1
|
Nierenberg TC, Thomas SM, Halliday I, Botty van den Bruele A, Chiba A, Modell Parrish KJ, Woriax HE, DiNome ML, Westbrook KE, Plichta JK. Survival outcomes after pathologic complete response with neoadjuvant endocrine therapy vs. neoadjuvant chemotherapy: a retrospective national database study. Breast Cancer Res Treat 2025; 212:161-172. [PMID: 40349259 DOI: 10.1007/s10549-025-07717-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Neoadjuvant therapies can result in pathologic complete response (pCR) in patients with breast cancer, which can be predictive of long-term outcomes. Patients with estrogen receptor positive (ER +) tumors may receive either neoadjuvant chemotherapy (NAC) or neoadjuvant endocrine therapy (NET). We sought to compare survival outcomes in those with non-metastatic ER + breast cancer who received NET or NAC and achieved pCR. METHODS All patients diagnosed with ER + /HER2- stage I-III breast cancer, who received neoadjuvant systemic therapy followed by surgery, and achieved pCR, were selected from the National Cancer Database (NCDB, 2010-2021). The Kaplan-Meier method was used to estimate overall survival (OS), and log-rank tests were used to test for differences in OS. Cox Proportional Hazards models were used to estimate the association of NAC vs NET with OS, after adjustment for covariates. RESULTS 3313 patients met eligibility criteria: 3148 received NAC and 165 NET. The median follow-up for the entire cohort was 82 months (95% CI 80.4-83.1). Patients who received NAC were significantly younger (median age: NAC 49y vs NET 64y; p < 0.001), more likely to have a comorbidity score of 0 (NAC 89.3% vs NET 81.2%, p = 0.004), and more likely to have private insurance (NAC 68.9% vs NET 44.2%, p < 0.001). There were no significant differences between the NAC and NET patients based on race and ethnicity, income, education, or community type (all p > 0.05). The NAC treated patients were more likely to have larger tumors [median tumor size (IQR): NAC 3 cm (2.0-4.3) vs NET 1.3 cm (0.7-2.8); p < 0.001)], ductal histology (NAC 92.6% vs 81.2%, p < 0.001), and grade 3 tumors (NAC 70.2% vs 10.3%, p < 0.001). In the unadjusted Kaplan-Meier analysis, there was no significant difference in OS between NAC vs NET [5-year OS: NAC 0.935 vs NET 0.916; p = 0.08]. After adjustment for demographics, disease characteristics, and treatments, there remained no association between OS and study group (NAC vs NET; p = 0.63). CONCLUSIONS Patients with ER + /HER2- early-stage breast cancer who achieved pCR had similar OS, regardless of whether they received NAC or NET. As such, pCR appears to have similar prognostic value irrespective of the type of systemic therapy used to obtain this favorable outcome.
Collapse
Affiliation(s)
- Tori C Nierenberg
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Samantha M Thomas
- Department of Biostatistics & Bioinformatics, Duke University, Durham, NC, USA
- Duke Cancer Institute, Durham, NC, USA
| | - Ian Halliday
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Astrid Botty van den Bruele
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
- Duke Cancer Institute, Durham, NC, USA
| | - Akiko Chiba
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
- Duke Cancer Institute, Durham, NC, USA
| | - Kendra J Modell Parrish
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
- Duke Cancer Institute, Durham, NC, USA
| | - Hannah E Woriax
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
- Duke Cancer Institute, Durham, NC, USA
| | - Maggie L DiNome
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
- Duke Cancer Institute, Durham, NC, USA
| | - Kelly E Westbrook
- Duke Cancer Institute, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Jennifer K Plichta
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA.
- Duke Cancer Institute, Durham, NC, USA.
- Department of Population Health Sciences, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
2
|
Beck AS, Earley M, Troxell M, Tsai J, Telli ML, Wapnir IL. Duration and effect of neoadjuvant endocrine therapy on invasive tumor cellularity in hormone receptor-positive breast cancer. Breast Cancer Res Treat 2025:10.1007/s10549-025-07722-6. [PMID: 40372533 DOI: 10.1007/s10549-025-07722-6] [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: 01/09/2025] [Accepted: 05/05/2025] [Indexed: 05/16/2025]
Abstract
PURPOSE Neoadjuvant chemotherapy has been used to evaluate tumor response and downstage hormone sensitive localized breast cancers. However, complete pathological responses are uncommon. Neoadjuvant endocrine therapy (NET) has been used sparingly for the treatment of hormone receptor (HR)-positive breast cancers and frequently for six months or less. There is no clear definition of response to NET nor well-defined parameters for duration of treatment. METHODS A retrospective chart review of patients with HR-positive localized invasive breast cancers treated with NET for at least 2 months was performed at a single institution. Clinical features, drug selection, duration of therapy, type of surgery as well as pathological characteristics, and residual tumor cellularity were analyzed. A multivariable linear regression model was used to examine the association between NET length and residual invasive tumor cellularity. RESULTS 104 evaluable HR-positive invasive breast cancers were treated with NET over an 11-year period. Median age was 69 (range: 31-89) and 88% were post-menopausal. The median duration of treatment was 8 months, with 61% having at least 7 months. Patients receiving NET for 7 months or longer had a significantly lower residual cellularity (20.4%) compared to those treated 2-6 months (34.9%) (p = 0.006). Both pre-treatment and post-treatment Ki-67 ≤ 10% were associated with a lower residual invasive tumor cellularity. Residual invasive cellularity was not associated with menopausal status or NET agent. CONCLUSION Longer NET duration is associated with greater tumor response. Mean residual invasive tumor cellularity was 42% lower among patients receiving 7 or more months of NET.
Collapse
Affiliation(s)
- Amanda Sutherland Beck
- Department of Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Michelle Earley
- Department of Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Megan Troxell
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Jacqueline Tsai
- Department of Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Melinda L Telli
- Department of Medicine, Stanford University School of Medicine, 900 Welch Road, Stanford, CA, 94305, USA
| | - Irene L Wapnir
- Department of Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA.
| |
Collapse
|
3
|
López-Velazco JI, Manzano S, Elorriaga K, Otaño M, Lahuerta A, Álvarez L, Etxabe I, Huarte M, Buch E, Gimenez J, Quiroga V, Fernandez M, Aragón S, Paré L, Prat A, Álvarez-López I, Caffarel MM, Urruticoechea A. Molecular characterisation of the residual disease after neoadjuvant endocrine therapy in ER+/HER2- breast cancer uncovers biomarkers of tumour response. Transl Oncol 2025; 57:102407. [PMID: 40349505 DOI: 10.1016/j.tranon.2025.102407] [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: 04/09/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Neoadjuvant endocrine therapy (NET) in oestrogen receptor-positive /HER2-negative breast cancer (ER+/HER2- BC) allows real-time evaluation of treatment sensitivity by monitoring tumour response and offers the opportunity of personalised therapy. However, the lack of reproducible biomarkers to assess response and long-term prognosis after NET is a significant barrier to increase its indications. METHODS In this study we searched for clinically relevant molecular reporters of response to NET in a multicentre population of ER+/HER2- BC patients (n = 87) by using: PAM50 gene expression panel and immunohistochemical evaluation of key proteins involved in tumorigenesis. RESULTS Our PAM50 analyses show that tumours changing from luminal A to normal-like subtype after NET presented better radiological and pathological tumour responses, a significant larger decrease in Ki67 at surgery, lower preoperative endocrine prognostic index score (PEPI) and lower tumour cellularity size (TCS) than those with persistent luminal A status. Patients with the highest response to NET showed the largest decrease in PAM50-derived risk of recurrence (ROR) following NET. In addition, the percentage of p53 positive cells was associated with decreased response to NET. CONCLUSIONS Our findings highlight the change of intrinsic subtype from luminal A to normal-like after NET as a putative biomarker characterising the patient population that obtains the highest benefit from NET. Our study also suggests that changes in PAM50-derived ROR score and p53 evaluation could also help to identify those patients. Thus, this study uncovers potential biomarkers of response to NET and prognosis, which should be validated in independent cohorts, helping to the implementation of NET in the clinical practice.
Collapse
Affiliation(s)
- Joanna I López-Velazco
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain
| | - Sara Manzano
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain
| | - Kepa Elorriaga
- Gipuzkoa Pathology Unit, OSI Donostialdea - Onkologikoa, San Sebastián, Spain
| | - Maria Otaño
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gipuzkoa Cancer Unit/OSI Donostialdea - Onkologikoa, San Sebastián, Spain
| | - Ainhara Lahuerta
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gipuzkoa Cancer Unit/OSI Donostialdea - Onkologikoa, San Sebastián, Spain
| | - Luis Álvarez
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gynecology and General Surgery Departments - Breast Unit, Onkologikoa, San Sebastián, Spain
| | - Inge Etxabe
- Gynecology and General Surgery Departments - Breast Unit, Onkologikoa, San Sebastián, Spain
| | - Miren Huarte
- Gynecology and General Surgery Departments - Breast Unit, Onkologikoa, San Sebastián, Spain
| | - Elvira Buch
- Hospital Clínico Universitario de Valencia, Spain
| | | | | | - Marta Fernandez
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gynecology Department - Breast Unit, OSI Donostialdea, San Sebastián, Spain
| | | | - Laia Paré
- Hospital Clinic, Barcelona - IDIBAPS, Barcelona, Spain
| | - Aleix Prat
- Hospital Clinic, Barcelona - IDIBAPS, Barcelona, Spain
| | - Isabel Álvarez-López
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gipuzkoa Cancer Unit/OSI Donostialdea - Onkologikoa, San Sebastián, Spain
| | - Maria M Caffarel
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| | - Ander Urruticoechea
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gipuzkoa Cancer Unit/OSI Donostialdea - Onkologikoa, San Sebastián, Spain.
| |
Collapse
|
4
|
Wang H, Zhang G, Gao W, Li S, Yang S, Zhang X, Wang J, Li C, Gao S, Geng C. Dalpiciclib plus aromatase inhibitor versus neoadjuvant chemotherapy for ER-positive, HER2-negative breast cancer. Front Oncol 2025; 15:1566146. [PMID: 40371225 PMCID: PMC12074949 DOI: 10.3389/fonc.2025.1566146] [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: 01/24/2025] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Background The combination of cyclin-dependent kinases 4 and 6 inhibitors (CDK4/6i) with endocrine therapy (ET) has emerged as an effective alternative to neoadjuvant chemotherapy (NCT) for patients with hormone receptor-positive (HR+) and human epidermal growth factor receptor 2 (HER2) -negative breast cancer (BC). This single-center study retrospectively evaluated the efficacy and safety of dalpiciclib combined with an aromatase inhibitor (AI) compared to NCT. Methods The clinicopathological data and treatment details of patients with HR+ HER2 negative BC who underwent either neoadjuvant endocrine therapy (NET) or NCT were collected from the Fourth Hospital of Hebei Medical University. The primary endpoint of the study was the Residual Cancer Burden (RCB), while secondary endpoints included breast pathological complete response (bpCR), clinical response rates (ORR), proliferation markers, and safety profiles. Results Between May 2022 and June 2023, a total of 36 patients were treated with dalpiciclib plus AI, while 137 patients received NCT for the final analysis. Prior to propensity score matching (PSM), the rates of RCB 0 were 0% in the NET group and 7.3% in the NCT group (p=0.205). The rates of bpCR were 2.8% for the NET group and 13.1% for the NCT group (p=0.142). The ORR was comparable between the two groups (p=0.397), as were the rates of BCS (p=0.608). Both treatment groups demonstrated significant reductions in Ki-67 levels, although the extent of reduction was similar (p=0.174). Notably, a Ki-67 level of ≤ 10% post-treatment was more prevalent in the NET group (p<0.0001). Only two patients in the NET group achieved a Preoperative Endocrine Prognostic Index (PEPI) 0 score. The incidence of grade 3-4 toxicities was significantly higher in the NCT group compared to the NET group (p<0.05). Following PSM, patients treated with dalpiciclib plus AI exhibited comparable clinical responses and a safety advantage relative to those receiving NCT. Conclusion This study indicates that the combination of dalpiciclib and AI elicits comparable responses and demonstrates improved safety profiles when compared to NCT in patients with HR+ HER2 negative breast tumors. Furthermore, RCB and pCR may not serve as optimal endpoints for evaluating the efficacy of CDK4/6i-based NET.
Collapse
Affiliation(s)
- Haoqi Wang
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Guoyu Zhang
- Department of Breast Surgery, Handan Central Hospital, Handan, Hebei, China
| | - Wei Gao
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sainan Li
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shan Yang
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xi Zhang
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiaxing Wang
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chunxiao Li
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shan Gao
- Gland Surgery, The Hebei Province People’s Hospital, Shijiazhuang, Hebei, China
| | - Cuizhi Geng
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
5
|
Yarlagadda S, de Oliveira Andrade M, Nanda R. CDK4/6 inhibition in early and advanced hormone receptor-positive, HER2-negative breast cancer. Expert Rev Anticancer Ther 2025:1-12. [PMID: 40285528 DOI: 10.1080/14737140.2025.2498994] [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: 02/07/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibition in combination with endocrine therapy is the mainstay of treatment for hormone receptor-positive, HER2-negative (HR + /HER2-) advanced breast cancer; the CDK4/6 inhibitors abemaciclib and ribociclib are also approved for high-risk, early-stage, HR + /HER2- breast cancer. Numerous studies exploring CDK4/6 inhibitors in the early-stage setting are ongoing, as well as many more exploring novel combinations in the metastatic setting. AREAS COVERED Here, we review the basis of CDK4/6 inhibition for HR +/HER2- breast cancer, the pivotal clinical trials which led to regulatory approval, and ongoing trials evaluating novel combinations to further improve outcomes for those with both early and advanced HR+/HER2- breast cancer. Current literature was reviewed by a comprehensive search of PubMed MEDLINE (1/1/2000-12/31/2024). EXPERT OPINION CDK4/6 inhibitors are integral in the management of both advanced and high-risk, early-stage HR + /HER2- breast cancer. Biomarkers predictive of CDK 4/6 inhibitor (CDK4/6i) benefit remain elusive, and clinical and pathological features remain key to identifying those who are candidates for CDK4/6 inhibition in the early-stage setting. Numerous trials evaluating the role of a CDK4/6i with novel endocrine therapy partners and other targeted agents are ongoing, with the goal of improving outcomes for those with HR + /HER2- disease.
