC, including prognostic biomarkers and clinical managements, with the aim of facilitating more personalized treatment strategies in the future. Research on DCIS has already yielded significant insights into breast carcinogenesis and will continue to pave the way for practical clinical applications.
Collapse
Affiliation(s)
- Jing Wang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Baizhou Li
- Department of Pathology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Meng Luo
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
- Department of Plastic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Huang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Zheng
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Suzhan Zhang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
| | - Jiaojiao Zhou
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
42
|
Tommasi C, Airò G, Pratticò F, Testi I, Corianò M, Pellegrino B, Denaro N, Demurtas L, Dessì M, Murgia S, Mura G, Wekking D, Scartozzi M, Musolino A, Solinas C. Hormone Receptor-Positive/HER2-Positive Breast Cancer: Hormone Therapy and Anti-HER2 Treatment: An Update on Treatment Strategies. J Clin Med 2024; 13:1873. [PMID: 38610638 PMCID: PMC11012464 DOI: 10.3390/jcm13071873] [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: 02/20/2024] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Hormone receptor (HR)-positive/HER2-positive breast cancer represents a distinct subtype expressing estrogen and progesterone receptors with an overexpression of HER2. Approximately 14% of female breast cancer cases are HER2-positive, with the majority being HR-positive. These tumors show a cross-talk between the hormonal and HER2 pathways; the interaction has implications for the treatment options for the disease. In this review, we analyze the biology of HR-positive/HER2-positive breast cancer and summarize the evidence concerning the standard of care options both in neoadjuvant/adjuvant settings and in advanced disease. Additionally, we focus on new trials and drugs for HR-positive/HER2-positive breast cancer and the new entity: HER2-low breast cancer.
Collapse
Affiliation(s)
- Chiara Tommasi
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Giulia Airò
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Fabiana Pratticò
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Irene Testi
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
| | - Matilde Corianò
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Benedetta Pellegrino
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Nerina Denaro
- Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Laura Demurtas
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, 09042 Monserrato, Italy (C.S.)
| | - Mariele Dessì
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, 09042 Monserrato, Italy (C.S.)
| | - Sara Murgia
- Medical Oncology, University of Cagliari, 09124 Cagliari, Italy
| | - Giovanni Mura
- Pathological Anatomy, Laboratory Valdès, 81200 Cagliari, Italy
| | - Demi Wekking
- Academic Medical Centre, Amsterdam University Medical Center, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Mario Scartozzi
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, 09042 Monserrato, Italy (C.S.)
- Medical Oncology, University of Cagliari, 09124 Cagliari, Italy
| | - Antonino Musolino
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy; (G.A.); (F.P.); (I.T.); (M.C.); (B.P.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
| | - Cinzia Solinas
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, 09042 Monserrato, Italy (C.S.)
| |
Collapse
|
43
|
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
|
44
|
Loibl S, André F, Bachelot T, Barrios CH, Bergh J, Burstein HJ, Cardoso MJ, Carey LA, Dawood S, Del Mastro L, Denkert C, Fallenberg EM, Francis PA, Gamal-Eldin H, Gelmon K, Geyer CE, Gnant M, Guarneri V, Gupta S, Kim SB, Krug D, Martin M, Meattini I, Morrow M, Janni W, Paluch-Shimon S, Partridge A, Poortmans P, Pusztai L, Regan MM, Sparano J, Spanic T, Swain S, Tjulandin S, Toi M, Trapani D, Tutt A, Xu B, Curigliano G, Harbeck N. Early breast cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2024; 35:159-182. [PMID: 38101773 DOI: 10.1016/j.annonc.2023.11.016] [Citation(s) in RCA: 261] [Impact Index Per Article: 261.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023] Open
Affiliation(s)
- S Loibl
- GBG Forschungs GmbH, Neu-Isenburg; Centre for Haematology and Oncology, Bethanien, Frankfurt, Germany
| | - F André
- Breast Cancer Unit, Medical Oncology Department, Gustave Roussy, Cancer Campus, Villejuif
| | - T Bachelot
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - C H Barrios
- Oncology Department, Latin American Cooperative Oncology Group and Oncoclínicas, Porto Alegre, Brazil
| | - J Bergh
- Department of Oncology-Pathology, Bioclinicum, Karolinska Institutet and Breast Cancer Centre, Karolinska Comprehensive Cancer Centre and University Hospital, Stockholm, Sweden
| | - H J Burstein
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - M J Cardoso
- Breast Unit, Champalimaud Foundation, Champalimaud Cancer Centre, Lisbon; Faculty of Medicine, Lisbon University, Lisbon, Portugal
| | - L A Carey
- Division of Medical Oncology, The University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Chapel Hill, USA
| | - S Dawood
- Department of Oncology, Mediclinic City Hospital, Dubai, UAE
| | - L Del Mastro
- Medical Oncology Clinic, IRCCS Ospedale Policlinico San Martino, Genoa; Department of Internal Medicine and Medical Specialities, School of Medicine, University of Genoa, Genoa, Italy
| | - C Denkert
- Institute of Pathology, Philipps-University Marburg and University Hospital Giessen and Marburg, Marburg
| | - E M Fallenberg
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - P A Francis
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - H Gamal-Eldin
- Department of Surgical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - K Gelmon
- Department of Medical Oncology, British Columbia Cancer, Vancouver, Canada
| | - C E Geyer
- Department of Internal Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, USA
| | - M Gnant
- Comprehensive Cancer Centre, Medical University of Vienna, Vienna, Austria
| | - V Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova; Oncology 2 Unit, Istituto Oncologico Veneto IOV IRCCS, Padova, Italy
| | - S Gupta
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India
| | - S B Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - D Krug
- Department of Radiation Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - M Martin
- Hospital General Universitario Gregorio Maranon, Universidad Complutense, GEICAM, Madrid, Spain
| | - I Meattini
- Department of Radiation Oncology, Azienda Ospedaliero-Universitaria Careggi, Florence; Department of Experimental and Clinical Biomedical Sciences 'M. Serio', University of Florence, Florence, Italy
| | - M Morrow
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - W Janni
- Department of Obstetrics and Gynaecology, University of Ulm, Ulm, Germany
| | - S Paluch-Shimon
- Sharett Institute of Oncology Department, Hadassah University Hospital & Faculty of Medicine Hebrew University, Jerusalem, Israel
| | - A Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - P Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Antwerp; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - L Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven
| | - M M Regan
- Division of Biostatistics, Dana-Farber Cancer Institute, Harvard Medical School, Boston
| | - J Sparano
- Department of Medicine, Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - T Spanic
- Europa Donna Slovenia, Ljubljana, Slovenia
| | - S Swain
- Medicine Department, Georgetown University Medical Centre and MedStar Health, Washington, USA
| | - S Tjulandin
- N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - M Toi
- Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Bunkyo-ku, Japan
| | - D Trapani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - A Tutt
- Breast Cancer Research Division, The Institute of Cancer Research, London; Comprehensive Cancer Centre, Division of Cancer Studies, Kings College London, London, UK
| | - B Xu
- Department of Medical Oncology, National Cancer Centre/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - G Curigliano
- Early Drug Development for Innovative Therapies Division, Istituto Europeo di Oncologia, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - N Harbeck
- Breast Centre, Department of Obstetrics & Gynaecology and Comprehensive Cancer Centre Munich, LMU University Hospital, Munich, Germany
| |
Collapse
|
45
|
Guy M, Teixeira A, Shrier A, Meschter C, Bolognese J, Chaturvedi P. Effects of orally administered crofelemer on the incidence and severity of neratinib-induced diarrhea in female dogs. PLoS One 2024; 19:e0282769. [PMID: 38265977 PMCID: PMC10807780 DOI: 10.1371/journal.pone.0282769] [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: 03/15/2023] [Accepted: 11/12/2023] [Indexed: 01/26/2024] Open
Abstract