Collapse
Affiliation(s)
- Sudha Yarlagadda
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| | | | - Rita Nanda
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, USA
| |
Collapse
|
6
|
Salahi‐Niri A, Zarand P, Shojaeian F, Mansouri N, Yazdani O, Esbati R, Safavi‐Naini SAA, Jahanbin B. Proliferative Markers in Breast Cancer and Chemotherapy Implications: A Comprehensive Review. Health Sci Rep 2025; 8:e70626. [PMID: 40201702 PMCID: PMC11976874 DOI: 10.1002/hsr2.70626] [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: 05/07/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 04/10/2025] Open
Abstract
Background and Aims Breast cancer is the most common cancer and a leading cause of cancer-related death among women globally. Determining which patients will benefit from chemotherapy remains challenging. Proliferative markers such as Ki-67, mini chromosome maintenance (MCM) proteins, and proliferating cell nuclear antigen (PCNA) offer valuable insights into tumor growth and treatment response. This review evaluates their clinical roles, with a focus on chemotherapy implications and emerging digital pathology techniques for marker quantification. Methods A narrative review was conducted by searching PubMed, Scopus, and Google Scholar for studies related to Ki-67, MCM, PCNA, breast cancer, and chemotherapy. Studies were thematically categorized into five areas. A bibliometric analysis of publications from 2000 to April 2023 was performed using the Bibliometrix R package and VOSviewer to assess research trends and thematic evolution. Results Eighty studies were included in the narrative synthesis. Ki-67 is the most commonly used marker, particularly useful in predicting response to neoadjuvant chemotherapy (NAC). MCM proteins show promise for identifying proliferative potential across tumor grades, while PCNA is associated with aggressive tumor features and poor prognosis. Post-chemotherapy changes in Ki-67 levels are linked to survival outcomes. Bibliometric analysis revealed a shift in research focus from basic mechanisms to clinical applications and digital quantification. Conclusion Proliferative markers play an essential role in breast cancer management. Ki-67 remains a key predictor of chemotherapy response, while MCM and PCNA offer complementary prognostic insights. Integration of these markers with digital pathology and AI-driven tools may enhance diagnostic accuracy and personalized treatment strategies. Standardization of assessment methods is crucial for broader clinical application.
Collapse
Affiliation(s)
- Aryan Salahi‐Niri
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Paniz Zarand
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Fatemeh Shojaeian
- Sidney Kimmel Comprehensive Cancer Research CenterJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Negar Mansouri
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Omid Yazdani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Romina Esbati
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Seyed Amir Ahmad Safavi‐Naini
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
- Division of Data‐Driven and Digital Medicine (D3M)Icahn School of Medicine at Mount SinaiNew YorkUSA
| | - Behnaz Jahanbin
- Cancer Institute, Pathology Department, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| |
Collapse
|
7
|
Berton Giachetti PPM, Carnevale Schianca A, Trapani D, Marra A, Toss A, Marchiò C, Dieci MV, Gentilini OD, Criscitiello C, Kalinsky K, Sparano JA, Curigliano G. Current controversies in the use of Oncotype DX in early breast cancer. Cancer Treat Rev 2025; 135:102887. [PMID: 40048856 DOI: 10.1016/j.ctrv.2025.102887] [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: 12/02/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 04/08/2025]
Abstract
Multigene prognostic genomic assays have become essential tools in the management of early breast cancer (BC), providing information that help in risk-stratification, to provide risk-adapted decision-making of adjuvant treatments. Clinical practice guidelines recommend refining the prognostic information provided by clinical and pathology features with the use of genomic tests, such as Oncotype DX®, to classify cancers into risk groups and inform adjuvant treatment strategies. However, the clinical value (i.e., prognostic and/or predictive) and applicability of these assays vary due to differences in the clinical setting, especially in those populations that were underrepresented in pivotal clinical trials. Oncotype DX® is a broadly utilized genomic test for breast cancer, having the highest level of supporting evidence to inform clinical practice. Our manuscript provides a comprehensive overview on this recurrence score assay, evaluates supporting evidence across patient populations, and discusses their impact on treatment decisions in those groups of patients underrepresented in pivotal clinical trials, where evidence is limited with the use of Oncotype DX.
Collapse
Affiliation(s)
- Pier Paolo M Berton Giachetti
- Division of New Drugs and Early Drug Development for Innovative Therapies European Institute of Oncology IRCCS Milano Italy; Department of Oncology and Hemato-Oncology University of Milano Milano Italy
| | - Ambra Carnevale Schianca
- Division of New Drugs and Early Drug Development for Innovative Therapies European Institute of Oncology IRCCS Milano Italy; Department of Oncology and Hemato-Oncology University of Milano Milano Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies European Institute of Oncology IRCCS Milano Italy; Department of Oncology and Hemato-Oncology University of Milano Milano Italy
| | - Antonio Marra
- Division of New Drugs and Early Drug Development for Innovative Therapies European Institute of Oncology IRCCS Milano Italy
| | - Angela Toss
- Department of Oncology and Hematology Azienda Ospedaliero-Universitaria di Modena Modena Italy; Department of Medical and Surgical Sciences University of Modena and Reggio Emilia Modena Italy
| | - Caterina Marchiò
- Division of Pathology Candiolo Cancer Institute FPO-IRCCS Candiolo Italy; Department of Medical Sciences University of Turin Turin Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2 35128 Padova, Italy; Oncology 2, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64 35128 Padova, Italy
| | - Oreste Davide Gentilini
- Breast Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies European Institute of Oncology IRCCS Milano Italy; Department of Oncology and Hemato-Oncology University of Milano Milano Italy
| | - Kevin Kalinsky
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Joseph A Sparano
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies European Institute of Oncology IRCCS Milano Italy; Department of Oncology and Hemato-Oncology University of Milano Milano Italy.
| |
Collapse
|
8
|
Vidal M, Falato C, Pascual T, Sanchez-Bayona R, Muñoz-Mateu M, Cebrecos I, Gonzalez-Farré X, Cortadellas T, Margelí Vila M, Luna MA, Siso C, Amillano K, Galván P, Bergamino MA, Ferrero-Cafiero JM, Salvador F, Espinosa Guerrero A, Pare L, Sanfeliu E, Prat A, Bellet M. Elacestrant in Women with Estrogen Receptor-Positive and HER2-Negative Early Breast Cancer: Results from the Preoperative Window-of-Opportunity ELIPSE Trial. Clin Cancer Res 2025; 31:1223-1232. [PMID: 39820652 PMCID: PMC11959270 DOI: 10.1158/1078-0432.ccr-24-2460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/04/2024] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE Elacestrant has shown significantly prolonged progression-free survival compared with standard-of-care endocrine therapy in estrogen receptor-positive (ER-positive), HER2-negative metastatic breast cancer, whereas potential benefit in early-stage disease requires further exploration. The SOLTI-ELIPSE window-of-opportunity trial investigated the biological changes induced by a short course of preoperative elacestrant in postmenopausal women with early breast cancer. PATIENTS AND METHODS Eligible patients with untreated T1c (≥1.5 cm)-T3, N0, ER-positive/HER2-negative breast cancer with locally assessed Ki67 ≥10% received elacestrant at a daily dose of 345 mg for 4 weeks. The primary efficacy endpoint was complete cell cycle arrest, defined as Ki67 ≤2.7%, on day 28. RESULTS Overall, 22 patients were evaluable for the primary endpoint. Elacestrant was associated with a complete cell cycle arrest rate of 27.3% and a statistically significant Ki67 geometric mean change of -52.9% (P = 0.007; 95% confidence interval, -67.4 to -32.1). Notably, the treatment with elacestrant led to a shift toward a more endocrine-sensitive and less proliferative tumor phenotype based on PAM50-based gene signatures. Elacestrant increased the expression of immune-response genes (GZMB, CD4, and CD8A) and suppressed proliferation and estrogen-signaling genes (MKI67, ESR1, and AR). These biological changes were independent of the levels of Ki67 suppression on day 28. The most common adverse events were grade 1 anemia (21.7%), hot flushes (8.7%), constipation (8.7%), and abdominal pain (8.7%). One patient experienced a grade 3 cutaneous rash, leading to treatment discontinuation. No other serious adverse events were reported. CONCLUSIONS Preoperative treatment with elacestrant in early breast cancer demonstrated relevant biological and molecular responses and exhibited a manageable safety profile. These findings support further investigation of elacestrant in the early setting.
Collapse
Affiliation(s)
- Maria Vidal
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Claudette Falato
- SOLTI Cancer Research Group, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Tomás Pascual
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Rodrigo Sanchez-Bayona
- SOLTI Cancer Research Group, Barcelona, Spain
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Montserrat Muñoz-Mateu
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Isaac Cebrecos
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Tomás Cortadellas
- Breast Unit, Department of Obstetrics and Gynaecology, Hospital Universitari General de Catalunya, Barcelona, Spain
| | - Mireia Margelí Vila
- SOLTI Cancer Research Group, Barcelona, Spain
- B-ARGO Group, Medical Oncology Department, ICO Badalona, Germans Trias I Pujol Institute, Badalona, Spain
- Medicine Department, Autonomous University, Barcelona, Spain
| | - Miguel A. Luna
- B-ARGO Group, Medical Oncology Department, ICO Badalona, Germans Trias I Pujol Institute, Badalona, Spain
| | | | - Kepa Amillano
- Hospital Universitari Sant Joan de Reus, Barcelona, Spain
| | - Patricia Galván
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Milana A. Bergamino
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- B-ARGO Group, Medical Oncology Department, ICO Badalona, Germans Trias I Pujol Institute, Badalona, Spain
| | | | | | | | - Laia Pare
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Esther Sanfeliu
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Pathology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Aleix Prat
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Meritxell Bellet
- SOLTI Cancer Research Group, Barcelona, Spain
- Medicine Department, Autonomous University, Barcelona, Spain
- Vall d’Hebron University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
9
|
Solaimani M, Hosseinzadeh S, Abasi M. Non-coding RNAs, a double-edged sword in breast cancer prognosis. Cancer Cell Int 2025; 25:123. [PMID: 40170036 PMCID: PMC11959806 DOI: 10.1186/s12935-025-03679-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
Cancer is a rising issue worldwide, and numerous studies have focused on understanding the underlying reasons for its occurrence and finding proper ways to defeat it. By applying technological advances, researchers are continuously uncovering and updating treatments in cancer therapy. Their vast functions in the regulation of cell growth and proliferation and their significant role in the progression of diseases, including cancer. This review provides a comprehensive analysis of ncRNAs in breast cancer, focusing on long non-coding RNAs such as HOTAIR, MALAT1, and NEAT1, as well as microRNAs such as miR-21, miR-221/222, and miR-155. These ncRNAs are pivotal in regulating cell proliferation, metastasis, drug resistance, and apoptosis. Additionally, we discuss experimental approaches that are useful for studying them and highlight the advantages and challenges of each method. We then explain the results of these clinical trials and offer insights for future studies by discussing major existing gaps. On the basis of an extensive number of studies, this review provides valuable insights into the potential of ncRNAs in cancer therapy. Key findings show that even though the functions of ncRNAs are vast and undeniable in cancer, there are still complications associated with their therapeutic use. Moreover, there is an absence of sufficient experiments regarding their application in mouse models, which is an area to work on. By emphasizing the crucial role of ncRNAs, this review underscores the need for innovative approaches and further studies to explore their potential in cancer therapy.
Collapse
Affiliation(s)
- Maryam Solaimani
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Sahar Hosseinzadeh
- Faculty of Pharmacy and Medical Biotechnology, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mozhgan Abasi
- Immunogenetics Research Center, Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, PO Box: 48175/861, Sari, Iran.
| |
Collapse
|
10
|
Danzinger S, Spornberger VH, Vietzen H, Tendl-Schulz K, Pfeiler G, Singer CF, Seifert M. Influence of histopathological changes after neoadjuvant chemotherapy on the survival of breast cancer patients. Cancer Treat Res Commun 2025; 43:100886. [PMID: 40031096 DOI: 10.1016/j.ctarc.2025.100886] [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: 07/30/2024] [Revised: 02/04/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Neoadjuvant chemotherapy (NACT) is an established form of therapy for early breast cancer (BC). The aim of our study was to analyze histopathological parameters before and after receiving NACT and to determine the influence of these changes on prognosis of BC patients. MATERIAL AND METHODS We retrospectively analyzed data of patients with primary early BC, diagnosed between January 2012 and December 2019, and NACT, followed by primary surgery. Patients achieving pathological complete response (pCR) were excluded. For the outcome analysis, disease-free survival (DFS) and overall survival (OS) were defined. RESULTS A total of 237 tumors were analyzed in the study. The conversion rates of tumor grade, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), Ki67 status, and BC subtype were 34.6 %, 3.4 %, 14.3 %, 4.6 %, 30.0 %, and 28.7 %, respectively. After a median follow-up of 58.03 months, we found an association between consistently negative ER/PR with the worst prognosis (DFS and OS) (ER p < 0.0001 for both; PR p = 0.0003, p = 0.0004, respectively). The conversion from Ki67 ≥14 % to <14 % led to an improved outcome compared to a constant Ki67 ≥14 % (DFS p = 0.003, OS p = 0.001). Tumor residuals with a non-triple-negative (nTN) subtype (TN → nTN) showed a better prognosis than those with TN subtype (nTN → TN) (DFS and OS p < 0.0001). CONCLUSIONS After NACT, tumor grade and Ki67 showed the highest conversion rates between primary biopsy and tumor residual. Depending on changes in ER, PR, Ki67, and subtype, we found significant differences in the prognosis of the patients.