Targeted therapies have increased cancer therapy-related diarrhea (CTD) burden, with high incidence and/or severity of diarrhea for some agents that inhibit epidermal growth factor receptor and receptor tyrosine kinases. Neratinib is a pan-HER tyrosine kinase inhibitor approved for breast cancer treatment and causes severe diarrhea in >95% of patients. Crofelemer, a novel intestinal chloride ion channel modulator, is an approved antidiarrheal drug for symptomatic relief of noninfectious diarrhea in patients with HIV/AIDS receiving antiretroviral therapy. The objective of this study was to evaluate the effectiveness of crofelemer prophylaxis in reducing the incidence /severity of neratinib-induced diarrhea without concomitant administration of loperamide in female beagle dogs. A pilot study using 3 dogs determined a maximum daily tolerated dose of neratinib was between 40 and 80 mg; this dose would induce a consistent incidence/severity of diarrhea without risking severe dehydration. In the definitive study, 24 female beagle dogs (8/group) received neratinib once daily and placebo capsules (CTR) four times/day, or neratinib once daily and crofelemer 125 mg delayed-release tablets given two times (BID), or neratinib once daily and crofelemer 125 mg delayed-release tablets given four times per day (QID). Fecal scores were collected twice daily using an established canine stool scoring scale called the Purina Fecal Scoring (PFS) System. After 28 days, using analysis of covariance (ANCOVA), dogs in the CTR group had a significantly higher average number of weekly loose/watery stools (PFS of 6 or 7) when compared to either crofelemer BID (8.71±2.2 vs. 5.96±2.2, p = 0.028) or crofelemer QID (8.70±2.2 vs. 5.74±2.2, p = 0.022) treatment groups. The average number of weekly loose/watery stools were not different between the crofelemer BID and QID treatment groups (p = 0.84). This study showed that crofelemer prophylaxis reduced the incidence/severity of neratinib-associated diarrhea in female beagle dogs without the need for any loperamide administration.
Collapse
Affiliation(s)
- Michael Guy
- Napo Pharmaceuticals, Inc., San Francisco, CA, United States of America
| | - Andre Teixeira
- Napo Pharmaceuticals, Inc., San Francisco, CA, United States of America
| | - Allison Shrier
- Napo Pharmaceuticals, Inc., San Francisco, CA, United States of America
| | - Carol Meschter
- Comparative Biosciences, Inc., Sunnyvale, CA, United States of America
| | | | - Pravin Chaturvedi
- Napo Pharmaceuticals, Inc., San Francisco, CA, United States of America
| |
Collapse
|
46
|
Rubin E, Shan KS, Dalal S, Vu DUD, Milillo-Naraine AM, Guaqueta D, Ergle A. Molecular Targeting of the Human Epidermal Growth Factor Receptor-2 (HER2) Genes across Various Cancers. Int J Mol Sci 2024; 25:1064. [PMID: 38256137 PMCID: PMC10816365 DOI: 10.3390/ijms25021064] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) belongs to the ErbB family, a group of four transmembrane glycoproteins with tyrosine kinase activity, all structurally related to epidermal growth factor receptor (EGFR). These tyrosine kinases are involved in the transmission of cellular signals controlling normal cell growth and differentiation. If this transmission goes awry, it can lead to dysregulated growth of the cell. HER2 specifically can be implicated in the pathogenesis of at least eight malignancies. HER2 positivity quickly became a well-characterized indicator of aggressiveness and poor prognosis, with high rates of disease progression and mortality. After realizing the implication of HER2, it first became investigated as a target for treatment in breast cancer, and later expanded to areas of research in other cancer types. To this day, the most therapeutic advancements of anti-HER2 therapy have been in breast cancer; however, there have been strong advancements made in the incorporation of anti-HER2 therapy in other cancer types as well. This comprehensive review dissects HER2 to its core, incorporating the most up to date information. The topics touched upon are discussed in detail and up to 200 published sources from the most highly recognized journals have been integrated. The importance of knowing about HER2 is exemplified by the groundbreaking advancements that have been made, and the change in treatment plans it has brought to the oncological world in the last twenty years. Since its groundbreaking discovery there have been significant breakthroughs in knowledge regarding the actual receptor, the receptors biology, its mechanism of action, and advancements in tests to detect HER2 and significant strides on how to best incorporate targeted treatment. Due to the success of this field thus far, the review concludes by discussing the future of novel anti-HER2 therapy currently in development that everyone should be aware of.
Collapse
Affiliation(s)
- Elizabeth Rubin
- Memorial Cancer Institute, Pembroke Pines, FL 33028, USA; (K.S.S.); (S.D.); (D.U.D.V.); (A.M.M.-N.); (D.G.); (A.E.)
| | | | | | | | | | | | | |
Collapse
|
47
|
Quiroga D, Wesolowski R, Zelinskas S, Pinette A, Benner B, Schwarz E, Savardekar H, Johnson C, Stiff A, Yu L, Macrae E, Lustberg M, Mrozek E, Ramaswamy B, Carson WE. An Open-Label Study of Subcutaneous CpG Oligodeoxynucleotide (PF03512676) in Combination with Trastuzumab in Patients with Metastatic HER2+ Breast Cancer. Cancer Control 2024; 31:10732748241250189. [PMID: 38797949 PMCID: PMC11129578 DOI: 10.1177/10732748241250189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 03/18/2024] [Accepted: 04/10/2024] [Indexed: 05/29/2024] Open
Abstract
OBJECTIVES CpG ODN is a Toll-like receptor 9 agonist with immunotherapeutic potential for many cancer types, including aggressive breast cancers. There is strong interest in utilizing CpG ODN as an adjuvant to improve clinical efficacy of current treatments and immunogenicity of breast cancers not traditionally responsive to active immunotherapy, such as those that are human epidermal growth factor receptor 2 (HER2)-positive. This study aimed to study the efficacy and safety of combination CpG ODN plus anti-HER2 antibody trastuzumab treatment in patients with advanced/metastatic breast cancer. METHODS This single-arm, open-label phase II clinical trial treated patients (n = 6) with advanced/metastatic HER2-positive breast cancer with weekly subcutaneous CpG ODN and trastuzumab. Patients may have received any number of prior therapies to be enrolled (most enrolled at median 1 prior line of chemotherapy). Peripheral blood was collected at baseline and weeks 2, 6, 12, and 18 for immune analyses. Six patients were enrolled and 50% achieved stable disease (SD) response. RESULTS Median PFS was 8.3 months. Three of the six patients enrolled opted to stop treatment due to tolerability issues. Multiplex assay for cytokine measurements revealed significantly higher VEGF-D levels at week 2 compared to baseline. Peripheral blood mononuclear cells analyzed by flow cytometry showed a significant increase in monocytic MDSC between weeks 6 and 12. Patients with progressive disease tended to have higher levels of week 6 monocytic MDSC and PD-1+ T cells than patients with SD. NK cell populations did not significantly change throughout treatment. CONCLUSIONS CpG ODN and trastuzumab treatment of metastatic HER2 + breast cancer was safe but was not tolerable for all patients. This combination did induce potentially predictive immune profile changes in treated patients with metastatic HER2 + breast cancer, the significance of which needs to be further explored.