Collapse
Affiliation(s)
- Sabine Danzinger
- Department of Obstetrics and Gynecology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Verena Heiss Spornberger
- Department of Obstetrics and Gynecology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Hannes Vietzen
- Center for Virology, Medical University of Vienna, Kinderspitalgasse 15, 1090 Vienna, Austria.
| | - Kristina Tendl-Schulz
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Georg Pfeiler
- Department of Obstetrics and Gynecology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Christian F Singer
- Department of Obstetrics and Gynecology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Michael Seifert
- Department of Obstetrics and Gynecology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
11
|
Marks JR, Zhang D, Hardman T, Chen YY, Hall A, Simpson L, Hieken T, Bedrosian I, Price E, Sheng J, Dai Y, Lee M, Sibley AB, Owzar K, Hwang ES. Genomic alterations are associated with response to aromatase inhibitor therapy for ER-positive postmenopausal ductal carcinoma in situ: (CALGB 40903, Alliance). Breast Cancer Res 2025; 27:26. [PMID: 39980051 PMCID: PMC11843815 DOI: 10.1186/s13058-025-01963-5] [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: 06/11/2024] [Accepted: 01/11/2025] [Indexed: 02/22/2025] Open
Abstract
PURPOSE CALGB 40903 (Alliance) was a phase II single arm multicenter trial conducted in postmenopausal patients diagnosed with estrogen-receptor (ER) positive breast ductal carcinoma in situ (DCIS) without invasion. Patients were treated with the aromatase inhibitor (AI) letrozole for 6 months prior to surgery with change in magnetic resonance imaging (MRI) enhancement volume compared to baseline as the primary endpoint. In the current study, we performed sequence analysis of pre- and post-treatment specimens to determine gene expression and DNA copy number parameters associated with treatment and response. EXPERIMENTAL DESIGN Paraffin sections from pretreatment biopsies and post-treatment surgical specimens were evaluated for presence of DCIS. Proliferation based on KI67 staining was quantified by a study pathologist. Macrodissection of the DCIS components from thin sections was the source of RNA and DNA. Whole-transcriptome RNA and shallow whole-genome DNA sequencing were performed. PAM50 analysis to assign intrinsic subtypes with associated probability of class membership was performed. Differential gene expression comparing responders versus non-responders and pre- versus post-treatment specimens was performed using a two-tiered approach based on candidate genes and a whole genome survey with appropriate multiple testing corrections. RESULTS Based on availability of specimens and presence of DCIS component, 29 patients (from the 70 who completed the treatment trial) were included in the final data set, including five who had a pathologic complete response (pCR). Response to treatment was qualified categorically based on a threshold of 10% KI67 in the post-treatment surgical specimen or pCR. Based on this criterion, six of the 29 DCIS were considered non-responders (> 10% KI67) and five subjects with pCR were assigned to the responder group. No standard clinical variables were associated with response. On the basis of gene expression analysis, 19 of the pre-treatment samples were classified as luminal A, all of which were classified as responders. PAM50 classification of the other ten pre-treatment samples included luminal B, HER2, basal, and normal-like, six of which were non-responders. PAM50 class membership shifted from baseline to post-treatment in eight cases, most often from luminal A to normal-like (five cases). Selected genes associated with estrogen receptor levels in invasive breast cancer were higher in AI responsive tumors. AI treatment resulted in reductions in estrogen and proliferation related genes. CONCLUSIONS Letrozole treatment produced an effective growth response, particularly in DCIS initially classified as luminal A. Study inclusion criteria of DCIS with at least 1% ER positive cells resulted in the inclusion of other subtypes that failed to respond. Treatment also induced both minor and major changes in intrinsic subtype based on PAM50 probabilities. Overall, these data indicate that response to AI treatment in ER( +) DCIS is variable and analogous to that observed in invasive breast cancers. TRANSLATIONAL RELEVANCE Treatment for breast DCIS ranges from active surveillance to mastectomy, often combined with adjuvant endocrine therapy. The work presented here based on a unique neoadjuvant trial provides direct information on hormone therapy responsiveness of this disease and further couples the biology of invasive breast cancer to its non-obligate precursor. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01439711.
Collapse
MESH Headings
- Humans
- Female
- Aromatase Inhibitors/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Postmenopause
- Carcinoma, Intraductal, Noninfiltrating/drug therapy
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Middle Aged
- Aged
- Receptors, Estrogen/metabolism
- Letrozole
- Treatment Outcome
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Profiling
- Biomarkers, Tumor
- DNA Copy Number Variations
- Genomics
Collapse
Affiliation(s)
- Jeffrey R Marks
- Department of Surgery, Duke University School of Medicine, 465 Seeley Mudd Building, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Dadong Zhang
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Timothy Hardman
- Department of Surgery, Duke University School of Medicine, 465 Seeley Mudd Building, Durham, NC, 27710, USA
| | - Yunn-Yi Chen
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Allison Hall
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lunden Simpson
- Department of Surgery, Duke University School of Medicine, 465 Seeley Mudd Building, Durham, NC, 27710, USA
| | | | | | - Elissa Price
- University of California at San Francisco, San Francisco, CA, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - Jeff Sheng
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Yanwan Dai
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Marissa Lee
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | | | - Kouros Owzar
- Duke Cancer Institute, Duke University, Durham, NC, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University School of Medicine, 465 Seeley Mudd Building, Durham, NC, 27710, USA.
- Duke Cancer Institute, Duke University, Durham, NC, USA.
| |
Collapse
|
12
|
Blanco R, Muñoz JP. Human Cytomegalovirus Infection and Breast Cancer: A Literature Review of Clinical and Experimental Data. BIOLOGY 2025; 14:174. [PMID: 40001942 PMCID: PMC11851556 DOI: 10.3390/biology14020174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 02/27/2025]
Abstract
Breast cancer (BC) remains a significant global health challenge, highlighting the need for continued research into novel risk factors, diagnostic approaches, and personalized treatments. Among emerging risk factors, viral infections have been implicated as potential contributors to breast carcinogenesis and BC progression. Recent evidence suggests that specific oncogenic strains of human cytomegalovirus (HCMV) may have the capacity to transform human mammary epithelial cells. This review assesses clinical data regarding HCMV presence in both tumor and non-tumor breast tissues, examining the role of HCMV oncoproteins in BC development and progression. Current findings indicate a higher prevalence of HCMV infection in breast carcinomas compared to non-tumor tissues, associated with an elevated risk of BC. Additionally, the HCMV-driven breast carcinogenesis model proposed here suggests that HCMV oncoproteins may activate multiple oncogenic pathways, fostering cell proliferation, survival, and tumor development. A deeper understanding of the role of HCMV in BC could enhance risk stratification and support the creation of targeted therapeutic strategies.
Collapse
Affiliation(s)
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| |
Collapse
|
13
|
Klocker EV, Egle D, Bartsch R, Rinnerthaler G, Gnant M. Efficacy and Safety of CDK4/6 Inhibitors: A Focus on HR+/HER2- Early Breast Cancer. Drugs 2025; 85:149-169. [PMID: 39820840 PMCID: PMC11802638 DOI: 10.1007/s40265-024-02144-y] [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] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) have revolutionized the treatment of hormone-receptor positive (HR+), HER2 negative (HER2-) metastatic breast cancer, and are now also established agents in the treatment of high-risk and intermediate-risk HR+ early breast cancer. Several strategies regarding CDK4/6i combinations or continuation beyond progression have been successfully evaluated in the metastatic setting, and are considered a standard of care. Mechanism of action of and resistance mechanisms against CDK4/6i in addition to endocrine resistance represent an important research topic, important for the treatment of HR+ breast cancer. Clinically, CDK4/6i are efficient substances that are usually well tolerated. However, side effects differing between the substances have been reported, and might lead to treatment discontinuation, including in the early disease setting. In the adjuvant setting, the addition of palbociclib to standard endocrine treatment has not improved outcomes, whereas large randomized phase III trials have demonstrated significant disease-free survival benefit for the addition of ribociclib (NATALEE trial) and abemaciclib (monarchE trial). Patient selection, treatment duration, endocrine backbone therapy, and other study details differ between these pivotal trials. This review focuses on both the scientific background as well as all available clinical data of CDK4/6i, with particular emphasis on their use in early breast cancer.
Collapse
Affiliation(s)
- Eva Valentina Klocker
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria
| | - Daniel Egle
- Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria
- Department of Gynecology, Breast Cancer Center Tirol, Medical University of Innsbruck, Innsbruck, Austria
| | - Rupert Bartsch
- Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Gabriel Rinnerthaler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria
| | - Michael Gnant
- Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
14
|
Rios-Hoyo A, Shan NL, Karn PL, Pusztai L. Clinical Implications of Breast Cancer Intrinsic Subtypes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:435-448. [PMID: 39821037 DOI: 10.1007/978-3-031-70875-6_21] [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
Estrogen receptor-positive (ER+) and estrogen receptor-negative (ER-) breast cancers have different genomic architecture and show large-scale gene expression differences consistent with different cellular origins, which is reflected in the luminal (i.e., ER+) versus basal-like (i.e., ER-) molecular class nomenclature. These two major molecular subtypes have distinct epidemiological risk factors and different clinical behaviors. Luminal cancers can be subdivided further based on proliferative activity and ER signaling. Those with a high expression of proliferation-related genes and a low expression of ER-associated genes, called luminal B, have a high risk of early recurrence (i.e., within 5 years), derive significant benefit from adjuvant chemotherapy, and may benefit from adding immunotherapy to chemotherapy. This subset of luminal cancers is identified as the genomic high-risk ER+ cancers by the MammaPrint, Oncotype DX Recurrence Score, EndoPredict, Prosigna, and several other molecular prognostic assays. Luminal A cancers are characterized by low proliferation and high ER-related gene expression. They tend to have excellent prognosis with adjuvant endocrine therapy. Adjuvant chemotherapy may not improve their outcome further. These cancers correspond to the genomic low-risk categories. However, these cancers remain at risk for distant recurrence for extended periods of time, and over 50% of distant recurrences occur after 5 years. Basal-like cancers are uniformly highly proliferative and tend to recur within 3-5 years of diagnosis. In the absence of therapy, basal-like breast cancers have the worst survival, but these also include many highly chemotherapy-sensitive cancers. Basal-like cancers are often treated with preoperative chemotherapy combined with an immune checkpoint inhibitor which results in 60-65% pathologic complete response rates that herald excellent long-term survival. Patients with residual cancer after neoadjuvant therapy can receive additional postoperative chemotherapy that improves their survival. Currently, there is no clinically actionable molecular subclassification for basal-like cancers, although cancers with high androgen receptor (AR)-related gene expression and those with high levels of immune infiltration have better prognosis, but currently their treatment is not different from basal-like cancers in general. A clinically important, minor subset of breast cancers are characterized by frequent HER2 gene amplification and high expression of a few dozen genes, many residing on the HER2 amplicon. This is an important subset because of the highly effective HER2 targeted therapies which are synergistic with endocrine therapy and chemotherapy. The clinical behavior of HER2-enriched cancers is dominated by the underlying ER subtype. ER+/HER2-enriched cancers tend to have more indolent course and lesser chemotherapy sensitivity than their ER counterparts.
Collapse
Affiliation(s)
| | - Naing-Lin Shan
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | | | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
15
|
Miras I, Gil A, Benavent M, Castilla MÁ, Vieites B, Dominguez-Cejudo MÁ, Molina-Pinelo S, Alfaro L, Frutos J, Ruiz-Borrego M, Falcón A, Cejuela M, Salvador-Bofill J. Predictive factors for complete pathologic response in luminal breast cancer: impact of ki67 and HER2 low expression. Ther Adv Med Oncol 2024; 16:17588359241309169. [PMID: 39734711 PMCID: PMC11672595 DOI: 10.1177/17588359241309169] [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: 08/12/2024] [Accepted: 12/06/2024] [Indexed: 12/31/2024] Open
Abstract
Background Complete pathological response to neoadjuvant treatment (NAT) in breast cancer is associated with prolonged survival. Compared to other breast cancer immunophenotypes, luminal tumors are the least chemosensitive with low rates of pathological response within this molecular subtype. Thus, finding predictors of response in this subset remains challenging. The emerging concept of low human epidermal growth factor receptor 2 (HER2) expression has led to a repurpose of the current prognostic system. Little is known about its correlation with response to NAT. Objectives This study aims to evaluate predictors of response in early-stage luminal breast cancer receiving neoadjuvant chemotherapy. Design A total of 252 luminal patients who received NAT were retrospectively assessed in this cohort study. Methods We analyzed the correlation of ki67 and HER2 low expression with the rate of pathologic response. Using ki67 as a continuous variable and applying the receiver operating characteristic curves method. Results We identified that in patients with a ki67 expression level >37%, the probability of having a complete pathological response was 4.80 times higher (odds ratio = 4.80, 95% confidence interval: 1.92-12.04). In Her2-low breast cancer patients, Her2 expression did not correlate with a better response rate. Conclusion In our study, a ki67 expression value greater than 37% constitutes a predictive biomarker of pathological complete response in the subgroup of patients with luminal B tumors and could be considered, therefore, an indicator for treatment decisions in this subgroup.
Collapse
Affiliation(s)
- Isabel Miras
- Institute of Biomedicine of Seville, HUVR, CSIC, University of Seville, Seville, Spain
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Ana Gil
- Institute of Biomedicine of Seville, HUVR, CSIC, University of Seville, Seville, Spain
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Marta Benavent
- Institute of Biomedicine of Seville, HUVR, CSIC, University of Seville, Seville, Spain
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | - Begoña Vieites
- Institute of Biomedicine of Seville, HUVR, CSIC, University of Seville, Seville, Spain
- Pathology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | - Sonia Molina-Pinelo
- Institute of Biomedicine of Seville, HUVR, CSIC, University of Seville, Seville, Spain
| | - Lina Alfaro
- Gynaecology and Obstetrics Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Javier Frutos
- Radiology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Manuel Ruiz-Borrego
- Institute of Biomedicine of Seville, HUVR, CSIC, University of Seville, Seville, Spain
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Alejandro Falcón
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Mónica Cejuela
- Institute of Biomedicine of Seville, HUVR, CSIC, University of Seville, Seville, Spain
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Javier Salvador-Bofill
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot, S/n, Seville 41013, Spain
- Institute of Biomedicine of Seville, HUVR, CSIC, University of Seville, Seville, Spain
| |
Collapse
|
16
|
Colomer R, González-Farré B, Ballesteros AI, Peg V, Bermejo B, Pérez-Mies B, de la Cruz S, Rojo F, Pernas S, Palacios J. Biomarkers in breast cancer 2024: an updated consensus statement by the Spanish Society of Medical Oncology and the Spanish Society of Pathology. Clin Transl Oncol 2024; 26:2935-2951. [PMID: 38869741 PMCID: PMC11564209 DOI: 10.1007/s12094-024-03541-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/14/2024]
Abstract
This revised consensus statement of the Spanish Society of Medical Oncology (SEOM) and the Spanish Society of Pathological Anatomy (SEAP) updates the recommendations for biomarkers use in the diagnosis and treatment of breast cancer that we first published in 2018. The expert group recommends determining in early breast cancer the estrogen receptor (ER), progesterone receptor (PR), Ki-67, and Human Epidermal growth factor Receptor 2 (HER2), as well as BReast CAncer (BRCA) genes in high-risk HER2-negative breast cancer, to assist prognosis and help in indicating the therapeutic options, including hormone therapy, chemotherapy, anti-HER2 therapy, and other targeted therapies. One of the four available genetic prognostic platforms (Oncotype DX®, MammaPrint®, Prosigna®, or EndoPredict®) may be used in ER-positive patients with early breast cancer to establish a prognostic category and help decide with the patient whether adjuvant treatment may be limited to hormonal therapy. In second-line advanced breast cancer, in addition, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) and estrogen receptor 1 (ESR1) should be tested in hormone-sensitive cases, BRCA gene mutations in HER2-negative cancers, and in triple-negative breast cancer (TNBC), programmed cell death-1 ligand (PD-L1). Newer biomarkers and technologies, including tumor-infiltrating lymphocytes (TILs), homologous recombination deficiency (HRD) testing, serine/threonine kinase (AKT) pathway activation, and next-generation sequencing (NGS), are at this point investigational.
Collapse
Affiliation(s)
- Ramon Colomer
- UAM Personalised Precision Medicine Chair & Medical Oncology Department, La Princesa University Hospital and Research Institute, C/Diego de León, 62, 28006, Madrid, Spain.
| | | | | | - Vicente Peg
- Pathological Anatomy Service, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Begoña Bermejo
- Medical Oncology Department, Biomedical Research Institute INCLIVA, Medicine Department of the University of Valencia and Clinic University Hospital, Valencia, Spain
| | - Belén Pérez-Mies
- Pathological Anatomy Service, Ramón y Cajal University Hospital, Faculty of Medicine, University of Alcalá, IRYCIS and CIBERONC, Madrid, Spain
| | - Susana de la Cruz
- Medical Oncology Department, Navarra University Hospital, Navarre, Spain
| | - Federico Rojo
- Anatomy Service, Fundación Jiménez Díaz University Hospital and CIBERONC, Madrid, Spain
| | - Sonia Pernas
- Oncology Department, Catalan Institute of Oncology (ICO)-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - José Palacios
- Pathological Anatomy Service, Department of Pathology, Ramón y Cajal University Hospital, Faculty of Medicine, University of Alcalá, IRYCIS and CIBERONC, Ctra. Colmenar Viejo, Km 9,1, 28034, Madrid, Spain.
| |
Collapse
|
17
|
Han X, Li H, Zhou SY, Dong SS, Zhang GL. Clinical efficacy of combined goserelin and anastrozole in neoadjuvant endocrine therapy for premenopausal women with hormone receptor-positive breast cancer. Discov Oncol 2024; 15:554. [PMID: 39397134 PMCID: PMC11471739 DOI: 10.1007/s12672-024-01418-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024] Open
Abstract
OBJECTIVE The objective of this study is to assess the efficacy and safety of combining goserelin with anastrozole in neoadjuvant endocrine therapy (NET) for patients diagnosed with premenopausal breast cancer. METHODS A retrospective analysis was conducted on the clinicopathological data of 34 patients diagnosed with premenopausal breast cancer who underwent NET in the Department of Breast Surgery at Baotou Cancer Hospital between March 2016 and December 2019. Additionally, the feasibility of using goserelin combined with anastrozole for premenopausal endocrine therapy was assessed. RESULTS The duration of NET ranged from 6 to 72 months, with a mean of 22.5 months and a median of 18 months. In patients with progressive disease, endocrine therapy was assessed over a period of 6 to 18 months, with a mean of 13.1 months and a median of 13 months. Among the 28 patients assessed, 12 (42.86%) were found to have stable disease, subsequently receiving chemotherapy. Of these, seven patients demonstrated good compliance, and 5 achieved a pathological complete response. Including the 2 patients who responded favorably to NET alone, a total of 7 patients attained a pathological complete response. Additionally, 16 patients achieved complete cell cycle arrest following treatment. A significant correlation was observed between the clinical efficacy assessment and the pathological assessment of NET (P < 0.05). CONCLUSION Although NET was safe for patients diagnosed with premenopausal breast cancer, it should not be considered in isolation from chemotherapy. Transitioning to chemotherapy in a timely manner can significantly enhance treatment outcomes. The duration of NET should be guided by clinical assessment rather than being constrained by a predetermined time frame.
Collapse
Affiliation(s)
- Xu Han
- Department of Breast Surgery, Baotou Cancer Hospital of Inner Mongolia, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Hui Li
- Department of Breast Surgery, Baotou Cancer Hospital of Inner Mongolia, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Shui-Ying Zhou
- Department of Breast Surgery, Baotou Cancer Hospital of Inner Mongolia, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Sha-Sha Dong
- Department of Breast Surgery, Baotou Cancer Hospital of Inner Mongolia, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - Gang-Ling Zhang
- Department of Breast Surgery, Baotou Cancer Hospital of Inner Mongolia, No.18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China.
| |
Collapse
|
18
|
Khorfan R, Vora HP, Namm JP, Solomon NL, Lum SS. Expanded Indications for Neoadjuvant Endocrine Therapy in Early-Stage Breast Cancer During the COVID-19 Pandemic. Ann Surg Oncol 2024; 31:7562-7568. [PMID: 39133445 PMCID: PMC11452450 DOI: 10.1245/s10434-024-15787-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/24/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND In response to the COVID-19 pandemic, the Pandemic Breast Cancer Consortium (PBCC) published recommendations for triage of breast cancer patients. The recommendations included neoadjuvant treatment of early-stage breast cancer patients experiencing delays in surgery. This study evaluated national patterns of neoadjuvant treatment according to triage guidelines. METHODS Patients treated with surgery (upfront or post-neoadjuvant) in 2018-2020 were collected from the National Cancer Database. The proportions of patients treated according to the PBCC triage guidelines were calculated in 2020 and compared with similar cohorts in 2018-2019. Patient and hospital factors were evaluated for association with treatment. RESULTS Among cT1N0 ER+/PR+/HER2- patients, those treated in 2020 were more likely to receive neoadjuvant endocrine therapy (NET) compared with those before that time (odds ratio [OR], 3.08; range, 2.93-3.24). Among the patients with cT2N0 or cT1N1 disease, NET was more common in 2020 (OR, 1.76; range, 1.65-1.88). Academic facility, black or Asian race, more comorbidities, and the New England/Middle Atlantic region were associated with NET use. CONCLUSIONS During the COVID-19 pandemic, expanded utilization of neoadjuvant therapy for surgical breast cancer patients was observed. Health care system limitations during the pandemic contributed to expanded adoption of neoadjuvant therapy for early breast cancer, contrary to usual practice. Long-term outcomes for patients treated according to PBCC recommendations should be closely monitored.
Collapse
Affiliation(s)
- Rhami Khorfan
- Surgical Oncology Division, Department of Surgery, Loma Linda University Health, Loma Linda, CA, USA
| | - Halley P Vora
- Surgical Oncology Division, Department of Surgery, Loma Linda University Health, Loma Linda, CA, USA
| | - Jukes P Namm
- Surgical Oncology Division, Department of Surgery, Loma Linda University Health, Loma Linda, CA, USA
| | - Naveenraj L Solomon
- Surgical Oncology Division, Department of Surgery, Loma Linda University Health, Loma Linda, CA, USA
| | - Sharon S Lum
- Surgical Oncology Division, Department of Surgery, Loma Linda University Health, Loma Linda, CA, USA.
| |
Collapse
|
19
|
Bischoff H, Espié M, Petit T. Neoadjuvant Therapy: Current Landscape and Future Horizons for ER-Positive/HER2-Negative and Triple-Negative Early Breast Cancer. Curr Treat Options Oncol 2024; 25:1210-1224. [PMID: 39145854 PMCID: PMC11416407 DOI: 10.1007/s11864-024-01251-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 08/16/2024]
Abstract
OPINION STATEMENT Navigating the complex landscape of breast cancer treatment involves distinct strategies for luminal and triple-negative subtypes. While neoadjuvant chemotherapy historically dominates the approach for aggressive triple-negative tumors, recent evidence highlights the transformative impact of immunotherapy, alongside chemotherapy, in reshaping treatment paradigms. In luminal cancers, endocrine therapy, notably aromatase inhibitors, demonstrates promising outcomes in postmenopausal patients with low-grade luminal A tumors. However, integrating targeted therapies like CDK4/6 inhibitors in neoadjuvant setting remains inconclusive. Identifying predictive factors for treatment response, especially in luminal tumors, poses a challenge, emphasizing the necessity for ongoing research. A multidisciplinary approach, tailored to individual patient profiles, is crucial for maximizing efficacy while minimizing toxicity. As we strive to optimize breast cancer management, a comprehensive understanding of the distinct characteristics and treatment implications of luminal and triple-negative subtypes, including the transformative role of immunotherapy, is essential for informed decision-making and personalized care.
Collapse
Affiliation(s)
- Hervé Bischoff
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, 67033, Strasbourg, France.
| | - Marc Espié
- Medical Oncology Department, Hôpital Saint Louis, Paris, France
| | - Thierry Petit
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, ICANS, 17 Rue Albert Calmette, 67033, Strasbourg, France
| |
Collapse
|
20
|
Miligy IM, Awasthi R, Mir Y, Khurana A, Sharma V, Chandaran U, Rakha E, Maurice Y, Kearns D, Oweis R, Asar A, Ironside A, Shaaban AM. Morphological and molecular changes of oestrogen receptor-positive breast cancer following bridging endocrine therapy: a United Kingdom multicentre study. Histopathology 2024; 85:405-417. [PMID: 38845397 DOI: 10.1111/his.15238] [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: 01/15/2024] [Revised: 05/21/2024] [Accepted: 05/25/2024] [Indexed: 08/09/2024]
Abstract
AIMS Standard neoadjuvant endocrine therapy (NAET) is used for 6-9 months to downstage hormone-receptor-positive breast cancer. Bridging ET was introduced during the COVID-19 pandemic to delay surgical intervention. There are no data in the literature on the effect of short course therapy on tumour response. We aimed to analyse the effect of bridging ET and validate the previously proposed neoadjuvant ET pathological reporting criteria. METHODS AND RESULTS This was a multicentre cohort of 256 patients who received bridging ET between March and October 2020. Assessment of paired pre- and post-NAET hormone receptors and HER2 and posttherapy Ki67 expression was done. The median duration of NAET was 45 days. In all, 86% of cases achieved partial pathological response and 9% showed minimal residual disease. Histological response to ET was observed from as early as day 6 posttherapy. Central scarring was noted in 32.8% of cases and lymphocytic infiltrate was seen in 43.4% of cases. Significant changes associated with the duration of ET were observed in tumour grade (21%), with downgrading identified in 12% of tumours (P < 0.001), progesterone receptor (PR) expression with switch to PR-negative status in 26% of cases (P < 0.001), and HER2 status with a switch from HER2-low to HER2-negative status in 32% of cases (P < 0.001). The median patient survival was 475 days, with an overall survival rate of 99.6%. CONCLUSIONS Changes characteristic of tumour regression and significant changes in PR and HER2 occurred following a short course of NAET. The findings support biomarker testing on pretreatment core biopsies and retesting following therapy.
Collapse
Affiliation(s)
- Islam M Miligy
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham, UK
- Histopathology Department, Menoufia University, Shebin El Kom, Egypt
| | - Rachna Awasthi
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham, UK
| | - Yasmeen Mir
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Anuj Khurana
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Vijay Sharma
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Usha Chandaran
- Histopathology Department, Salford Royal Hospital, Salford, UK
| | - Emad Rakha
- Histopathology Department, Nottingham City Hospital, Nottingham, UK
| | - Yasmine Maurice
- Histopathology Department, Heartlands General Hospital, Birmingham, UK
| | - Daniel Kearns
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham, UK
| | - Rami Oweis
- Histopathology Department, Rotherham Foundation Trust, Rotherham, UK
| | - Amal Asar
- Histopathology Department, Rotherham Foundation Trust, Rotherham, UK
| | | | - Abeer M Shaaban
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
21
|
Schettini F, Saracchini S, Bassini A, Marus W, Corsetti S, Specogna I, Bertola M, Micheli E, Wirtz RM, Laible M, Şahin U, Strina C, Milani M, Aguggini S, Tancredi R, Fiorio E, Sulfaro S, Generali D. Prediction of response to neoadjuvant chemotherapy by MammaTyper® across breast cancer subtypes: A retrospective cross-sectional study. Breast 2024; 76:103753. [PMID: 38815444 PMCID: PMC11166895 DOI: 10.1016/j.breast.2024.103753] [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: 10/18/2023] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT) is widely used in the treatment of triple-negative and HER2-positive breast cancer (BC), but its use in estrogen receptor (ER) and/or progesterone receptor (PR) positive/HER2-negative BC is questioned because of the low pathologic complete response (pCR) rates. This retrospective study assessed the mRNA-based MammaTyper® assay's capability of predicting pCR with NACT, and ER, PR, Ki67, and HER2 status at immunohistochemical (IHC) through transcriptomics. METHODS Diagnostic biopsies from 76 BC patients treated at the Cremona Hospital between 2012-2018 were analyzed. Relative mRNA expression levels of ERBB2, ESR1, PGR, and MKI67 were measured using the MammaTyper® kit and integrated into a pCR score. Predicting capability of pCR and standard IHC biomarkers could be assessed with ROC curves in 75 and 76 patients, respectively. RESULTS Overall, 68.0% patients obtained a MammaTyper® high-score and 32.0% a MammaTyper® low-score. Among high-score patients, 62.7% achieved pCR, compared to 16.7% in the low-score group (p = 0.0003). The binary MammaTyper® score showed good prediction of pCR in the overall cohort (area under curve [AUC] = 0.756) and in HR+/HER2-negative cases (AUC = 0.774). In cases with residual disease, the continuous MammaTyper® score correlated moderately with residual tumor size and decrease in tumor size. MammaTyper® showed substantial agreement with IHC for ESR1/ER and ERBB2/HER2, and moderate agreement for PGR/PR and MKI67/Ki67. CONCLUSION Overall, MammaTyper® pCR score may serve as a standardized tool for predicting NACT response in HR+/HER2-negative BC, potentially guiding treatment strategies. Additionally, it could provide a more standardized and reproducible assessment of ER, PR, HER2, and Ki67 status.