Collapse
Affiliation(s)
- Dionisia Quiroga
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Robert Wesolowski
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Sara Zelinskas
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ashley Pinette
- Department of Surgery, The Ohio State University, Columbus, OH, USA
- Miami Valley Hospital, Dayton, OH, USA
| | - Brooke Benner
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Emily Schwarz
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Himanshu Savardekar
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Courtney Johnson
- Department of Surgery, The Ohio State University, Columbus, OH, USA
- Miami Valley Hospital, Dayton, OH, USA
| | - Andrew Stiff
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Erin Macrae
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
- Columbus Oncology Associates, Columbus, OH, USA
| | - Maryam Lustberg
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
- Yale School of Medicine, New Haven, CN, USA
| | - Ewa Mrozek
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
- St. Rita’s Cancer Center, Lima, OH, USA
| | - Bhuvaneswari Ramaswamy
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - William E. Carson
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
48
|
Hajjo R, Sabbah DA, Bardaweel SK, Zhong HA. Targeting the EGFR/RAS/RAF signaling pathway in anticancer research: a recent update on inhibitor design and clinical trials (2020-2023). Expert Opin Ther Pat 2024; 34:51-69. [PMID: 38450537 DOI: 10.1080/13543776.2024.2327307] [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: 11/01/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024]
Abstract
INTRODUCTION Recent years have seen significant strides in drug developmenttargeting the EGFR/RAS/RAF signaling pathway which is critical forcell growth and proliferation. Protein-protein interaction networksamong EGFR, RAS, and RAF proteins offer insights for drug discovery. This review discusses the drug design and development efforts ofinhibitors targeting these proteins over the past 3 years, detailingtheir structures, selectivity, efficacy, and combination therapy.Strategies to combat drug resistance and minimize toxicities areexplored, along with future research directions. AREA COVERED This review encompasses clinical trials and patents on EGFR, KRAS,and BRAF inhibitors from 2020 to 2023, including advancements indesign and synthesis of proteolysis targeting chimeras (PROTACs) forprotein degradation. EXPERT OPINION To tackle drug resistance, designing allosteric fourth-generationEGFR inhibitors is vital. Covalent, allosteric, or combinationaltherapies, along with PROTAC degraders, are key methods to addressresistance and toxicity in KRAS and BRAF inhibitors.
Collapse
Affiliation(s)
- Rima Hajjo
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Board Member, National Center for Epidemics and Communicable Disease Control (JCDC), Amman, Jordan
| | - Dima A Sabbah
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Sanaa K Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman, Jordan
| | - Haizhen A Zhong
- DSC 309, Department of Chemistry, The University of Nebraska at Omaha, Omaha, NE, USA
| |
Collapse
|
49
|
Heater NK, Somayaji K, Gradishar W. Treatment of residual disease following neoadjuvant therapy in breast cancer. J Surg Oncol 2024; 129:18-25. [PMID: 37990834 DOI: 10.1002/jso.27523] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/23/2023]
Abstract
Substantial advances have been made in the systemic treatment of breast cancer with residual disease following neoadjuvant therapy. We reviewed recent and ongoing studies informing the standard clinical management of residual disease by subtype: HER2+, TNBC, and HR+/HER2-, as well as strategies for BRCA+ disease. We conclude with a discussion of ongoing clinical trials and current controversies regarding the treatment of residual disease in breast cancer.
Collapse
Affiliation(s)
- Natalie K Heater
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, Illinois, USA
| | - Khyati Somayaji
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, Illinois, USA
| | - William Gradishar
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
50
|
Fernandes CL, Silva DJ, Mesquita A. Novel HER-2 Targeted Therapies in Breast Cancer. Cancers (Basel) 2023; 16:87. [PMID: 38201515 PMCID: PMC10778064 DOI: 10.3390/cancers16010087] [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: 11/18/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Human epidermal growth factor 2 (HER-2)-positive breast cancer represents 15-20% of all breast cancer subtypes and has an aggressive biological behavior with worse prognosis. The development of HER-2-targeted therapies has changed the disease's course, having a direct impact on survival rates and quality of life. Drug development of HER-2-targeting therapies is a prolific field, with numerous new therapeutic strategies showing survival benefits and gaining regulatory approval in recent years. Furthermore, the acknowledgement of the survival impact of HER-2-directed therapies on HER-2-low breast cancer has contributed even more to advances in the field. The present review aims to summarize the newly approved therapeutic strategies for HER-2-positive breast cancer and review the new and exploratory HER-2-targeted therapies currently under development.
Collapse
Affiliation(s)
- Catarina Lopes Fernandes
- Medical Oncology Department, Pedro Hispano Hospital, 4464-513 Matosinhos, Portugal; (D.J.S.); (A.M.)
| | - Diogo J. Silva
- Medical Oncology Department, Pedro Hispano Hospital, 4464-513 Matosinhos, Portugal; (D.J.S.); (A.M.)
| | - Alexandra Mesquita
- Medical Oncology Department, Pedro Hispano Hospital, 4464-513 Matosinhos, Portugal; (D.J.S.); (A.M.)
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|