Collapse
MESH Headings
- Humans
- Female
- Neoadjuvant Therapy
- Retrospective Studies
- Middle Aged
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/analysis
- Adult
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/analysis
- Cross-Sectional Studies
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/analysis
- Aged
- Chemotherapy, Adjuvant
- Ki-67 Antigen/analysis
- Ki-67 Antigen/metabolism
- Immunohistochemistry
- Predictive Value of Tests
- Treatment Outcome
- RNA, Messenger/analysis
- RNA, Messenger/metabolism
- ROC Curve
Collapse
Affiliation(s)
- Francesco Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain.
| | | | - Anna Bassini
- Azienda per l'Assistenza Sanitaria 5 Friuli Occidentale, "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - Wally Marus
- Azienda per l'Assistenza Sanitaria 5 Friuli Occidentale, "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | | | - Ilaria Specogna
- Azienda per l'Assistenza Sanitaria 5 Friuli Occidentale, "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | | | - Elvia Micheli
- Azienda per l'Assistenza Sanitaria 5 Friuli Occidentale, "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - Ralph M Wirtz
- STRATIFYER Molecular Pathology GmbH, Cologne, Germany
| | | | | | - Carla Strina
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Manuela Milani
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Sergio Aguggini
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Richard Tancredi
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Elena Fiorio
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, 37134 Verona, Italy
| | - Sandro Sulfaro
- Azienda per l'Assistenza Sanitaria 5 Friuli Occidentale, "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - Daniele Generali
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.
| |
Collapse
|
22
|
Fjermeros K, Ghannoum S, Geisler SB, Bhargava S, Tahiri A, Klajic J, Lüders T, Fongård M, Nawaz MS, Bosnjak-Olsen T, Buvarp UCE, Rosenskiold AKJ, Nguyen NT, Sletbak TT, Seyedzadeh M, Selsås K, Porojnicu AC, Skjerven HK, Hovda T, Sahlberg KK, Torland LA, Lyngra M, Hammarström CL, Hönigsperger EB, Noone JC, Mathiassen S, Hurtado A, Goel S, Koff A, Tekpli X, Kristensen VN, Geisler J. The NEOLETRIB trial: neoadjuvant treatment with Letrozole and Ribociclib in ER-positive, HER2-negative breast cancer. Future Oncol 2024; 20:2457-2466. [PMID: 39073142 PMCID: PMC11520546 DOI: 10.1080/14796694.2024.2377531] [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: 05/13/2024] [Accepted: 07/04/2024] [Indexed: 07/30/2024] Open
Abstract
Chemotherapy is used as neoadjuvant therapy for all subgroups of breast cancer, including ER-positive, and HER2-negative cases. However, studies have suggested that using aromatase inhibitors combined with CDK4/6-inhibitors might be an appropriate alternative in selected patients. Thus, the NEOLETRIB trial evaluates the response of ER-positive, HER2-negative luminal A/B breast cancer to the combination of letrozole and ribociclib in the neoadjuvant setting. Comprehensive molecular biology procedures, including sequential single-cell RNA-sequencing of tumor biopsies, are performed during 6 months of treatment with extensive biobanking of blood samples, tumor biopsies and gut microbiome specimens. Our findings will hopefully contribute to an improved selection of patients who may benefit from this drug combination and give new insights into the intra-tumoral changes during this treatment.Trial registration number: NCT05163106 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Kamilla Fjermeros
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Salim Ghannoum
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | | | - Sameer Bhargava
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
- Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Andliena Tahiri
- Department of Clinical Molecular Biology (EpiGen), Akershus University Hospital, Lørenskog, Norway
| | - Jovana Klajic
- Department of Clinical Molecular Biology (EpiGen), Akershus University Hospital, Lørenskog, Norway
| | - Torben Lüders
- Department of Clinical Molecular Biology (EpiGen), Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Marie Fongård
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Meh Sameen Nawaz
- Department of Clinical Molecular Biology (EpiGen), Akershus University Hospital, Lørenskog, Norway
- Department of Health and Exercise, School of Health Sciences, Kristiania University College, Oslo, Norway
| | | | | | | | - Nam Thi Nguyen
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | | | | | - Knut Selsås
- Department of Endocrine & Breast Surgery, Akershus University Hospital, Lørenskog, Norway
| | | | - Helle Kristine Skjerven
- Department of Breast & Endocrine Surgery, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Tone Hovda
- Department of Radiology, Drammen Hospital, Vestre Viken Hospital Trust, Norway
| | - Kristine Kleivi Sahlberg
- Department of Research & Innovation, Vestre Viken Hospital Trust, Drammen, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Lilly Anne Torland
- Department of Research & Innovation, Vestre Viken Hospital Trust, Drammen, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Marianne Lyngra
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | | | | | | | - Silje Mathiassen
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Antoni Hurtado
- Functional Genomics group & Molecular Pathology Unit, Centro de Investigación del Cáncer (CSIC-Universidad de Salamanca), Campus Universitario Miguel de Unamuno s/n. 37007, Salamanca, Spain
| | - Shom Goel
- Peter MacCallum Cancer Centre, Australia & The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Andrew Koff
- Program in Molecular Biology, Memorial Sloan Kettering Cancer Center & Weill Cornell Medical College, New York, NY USA
| | - Xavier Tekpli
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Vessela N. Kristensen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|
23
|
Miligy IM, Badr N, Stevens A, Spooner D, Awasthi R, Mir Y, Khurana A, Sharma V, Chandaran U, Rakha EA, Maurice Y, Kearns D, Oweis R, Asar A, Ironside A, Shaaban AM. Pathological Changes Following Neoadjuvant Endocrine Therapy (NAET): A Multicentre Study of 391 Breast Cancers. Int J Mol Sci 2024; 25:7381. [PMID: 39000487 PMCID: PMC11242101 DOI: 10.3390/ijms25137381] [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: 04/19/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Oestrogen receptor (ER)-positive breast cancer (BC) is generally well responsive to endocrine therapy. Neoadjuvant endocrine therapy (NAET) is increasingly being used for downstaging ER-positive tumours. This study aims to analyse the effect of NAET on a well-characterised cohort of ER-positive BC with particular emphasis on receptor expression. This is a retrospective United Kingdom (UK) multicentre study of 391 patients who received NAET between October 2012 and October 2020. Detailed analyses of the paired pre- and post-NAET morphological changes and hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2) expression were performed. The median duration of NAET was 86 days, with median survival and overall survival rates of 380 days and 93.4%, respectively. A total of 90.3% of cases achieved a pathological partial response, with a significantly higher rate of response in the HER2-low cancers. Following NAET, BC displayed some pathological changes involving the tumour stroma including central scarring and an increase in tumour infiltrating lymphocytes (TILs) and tumour cell morphology. Significant changes associated with the duration of NAET were observed in tumour grade (30.6% of cases), with downgrading identified in 19.3% of tumours (p < 0.001). The conversion of ER status from positive to low or negative was insignificant. The conversion of progesterone receptor (PR) and HER2 status to negative status was observed in 31.3% and 38.1% of cases, respectively (p < 0.001). HER2-low breast cancer decreased from 63% to 37% following NAET in the paired samples. Significant morphological and biomarker changes involving PR and HER2 expression occurred following NAET. The findings support biomarker testing on pre-treatment core biopsies and post-treatment residual carcinoma.
Collapse
Affiliation(s)
- Islam M. Miligy
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (I.M.M.); (R.A.); (D.K.)
- Histopathology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom 11352, Egypt;
| | - Nahla Badr
- Histopathology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom 11352, Egypt;
| | - Andrea Stevens
- Oncology Department, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (A.S.); (D.S.)
| | - David Spooner
- Oncology Department, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (A.S.); (D.S.)
| | - Rachna Awasthi
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (I.M.M.); (R.A.); (D.K.)
| | - Yasmeen Mir
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK; (Y.M.); (A.K.); (V.S.)
| | - Anuj Khurana
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK; (Y.M.); (A.K.); (V.S.)
| | - Vijay Sharma
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK; (Y.M.); (A.K.); (V.S.)
| | - Usha Chandaran
- Histopathology Department, Salford Royal Hospital, Salford M6 8HD, UK;
| | - Emad A. Rakha
- Histopathology Department, Nottingham City Hospital, Nottingham NG5 1PB, UK;
| | - Yasmine Maurice
- Histopathology Department, Heartlands General Hospital, Birmingham B9 5SS, UK;
| | - Daniel Kearns
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (I.M.M.); (R.A.); (D.K.)
| | - Rami Oweis
- Histopathology Department, Rotherham Foundation Trust, Rotherham S60 2UD, UK; (R.O.); (A.A.)
| | - Amal Asar
- Histopathology Department, Rotherham Foundation Trust, Rotherham S60 2UD, UK; (R.O.); (A.A.)
| | | | - Abeer M. Shaaban
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham B15 2GW, UK; (I.M.M.); (R.A.); (D.K.)
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK
| |
Collapse
|
24
|
Shimomura A, Sagara Y, Koto R, Fujiwara M, Kanemura Y, Kitagawa H, Saji S. Real-world data of HER2-negative early breast cancer patients treated with anthracycline and/or taxane regimens in Japan. Breast Cancer 2024; 31:581-592. [PMID: 38679657 PMCID: PMC11194198 DOI: 10.1007/s12282-024-01572-8] [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: 12/21/2023] [Accepted: 03/17/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Anthracycline- and taxane-based chemotherapy regimens are established treatments for human epidermal growth factor receptor (HER)2-negative early-stage breast cancer with high risk of recurrence. This study examined the prevalence of these chemotherapy regimens as perioperative therapy, the patterns of retreatment, and factors influencing prescription choices in Japan. METHODS This observational cohort study focused on high-risk early-stage breast cancer patients not undergoing anti-HER2 therapy, utilizing data from a hospital-based claims database in Japan spanning from April 2008 to September 2021. RESULTS Of 42,636 high-risk patients who underwent breast cancer surgery, 32,133 (75.4%) were categorized as having luminal-type (received endocrine therapy) and 10,503 (24.6%) as having triple-negative cancer (not receiving any endocrine therapies). Most patients (98.7%) with luminal-type breast cancer received perioperative therapy, and 40.3% of those received anthracycline/taxane. In the triple-negative group, 57.0% of all patients received perioperative therapy and of those, 93.4% received anthracycline/taxane. Being over 40 years old, having an early stage (clinical stage ≤ II), and receiving treatment in non-specialized facilities were associated with less use of anthracycline/taxane in the luminal-type group. For the triple-negative group, associated factors with less use of anthracycline/taxane included being over 60 years old, treatment in small hospital (capacity < 200 beds), and treatment in non-specialized facilities. CONCLUSIONS Approximately half the patients in both the luminal-type and triple-negative groups were prescribed anthracycline and/or taxane for perioperative chemotherapy. The choice was associated with patient age, cancer stage, and the scale and specialization of the treatment facilities. This study sheds light on the current state of breast cancer treatment practices in Japan.
Collapse
Affiliation(s)
- Akihiko Shimomura
- Department of Breast and Medical Oncology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yasuaki Sagara
- Department of Breast and Thyroid Surgical Oncology, Social Medical Corporation Hakuaikai Sagara Hospital, Kagoshima, Japan
| | - Ryo Koto
- Medical Department, AstraZeneca K.K., Osaka, Japan
| | | | | | | | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, 1 Hikariga-oka, Fukushima, Fukushima, 960-1295, Japan.
| |
Collapse
|
25
|
Zhao H, Li D, Li Q, Zhang B, Xiao C, Zhao Y, Ge J, Yu Y, Jia Y, Guo X, Cao X, Wang X. Tucidinostat Plus Exemestane as a Neoadjuvant in Early-Stage, Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Breast Cancer. Oncologist 2024; 29:e763-e770. [PMID: 38459836 PMCID: PMC11144976 DOI: 10.1093/oncolo/oyae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/06/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND To assess the efficacy and safety of tucidinostat plus exemestane as a neoadjuvant strategy in early-stage breast cancer. METHODS This prospective, open-label, single-arm phase II trial enrolled patients with stage II-III breast cancer with hormone receptor-positive and human epidermal growth factor receptor 2 (HER2)-negative. Eligible patients received tucidinostat plus exemestane, and then breast-conserving surgery (BCS) or modified radical mastectomy. RESULTS Among 20 enrolled patients, 3 of them achieved preoperative endocrine prognostic index (PEPI) score of 0. Additionally, complete cell cycle arrest was observed in 7, radiologic objective response rate in 10, and disease control rate in 20 patients, pathological complete response in 1 patient, and 5 patients performed BCS. Ki67 suppression from baseline to surgery was observed in 17 of patients, with the Ki67 change ratio of -73.5%. Treatment-emergent adverse event included neutropenia, leukopenia, thrombocytopenia, lymphopenia, hypoalbuminemia, aspartate aminotransferase elevation, glutamyl transpeptidase elevation, anemia, and alanine aminotransferase elevation. CONCLUSIONS Despite the rate of PEPI score 0 was not high, tucidinostat plus exemestane as a neoadjuvant therapy might be well tolerated and showed promising clinical responses in patients with early hormone receptor-positive, HER2-negative breast cancer. To clarify the safety and efficacy of this strategy, further investigation is warranted. CLINICAL TRIAL REGISTRATION ChiCTR2100046678.
Collapse
Affiliation(s)
- Hongmeng Zhao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Dan Li
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Qian Li
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Bin Zhang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Chunhua Xiao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Ying Zhao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Jie Ge
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Yue Yu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Yumian Jia
- The Department of Breast Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Xiaojing Guo
- The Department of Breast Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Xuchen Cao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| | - Xin Wang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, People’s Republic of China
| |
Collapse
|
26
|
Bossuyt V, Provenzano E, Symmans WF, Webster F, Allison KH, Dang C, Gobbi H, Kulka J, Lakhani SR, Moriya T, Quinn CM, Sapino A, Schnitt S, Sibbering DM, Slodkowska E, Yang W, Tan PH, Ellis I. A dedicated structured data set for reporting of invasive carcinoma of the breast in the setting of neoadjuvant therapy: recommendations from the International Collaboration on Cancer Reporting (ICCR). Histopathology 2024; 84:1111-1129. [PMID: 38443320 DOI: 10.1111/his.15165] [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: 12/21/2023] [Accepted: 02/11/2024] [Indexed: 03/07/2024]
Abstract
AIMS The International Collaboration on Cancer Reporting (ICCR), a global alliance of major (inter-)national pathology and cancer organisations, is an initiative aimed at providing a unified international approach to reporting cancer. ICCR recently published new data sets for the reporting of invasive breast carcinoma, surgically removed lymph nodes for breast tumours and ductal carcinoma in situ, variants of lobular carcinoma in situ and low-grade lesions. The data set in this paper addresses the neoadjuvant setting. The aim is to promote high-quality, standardised reporting of tumour response and residual disease after neoadjuvant treatment that can be used for subsequent management decisions for each patient. METHODS The ICCR convened expert panels of breast pathologists with a representative surgeon and oncologist to critically review and discuss current evidence. Feedback from the international public consultation was critical in the development of this data set. RESULTS The expert panel concluded that a dedicated data set was required for reporting of breast specimens post-neoadjuvant therapy with inclusion of data elements specific to the neoadjuvant setting as core or non-core elements. This data set proposes a practical approach for handling and reporting breast resection specimens following neoadjuvant therapy. The comments for each data element clarify terminology, discuss available evidence and highlight areas with limited evidence that need further study. This data set overlaps with, and should be used in conjunction with, the data sets for the reporting of invasive breast carcinoma and surgically removed lymph nodes from patients with breast tumours, as appropriate. Key issues specific to the neoadjuvant setting are included in this paper. The entire data set is freely available on the ICCR website. CONCLUSIONS High-quality, standardised reporting of tumour response and residual disease after neoadjuvant treatment are critical for subsequent management decisions for each patient.
Collapse
Affiliation(s)
- Veerle Bossuyt
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Provenzano
- Department of Histopathology, Addenbrookes Hospital, Cambridge, UK
| | - W Fraser Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fleur Webster
- International Collaboration on Cancer Reporting, Surry Hills, NSW, Australia
| | - Kimberly H Allison
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chau Dang
- Memorial Sloan Kettering Cancer Center, West Harrison, NY, USA
| | - Helenice Gobbi
- Department of Surgical Clinic, Federal University of Triangulo Mineiro, Uberaba, MG, Brazil
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Sunil R Lakhani
- Centre for Clinical Research, and Pathology Queensland, University of Queensland, Brisbane, Qld, Australia
| | - Takuya Moriya
- Department of Pathology, Kawasaki Medical School, Okayama, Japan
| | - Cecily M Quinn
- Department of Histopathology, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College, Dublin, Ireland
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Stuart Schnitt
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - D Mark Sibbering
- University Hospitals of Derby and Burton NHS Trust, Royal Derby Hospital, Derby, UK
| | - Elzbieta Slodkowska
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | | | - Ian Ellis
- Department of Histopathology, Nottingham City Hospital, London, UK
| |
Collapse
|
27
|
Yang H, Ruan Y, Sun Y, Wang P, Qiao J, Wang C, Liu Z. Assessment of the impact of residual tumors at different sites post-neoadjuvant chemotherapy on prognosis in breast cancer patients and development of a disease-free survival prediction model. Ther Adv Med Oncol 2024; 16:17588359241249578. [PMID: 38736552 PMCID: PMC11085027 DOI: 10.1177/17588359241249578] [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: 12/17/2023] [Accepted: 04/09/2024] [Indexed: 05/14/2024] Open
Abstract
Background Residual disease after neoadjuvant chemotherapy (NAC) in breast cancer patients predicts worse outcomes than pathological complete response. Differing prognostic impacts based on the anatomical site of residual tumors are not well studied. Objectives The study aims to assess disease-free survival (DFS) in breast cancer patients with different residual tumor sites following NAC and to develop a nomogram for predicting 1- to 3-year DFS in these patients. Design A retrospective cohort study. Methods Retrospective analysis of 953 lymph node-positive breast cancer patients with residual disease post-NAC. Patients were categorized into three groups: residual disease in breast (RDB), residual disease in lymph nodes (RDN), and residual disease in both (RDBN). DFS compared among groups. Patients were divided into a training set and a validation set in a 7:3 ratio. Prognostic factors for DFS were analyzed to develop a nomogram prediction model. Results RDB patients had superior 3-year DFS of 94.6% versus 85.2% for RDN and 81.8% for RDBN (p < 0.0001). Clinical T stage, N stage, molecular subtype, and postoperative pN stage were independently associated with DFS on both univariate and multivariate analyses. Nomogram integrating clinical tumor-node-metastasis (TNM) stage, molecular subtype, pathological response demonstrated good discrimination (C-index 0.748 training, 0.796 validation cohort), and calibration. Conclusion The location of residual disease has prognostic implications, with nodal residuals predicting poorer DFS. The validated nomogram enables personalized DFS prediction to guide treatment decisions.
Collapse
Affiliation(s)
- Hanzhao Yang
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Yuxia Ruan
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Yadong Sun
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Peili Wang
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jianghua Qiao
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Chengzheng Wang
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Zhenzhen Liu
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, No. 127, Dongming Road, Zhengzhou 450008, China
| |
Collapse
|
28
|
Camargo-Herrera V, Castellanos G, Rangel N, Jiménez-Tobón GA, Martínez-Agüero M, Rondón-Lagos M. Patterns of Chromosomal Instability and Clonal Heterogeneity in Luminal B Breast Cancer: A Pilot Study. Int J Mol Sci 2024; 25:4478. [PMID: 38674062 PMCID: PMC11049937 DOI: 10.3390/ijms25084478] [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: 01/26/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 04/28/2024] Open
Abstract
Chromosomal instability (CIN), defined by variations in the number or structure of chromosomes from cell to cell, is recognized as a distinctive characteristic of cancer associated with the ability of tumors to adapt to challenging environments. CIN has been recognized as a source of genetic variation that leads to clonal heterogeneity (CH). Recent findings suggest a potential association between CIN and CH with the prognosis of BC patients, particularly in tumors expressing the epidermal growth factor receptor 2 (HER2+). In fact, information on the role of CIN in other BC subtypes, including luminal B BC, is limited. Additionally, it remains unknown whether CIN in luminal B BC tumors, above a specific threshold, could have a detrimental effect on the growth of human tumors or whether low or intermediate CIN levels could be linked to a more favorable BC patient prognosis when contrasted with elevated levels. Clarifying these relationships could have a substantial impact on risk stratification and the development of future therapeutic strategies aimed at targeting CIN in BC. This study aimed to assess CIN and CH in tumor tissue samples from ten patients with luminal B BC and compare them with established clinicopathological parameters. The results of this study reveal that luminal B BC patients exhibit intermediate CIN and stable aneuploidy, both of which correlate with lymphovascular invasion. Our results also provide valuable preliminary data that could contribute to the understanding of the implications of CIN and CH in risk stratification and the development of future therapeutic strategies in BC.
Collapse
Affiliation(s)
- Valentina Camargo-Herrera
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia; (V.C.-H.).; (G.C.)
| | - Giovanny Castellanos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia; (V.C.-H.).; (G.C.)
| | - Nelson Rangel
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia;
| | - Guillermo Antonio Jiménez-Tobón
- Laboratorio de Patología, Hospital Universitario Mayor-Méderi, Bogotá 110311, Colombia;
- Grupo BIOmedUR, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 110231, Colombia
| | - María Martínez-Agüero
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 110231, Colombia
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia; (V.C.-H.).; (G.C.)
| |
Collapse
|
29
|
Conforti F, Pala L, De Pas T, Zattarin E, Catania C, Cocorocchio E, Rossi G, Laszlo D, Colleoni M, Zambelli A, Hortobagyi GN, Cortes J, Piccart MJ, Dowsett M, Gelber RD, Viale G. Fine-Tuning Adjuvant Endocrine Therapy for Early-Stage Breast Cancer: An Expert Consensus on Open Issues for Future Research. Clin Cancer Res 2024; 30:1093-1103. [PMID: 37906083 DOI: 10.1158/1078-0432.ccr-23-1836] [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: 06/19/2023] [Revised: 08/29/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
After decades of research, improving the efficacy of adjuvant endocrine therapy (ET) for early-stage breast cancer becomes increasingly difficult. Beyond technological breakthroughs and the availability of new classes of drugs, further improvement of adjuvant ET will require applying a rigorous research approach in poorly investigated areas. We critically discuss some key principles that should inform future research to improve ET efficacy, including identifying specific subgroups of patients who can benefit from escalating or de-escalating approaches, optimizing available and new treatment strategies for different clinical contexts, and dissecting the direct and indirect biological effects of therapeutic interventions. Four main issues regarding adjuvant ET were identified as relevant areas, where a better application of such principles can provide positive results in the near future: (i) tailoring the optimal duration of adjuvant ET, (ii) optimizing ovarian function suppression for premenopausal women, (iii) dissecting the biological effects of estrogen receptor manipulation, and (iv) refining the selection of patients to candidate for treatments escalation.
Collapse
Affiliation(s)
- Fabio Conforti
- Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Laura Pala
- Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Tommaso De Pas
- Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Emma Zattarin
- Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Chiara Catania
- Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Emilia Cocorocchio
- Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Giovanna Rossi
- Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Daniele Laszlo
- Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Marco Colleoni
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Alberto Zambelli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Gabriel N Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Javier Cortes
- International Breast Cancer Center, Pangaea Oncology, Quironsalud Group, Madrid and Barcelona, Spain
- Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| | - Martine J Piccart
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Mitch Dowsett
- Breast Unit, Royal Marsden Hospital, London, United Kingdom
| | - Richard D Gelber
- Department of Data Science, Division of Biostatistics, Dana-Farber Cancer Institute, Harvard Medical School, Harvard T.H. Chan School of Public Health, and Frontier Science Foundation, Boston, Massachusetts
| | - Giuseppe Viale
- Department of Pathology, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| |
Collapse
|
30
|
Hoshina H, Sakatani T, Kawamoto Y, Ohashi R, Takei H. Cytomorphological Disparities in Invasive Breast Cancer Cells following Neoadjuvant Endocrine Therapy and Chemotherapy. Pathobiology 2024; 91:288-298. [PMID: 38447546 PMCID: PMC11309077 DOI: 10.1159/000538227] [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] [Accepted: 03/04/2024] [Indexed: 03/08/2024] Open
Abstract
INTRODUCTION Neoadjuvant endocrine therapy (NAE) offers a breast-conserving surgery rate and clinical response rate similar to those of neoadjuvant chemotherapy (NAC), while presenting fewer adverse events and lower pathological complete response rates. The assessment of pathological response determines degenerative changes and predicts the prognosis of breast cancer treated with NAC. This study clarified the degenerative changes occurring in breast cancer following NAE. METHODS Our study encompassed two groups: NAE, consisting of 15 patients, and NAC, comprising 18 patients. Tissue samples were obtained from core needle biopsies and surgeries. Nuclear and cell areas were calculated using Autocell analysis. Furthermore, we assessed markers associated with microtubule depolymerization (KIF2A) and initiators of apoptosis (caspase-9). RESULTS In the NAC group, we observed significant increases in both cytoplasmic and cell areas. These changes in cytoplasm and cells were notably more pronounced in the NAC group compared to the NAE group. After treatment, KIF2A exhibited a decrease, with the magnitude of change being greater in the NET group than in the NAC group. However, no discernible differences were found in caspase-9 expression between the two groups. CONCLUSION Our findings indicate that NAE induces condensation in cancer cells via cell cycle arrest or apoptosis. Conversely, NAC leads to cell enlargement due to the absence of microtubule depolymerization. These discrepancies underscore the importance of accounting for these distinctions when establishing criteria for evaluating pathological responses.
Collapse
Affiliation(s)
- Hideko Hoshina
- Department of Breast Surgery and Oncology, Nippon Medical School, Tokyo, Japan,
| | - Takashi Sakatani
- Department of Diagnostic Pathology, Nippon Medical School Hospital, Tokyo, Japan
| | - Yoko Kawamoto
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Ryuji Ohashi
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Hiroyuki Takei
- Department of Breast Surgery and Oncology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
31
|
Kang D, Wang C, Han Z, Zheng L, Guo W, Fu F, Qiu L, Han X, He J, Li L, Chen J. Exploration of the relationship between tumor-infiltrating lymphocyte score and histological grade in breast cancer. BMC Cancer 2024; 24:318. [PMID: 38454386 PMCID: PMC10921807 DOI: 10.1186/s12885-024-12069-0] [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: 09/21/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND The histological grade is an important factor in the prognosis of invasive breast cancer and is vital to accurately identify the histological grade and reclassify of Grade2 status in breast cancer patients. METHODS In this study, data were collected from 556 invasive breast cancer patients, and then randomly divided into training cohort (n = 335) and validation cohort (n = 221). All patients were divided into actual low risk group (Grade1) and high risk group (Grade2/3) based on traditional histological grade, and tumor-infiltrating lymphocyte score (TILs-score) obtained from multiphoton images, and the TILs assessment method proposed by International Immuno-Oncology Biomarker Working Group (TILs-WG) were also used to differentiate between high risk group and low risk group of histological grade in patients with invasive breast cancer. Furthermore, TILs-score was used to reclassify Grade2 (G2) into G2 /Low risk and G2/High risk. The coefficients for each TILs in the training cohort were retrieved using ridge regression and TILs-score was created based on the coefficients of the three kinds of TILs. RESULTS Statistical analysis shows that TILs-score is significantly correlated with histological grade, and is an independent predictor of histological grade (odds ratio [OR], 2.548; 95%CI, 1.648-3.941; P < 0.0001), but TILs-WG is not an independent predictive factor for grade (P > 0.05 in the univariate analysis). Moreover, the risk of G2/High risk group is higher than that of G2/Low risk group, and the survival rate of patients with G2/Low risk is similar to that of Grade1, while the survival rate of patients with G2/High risk is even worse than that of patients with G3. CONCLUSION Our results suggest that TILs-score can be used to predict the histological grade of breast cancer and potentially to guide the therapeutic management of breast cancer patients.
Collapse
Affiliation(s)
- Deyong Kang
- Department of Pathology, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Chuan Wang
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Zhonghua Han
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Liqin Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China
| | - Wenhui Guo
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Fangmeng Fu
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Lida Qiu
- College of Physics and Electronic Information Engineering, Minjiang University, 350108, Fuzhou, P. R. China
| | - Xiahui Han
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China
| | - Jiajia He
- School of Science, Jimei University, 361021, Xiamen, P. R. China.
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China.
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China.
| |
Collapse
|
32
|
Ma CX, Suman VJ, Sanati S, Vij K, Anurag M, Leitch AM, Unzeitig GW, Hoog J, Fernandez-Martinez A, Fan C, Gibbs RA, Watson MA, Dockter TJ, Hahn O, Guenther JM, Caudle A, Crouch E, Tiersten A, Mita M, Razaq W, Hieken TJ, Wang Y, Rimawi MF, Weiss A, Winer EP, Hunt KK, Perou CM, Ellis MJ, Partridge AH, Carey LA. Endocrine-Sensitive Disease Rate in Postmenopausal Patients With Estrogen Receptor-Rich/ERBB2-Negative Breast Cancer Receiving Neoadjuvant Anastrozole, Fulvestrant, or Their Combination: A Phase 3 Randomized Clinical Trial. JAMA Oncol 2024; 10:362-371. [PMID: 38236590 PMCID: PMC10797521 DOI: 10.1001/jamaoncol.2023.6038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/29/2023] [Indexed: 01/19/2024]
Abstract
Importance Adding fulvestrant to anastrozole (A+F) improved survival in postmenopausal women with advanced estrogen receptor (ER)-positive/ERBB2 (formerly HER2)-negative breast cancer. However, the combination has not been tested in early-stage disease. Objective To determine whether neoadjuvant fulvestrant or A+F increases the rate of pathologic complete response or ypT1-2N0/N1mic/Ki67 2.7% or less residual disease (referred to as endocrine-sensitive disease) over anastrozole alone. Design, Setting, and Participants A phase 3 randomized clinical trial assessing differences in clinical and correlative outcomes between each of the fulvestrant-containing arms and the anastrozole arm. Postmenopausal women with clinical stage II to III, ER-rich (Allred score 6-8 or >66%)/ERBB2-negative breast cancer were included. All analyses were based on data frozen on March 2, 2023. Interventions Patients received anastrozole, fulvestrant, or a combination for 6 months preoperatively. Tumor Ki67 was assessed at week 4 and optionally at week 12, and if greater than 10% at either time point, the patient switched to neoadjuvant chemotherapy or immediate surgery. Main Outcomes and Measures The primary outcome was the endocrine-sensitive disease rate (ESDR). A secondary outcome was the percentage change in Ki67 after 4 weeks of neoadjuvant endocrine therapy (NET) (week 4 Ki67 suppression). Results Between February 2014 and November 2018, 1362 female patients (mean [SD] age, 65.0 [8.2] years) were enrolled. Among the 1298 evaluable patients, ESDRs were 18.7% (95% CI, 15.1%-22.7%), 22.8% (95% CI, 18.9%-27.1%), and 20.5% (95% CI, 16.8%-24.6%) with anastrozole, fulvestrant, and A+F, respectively. Compared to anastrozole, neither fulvestrant-containing regimen significantly improved ESDR or week 4 Ki67 suppression. The rate of week 4 or week 12 Ki67 greater than 10% was 25.1%, 24.2%, and 15.7% with anastrozole, fulvestrant, and A+F, respectively. Pathologic complete response/residual cancer burden class I occurred in 8 of 167 patients and 17 of 167 patients, respectively (15.0%; 95% CI, 9.9%-21.3%), after switching to neoadjuvant chemotherapy due to week 4 or week 12 Ki67 greater than 10%. PAM50 subtyping derived from RNA sequencing of baseline biopsies available for 753 patients (58%) identified 394 luminal A, 304 luminal B, and 55 nonluminal tumors. A+F led to a greater week 4 Ki67 suppression than anastrozole alone in luminal B tumors (median [IQR], -90.4% [-95.2 to -81.9%] vs -76.7% [-89.0 to -55.6%]; P < .001), but not luminal A tumors. Thirty-six nonluminal tumors (65.5%) had a week 4 or week 12 Ki67 greater than 10%. Conclusions and Relevance In this randomized clinical trial, neither fulvestrant nor A+F significantly improved the 6-month ESDR over anastrozole in ER-rich/ERBB2-negative breast cancer. Aromatase inhibition remains the standard-of-care NET. Differential NET response by PAM50 subtype in exploratory analyses warrants further investigation. Trial Registration ClinicalTrials.gov Identifier: NCT01953588.
Collapse
Affiliation(s)
- Cynthia X. Ma
- Washington University School of Medicine, St Louis, Missouri
| | - Vera J. Suman
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota
| | - Souzan Sanati
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Kiran Vij
- Washington University School of Medicine, St Louis, Missouri
| | | | | | | | - Jeremy Hoog
- Washington University School of Medicine, St Louis, Missouri
| | | | - Cheng Fan
- University of North Carolina at Chapel Hill
| | | | - Mark A. Watson
- Washington University School of Medicine, St Louis, Missouri
| | - Travis J. Dockter
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota
| | - Olwen Hahn
- University of Chicago, Chicago, Illinois
| | | | | | - Erika Crouch
- Washington University School of Medicine, St Louis, Missouri
| | | | - Monica Mita
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Wajeeha Razaq
- University of Oklahoma Health Sciences Center, Oklahoma City
| | | | - Yang Wang
- Presbyterian Kaseman Hospital, Albuquerque, New Mexico
| | | | - Anna Weiss
- University of Rochester, Rochester, New York
| | | | | | | | | | | | | |
Collapse
|
33
|
Stravodimou A, Voutsadakis IA. Neo-adjuvant therapies for ER positive/HER2 negative breast cancers: from chemotherapy to hormonal therapy, CDK inhibitors, and beyond. Expert Rev Anticancer Ther 2024; 24:117-135. [PMID: 38475990 DOI: 10.1080/14737140.2024.2330601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 02/02/2024] [Indexed: 03/14/2024]
Abstract
INTRODUCTION Chemotherapy has been traditionally used as neo-adjuvant therapy in breast cancer for down-staging of locally advanced disease in all sub-types. In the adjuvant setting, genomic assays have shown that a significant proportion of ER positive/HER2 negative patients do not derive benefit from the addition of chemotherapy to adjuvant endocrine therapy. An interest in hormonal treatments as neo-adjuvant therapies in ER positive/HER2 negative cancers has been borne by their documented success in the adjuvant setting. Moreover, cytotoxic chemotherapy is less effective in ER positive/HER2 negative disease compared with other breast cancer subtypes in obtaining pathologic complete responses. AREAS COVERED Neo-adjuvant therapies for ER positive/HER2 negative breast cancers and associated biomarkers are reviewed, using a Medline survey. A focus of discussion is the prediction of patients that are unlikely to derive extra benefit from chemotherapy and have the highest probabilities of benefiting from hormonal and other targeted therapies. EXPERT OPINION Predictive biomarkers of response to neo-adjuvant chemotherapy and hormonal therapies are instrumental for selecting ER positive/HER2 negative breast cancer patients for each treatment. Chemotherapy remains the standard of care for many of those patients requiring neo-adjuvant treatment, but other neo-adjuvant therapies are increasingly used.
Collapse
Affiliation(s)
- Athina Stravodimou
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Ioannis A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste Marie, Ontario, Canada
- Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| |
Collapse
|
34
|
Bacci M, Lorito N, Smiriglia A, Subbiani A, Bonechi F, Comito G, Morriset L, El Botty R, Benelli M, López-Velazco JI, Caffarel MM, Urruticoechea A, Sflomos G, Malorni L, Corsini M, Ippolito L, Giannoni E, Meattini I, Matafora V, Havas K, Bachi A, Chiarugi P, Marangoni E, Morandi A. Acetyl-CoA carboxylase 1 controls a lipid droplet-peroxisome axis and is a vulnerability of endocrine-resistant ER + breast cancer. Sci Transl Med 2024; 16:eadf9874. [PMID: 38416843 DOI: 10.1126/scitranslmed.adf9874] [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: 11/24/2022] [Accepted: 01/30/2024] [Indexed: 03/01/2024]
Abstract
Targeting aromatase deprives ER+ breast cancers of estrogens and is an effective therapeutic approach for these tumors. However, drug resistance is an unmet clinical need. Lipidomic analysis of long-term estrogen-deprived (LTED) ER+ breast cancer cells, a model of aromatase inhibitor resistance, revealed enhanced intracellular lipid storage. Functional metabolic analysis showed that lipid droplets together with peroxisomes, which we showed to be enriched and active in the LTED cells, controlled redox homeostasis and conferred metabolic adaptability to the resistant tumors. This reprogramming was controlled by acetyl-CoA-carboxylase-1 (ACC1), whose targeting selectively impaired LTED survival. However, the addition of branched- and very long-chain fatty acids reverted ACC1 inhibition, a process that was mediated by peroxisome function and redox homeostasis. The therapeutic relevance of these findings was validated in aromatase inhibitor-treated patient-derived samples. Last, targeting ACC1 reduced tumor growth of resistant patient-derived xenografts, thus identifying a targetable hub to combat the acquisition of estrogen independence in ER+ breast cancers.
Collapse
Affiliation(s)
- Marina Bacci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Nicla Lorito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Alfredo Smiriglia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Angela Subbiani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Francesca Bonechi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Ludivine Morriset
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France
| | - Rania El Botty
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France
| | - Matteo Benelli
- Department of Medical Oncology, Azienda USL Toscana Centro, Hospital of Prato, Via Suor Niccolina Infermiera 20, 59100 Prato, Italy
| | - Joanna I López-Velazco
- Biodonostia Health Research Institute, Paseo Dr Begiristain s/n, 20014 San Sebastian, Spain
| | - Maria M Caffarel
- Biodonostia Health Research Institute, Paseo Dr Begiristain s/n, 20014 San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Ander Urruticoechea
- Biodonostia Health Research Institute, Paseo Dr Begiristain s/n, 20014 San Sebastian, Spain
- Gipuzkoa Cancer Unit, OSI Donostialdea-Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014 San Sebastian, Spain
| | - George Sflomos
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Luca Malorni
- Department of Medical Oncology, Azienda USL Toscana Centro, Hospital of Prato, Via Suor Niccolina Infermiera 20, 59100 Prato, Italy
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, 25123 Brescia, Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Icro Meattini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Vittoria Matafora
- IFOM ETS-AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Kristina Havas
- IFOM ETS-AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Angela Bachi
- IFOM ETS-AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Elisabetta Marangoni
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| |
Collapse
|
35
|
Cantini L, Trapani D, Guidi L, Boscolo Bielo L, Scafetta R, Koziej M, Vidal L, Saini KS, Curigliano G. Neoadjuvant therapy in hormone Receptor-Positive/HER2-Negative breast cancer. Cancer Treat Rev 2024; 123:102669. [PMID: 38141462 DOI: 10.1016/j.ctrv.2023.102669] [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: 09/20/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/25/2023]
Abstract
Neoadjuvant therapy is commonly used in patients with locally advanced or inoperable breast cancer (BC). Neoadjuvant chemotherapy (NACT) represents an established treatment modality able to downstage tumours, facilitate breast-conserving surgery, yet also achieve considerable pathologic complete response (pCR) rates in HER2-positive and triple-negative BC. For patients with HR+/HER2- BC, the choice between NACT and neoadjuvant endocrine therapy (NET) is still based on clinical and pathological features and not guided by biomarkers of defined clinical utility, differently from the adjuvant setting where gene-expression signatures have been widely adopted to drive decision-making. In this review, we summarize the evidence supporting the choice of NACT vs NET in HR+/HER2- BC, discussing the issues surrounding clinical trial design and proper selection of patients for every treatment. It is time to question the binary paradigm of responder vs non-responders as well as the "one size fits all" approach in luminal BC, supporting the utilization of continuous endpoints and the adoption of tissue and plasma-based biomarkers at multiple timepoints. This will eventually unleash the full potential of neoadjuvant therapy which is to modulate patient treatment based on treatment sensitivity and surgical outcomes. We also reviewed the current landscape of neoadjuvant studies for HR+/HER2- BC, focusing on antibody-drug conjugates (ADCs) and immunotherapy combinations. Finally, we proposed a roadmap for future neoadjuvant approaches in HR+/HER2- BC, which should be based on a staggered biomarker-driven treatment selection aiming at impacting long-term relevant endpoints.
Collapse
Affiliation(s)
| | - Dario Trapani
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenzo Guidi
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Luca Boscolo Bielo
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Roberta Scafetta
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; Department of medical oncology, Campus Bio-Medico, University of Rome, Rome, Italy
| | | | | | | | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy.
| |
Collapse
|
36
|
Paiva CE, Zonta MPM, Granero RC, Guimarães VS, Pimenta LM, Teixeira GR, Paiva BSR. The Magee 3 Equation Predicts Favorable Pathologic Response to Neoadjuvant Endocrine Therapy in Breast Cancer Patients. Cancers (Basel) 2024; 16:339. [PMID: 38254828 PMCID: PMC10813970 DOI: 10.3390/cancers16020339] [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: 09/25/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Breast cancer (BC) remains a significant health care challenge, and treatment approaches continue to evolve. Among these, neoadjuvant endocrine therapy (NET) has gained prominence, particularly for postmenopausal, hormone-receptor positive, HER2-negative (HR+/HER2-) BC patients. Despite this, a significant gap exists in identifying patients who stand to benefit from NET. The objective of this study was to assess whether Magee equations (MEs) could serve as predictors of response to NET. METHODS This retrospective study included adult patients with invasive BC who underwent NET followed by curative surgery. Assessment of sociodemographic, clinical, and tumor-related variables was conducted. The ME1, ME2, ME3, and ME mean were analyzed to explore their predictive role for NET response. Receiver operating characteristic (ROC) curves were employed, along with the determination of optimal cutoff points. Logistic regression models were utilized to identify the most significant predictors of pathological response. RESULTS Among the 75 female participants, the mean age was 69.4 years, with the majority being postmenopausal (n = 72, 96%) and having an ECOG-PS of 0/1 (n = 63, 84%). Most patients were classified as luminal A (n = 41, 54.7%). ME3 emerged as a promising predictor, boasting an AUC of 0.734, with sensitivity of 90.62% and specificity of 57.50% when the threshold was ≤ 19.97. In univariate analysis, clinical staging (p = 0.002), molecular subtype (p = 0.001), and ME3 (continuous = 0.001, original 3-tier: p = 0.013, new 2-tier: <0.001) categories exhibited significant associations with pathological response. In the multivariate model, clinical staging and new 2-tier ME3 (<20 vs. ≥20) were included as significant variables. CONCLUSIONS Patients with ME3 < 20 have a higher likelihood of presenting a pathological response, offering a cost-effective alternative tool to Oncotype DX. Larger future studies with a prospective design are awaited to confirm our findings.
Collapse
Affiliation(s)
- Carlos Eduardo Paiva
- Department of Clinical Oncology, Barretos Cancer Hospital, Barretos 14784-400, SP, Brazil;
| | - Maria Paola Montesso Zonta
- Barretos School of Health Sciences Dr. Paulo Prata—FACISB, Barretos 14785-002, SP, Brazil; (M.P.M.Z.); (R.C.G.); (G.R.T.)
| | - Rafaela Carvalho Granero
- Barretos School of Health Sciences Dr. Paulo Prata—FACISB, Barretos 14785-002, SP, Brazil; (M.P.M.Z.); (R.C.G.); (G.R.T.)
| | - Vitor Souza Guimarães
- Department of Clinical Oncology, Barretos Cancer Hospital, Barretos 14784-400, SP, Brazil;
| | - Layla Melo Pimenta
- Department of Pathology, Barretos Cancer Hospital, Barretos 14784-400, SP, Brazil;
| | - Gustavo Ramos Teixeira
- Barretos School of Health Sciences Dr. Paulo Prata—FACISB, Barretos 14785-002, SP, Brazil; (M.P.M.Z.); (R.C.G.); (G.R.T.)
- Department of Pathology, Barretos Cancer Hospital, Barretos 14784-400, SP, Brazil;
| | | |
Collapse
|
37
|
López-Velazco JI, Manzano S, Otaño M, Elorriaga K, Bultó N, Herrero J, Lahuerta A, Segur V, Álvarez-López I, Caffarel MM, Urruticoechea A. A prospective study on tumour response assessment methods after neoadjuvant endocrine therapy in early oestrogen receptor-positive breast cancer. Breast Cancer Res 2024; 26:3. [PMID: 38173005 PMCID: PMC10765775 DOI: 10.1186/s13058-023-01756-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Neoadjuvant endocrine therapy (NET) in oestrogen receptor-positive (ER+) /HER2-negative (HER2-) breast cancer allows real-time evaluation of drug efficacy as well as investigation of the biological and molecular changes that occur after estrogenic deprivation. Clinical and pathological evaluation after NET may be used to obtain prognostic and predictive information of tumour response to decide adjuvant treatment. In this setting, clinical scales developed to evaluate response after neoadjuvant chemotherapy are not useful and there are not validated biomarkers to assess response to NET beyond Ki67 levels and preoperative endocrine prognostic index score (mPEPI). METHODS In this prospective study, we extensively analysed radiological (by ultrasound scan (USS) and magnetic resonance imaging (MRI)) and pathological tumour response of 104 postmenopausal patients with ER+ /HER2- resectable breast cancer, treated with NET for a mean of 7 months prior to surgery. We defined a new score, tumour cellularity size (TCS), calculated as the product of the residual tumour cellularity in the surgical specimen and the tumour pathological size. RESULTS Our results show that radiological evaluation of response to NET by both USS and MRI underestimates pathological tumour size (path-TS). Tumour size [mean (range); mm] was: path-TS 20 (0-80); radiological-TS by USS 9 (0-31); by MRI: 12 (0-60). Nevertheless, they support the use of MRI over USS to clinically assess radiological tumour response (rad-TR) due to the statistically significant association of rad-TR by MRI, but not USS, with Ki67 decrease (p = 0.002 and p = 0.3, respectively) and mPEPI score (p = 0.002 and p = 0.6, respectively). In addition, we propose that TCS could become a new tool to standardize response assessment to NET given its simplicity, reproducibility and its good correlation with existing biomarkers (such as ΔKi67, p = 0.001) and potential added value. CONCLUSION Our findings shed light on the dynamics of tumour response to NET, challenge the paradigm of the ability of NET to decrease surgical volume and point to the utility of the TCS to quantify the scattered tumour response usually produced by endocrine therapy. In the future, these results should be validated in independent cohorts with associated survival data.
Collapse
Affiliation(s)
- Joanna I López-Velazco
- Biogipuzkoa (Previously Known As Biodonostia) Health Research Institute, Paseo Dr Begiristain S/N, 20014, San Sebastian, Spain
| | - Sara Manzano
- Biogipuzkoa (Previously Known As Biodonostia) Health Research Institute, Paseo Dr Begiristain S/N, 20014, San Sebastian, Spain
| | - María Otaño
- Gipuzkoa Cancer Unit, OSI Donostialdea - Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014, San Sebastian, Spain
| | - Kepa Elorriaga
- Gipuzkoa Cancer Unit, OSI Donostialdea - Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014, San Sebastian, Spain
| | - Núria Bultó
- Gipuzkoa Cancer Unit, OSI Donostialdea - Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014, San Sebastian, Spain
| | - Julio Herrero
- Gipuzkoa Cancer Unit, OSI Donostialdea - Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014, San Sebastian, Spain
| | - Ainhara Lahuerta
- Biogipuzkoa (Previously Known As Biodonostia) Health Research Institute, Paseo Dr Begiristain S/N, 20014, San Sebastian, Spain
- Gipuzkoa Cancer Unit, OSI Donostialdea - Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014, San Sebastian, Spain
| | - Virginia Segur
- Gipuzkoa Cancer Unit, OSI Donostialdea - Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014, San Sebastian, Spain
| | - Isabel Álvarez-López
- Biogipuzkoa (Previously Known As Biodonostia) Health Research Institute, Paseo Dr Begiristain S/N, 20014, San Sebastian, Spain
- Gipuzkoa Cancer Unit, OSI Donostialdea - Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014, San Sebastian, Spain
| | - Maria M Caffarel
- Biogipuzkoa (Previously Known As Biodonostia) Health Research Institute, Paseo Dr Begiristain S/N, 20014, San Sebastian, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| | - Ander Urruticoechea
- Biogipuzkoa (Previously Known As Biodonostia) Health Research Institute, Paseo Dr Begiristain S/N, 20014, San Sebastian, Spain.
- Gipuzkoa Cancer Unit, OSI Donostialdea - Onkologikoa Foundation, Paseo Dr Begiristain 121, 20014, San Sebastian, Spain.
| |
Collapse
|
38
|
Paiva CE, Silva ATF, Oliveira IDS, Guimarães VS, Lacerda DC, Teixeira GR, Watanabe AHU, Onari N, Paiva BSR, de Oliveira-Junior I, Marques MMC, Maia YCDP. A Research Protocol for a Phase II Single-Arm Clinical Trial Assessing the Feasibility and Efficacy of Neoadjuvant Anastrozole in Patients With Luminal Breast Cancer and Low Proliferative Index: The ANNE Trial. Cancer Control 2024; 31:10732748241272463. [PMID: 39140157 PMCID: PMC11325316 DOI: 10.1177/10732748241272463] [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: 01/19/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
INTRODUCTION Neoadjuvant endocrine therapy (NET) is recommended for the treatment of invasive breast cancer (BC), particularly luminal subtypes, in locally advanced stages. Previous randomized studies have demonstrated the benefits of aromatase inhibitors in this context. However, NET is typically reserved for elderly or frail patients who may not tolerate neoadjuvant chemotherapy. Identifying non-responsive patients early and extending treatment for responsive ones would be ideal, yet optimal strategies are awaited. AIMS This non-randomized phase 2 clinical trial aims to assess NET feasibility and efficacy in postmenopausal stage II and III luminal BC patients, identifying predictive therapeutic response biomarkers. Efficacy will be gauged by patients with Ki67 ≤ 10% after 4 weeks and Preoperative Endocrine Prognostic Index (PEPI) scores 0 post-surgery. Study feasibility will be determined by participation acceptance rate (recruitment rate ≥50%) and inclusion rate (>2 patients/month). METHODS Postmenopausal women with luminal, HER2-tumors in stages II and III undergo neoadjuvant anastrozole treatment, evaluating continuing NET or receiving chemotherapy through early Ki67 analysis after 2 to 4 weeks. The study assesses NET extension for up to 10 months, using serial follow-ups with standardized breast ultrasound and clinical criteria-based NET suspension. Clinical and pathological responses will be measured overall and in the luminal tumor A subgroup. Toxicity, health-related quality of life, and circulating biomarkers predicting early NET response will also be evaluated.
Collapse
Affiliation(s)
- Carlos Eduardo Paiva
- Deparment of Clinical Oncology, Barretos Cancer Hospital, Barretos-SP, Brazil
- Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, Barretos-SP, Brazil
| | - Alinne Tatiane Faria Silva
- Nutrition and Molecular Biology Research Group, School of Medicine, Federal University of Uberlandia, Uberlandia, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, Brazil
| | - Izabella da Silva Oliveira
- Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, Barretos-SP, Brazil
| | - Vitor Souza Guimarães
- Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, Barretos-SP, Brazil
| | | | - Gustavo Ramos Teixeira
- Department of Pathology, Barretos Cancer Hospital, Barretos-SP, Brazil
- Barretos School of Health Sciences Dr. Paulo Prata – FACISB, Barretos-SP, Brazil
| | | | - Nilton Onari
- Department of Breast Radiology, Barretos Cancer Hospital, Barretos-SP, Brazil
| | | | | | | | - Yara Cristina de Paiva Maia
- Nutrition and Molecular Biology Research Group, School of Medicine, Federal University of Uberlandia, Uberlandia, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, Brazil
| |
Collapse
|
39
|
Kim H, Whitman AA, Wisniewska K, Kakati RT, Garcia-Recio S, Calhoun BC, Franco HL, Perou CM, Spanheimer PM. Tamoxifen Response at Single-Cell Resolution in Estrogen Receptor-Positive Primary Human Breast Tumors. Clin Cancer Res 2023; 29:4894-4907. [PMID: 37747807 PMCID: PMC10690085 DOI: 10.1158/1078-0432.ccr-23-1248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/18/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
PURPOSE In estrogen receptor-positive (ER+)/HER2- breast cancer, multiple measures of intratumor heterogeneity are associated with a worse response to endocrine therapy. We sought to develop a novel experimental model to measure heterogeneity in response to tamoxifen treatment in primary breast tumors. EXPERIMENTAL DESIGN To investigate heterogeneity in response to treatment, we developed an operating room-to-laboratory pipeline for the collection of live normal breast specimens and human tumors immediately after surgical resection for processing into single-cell workflows for experimentation and genomic analyses. Live primary cell suspensions were treated ex vivo with tamoxifen (10 μmol/L) or control media for 12 hours, and single-cell RNA libraries were generated using the 10X Genomics droplet-based kit. RESULTS In total, we obtained and processed normal breast tissue from two women undergoing reduction mammoplasty and tumor tissue from 10 women with ER+/HER2- invasive breast carcinoma. We demonstrate differences in tamoxifen response by cell type and identify distinctly responsive and resistant subpopulations within the malignant cell compartment of human tumors. Tamoxifen resistance signatures from resistant subpopulations predict poor outcomes in two large cohorts of ER+ breast cancer patients and are enriched in endocrine therapy-resistant tumors. CONCLUSIONS This novel ex vivo model system now provides the foundation to define responsive and resistant subpopulations within heterogeneous human tumors, which can be used to develop precise single cell-based predictors of response to therapy and to identify genes and pathways driving therapeutic resistance.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Austin A. Whitman
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Kamila Wisniewska
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Rasha T. Kakati
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Susana Garcia-Recio
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Benjamin C. Calhoun
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hector L. Franco
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
- Computational Medicine Program, University of North Carolina, Chapel Hill, North Carolina
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
- Computational Medicine Program, University of North Carolina, Chapel Hill, North Carolina
| | - Philip M. Spanheimer
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
40
|
Martins-Branco D, Nader-Marta G, Molinelli C, Ameye L, Paesmans M, Ignatiadis M, Aftimos P, Salgado R, de Azambuja E. Ki-67 index after neoadjuvant endocrine therapy as a prognostic biomarker in patients with ER-positive/HER2-negative early breast cancer: a systematic review and meta-analysis. Eur J Cancer 2023; 194:113358. [PMID: 37857118 DOI: 10.1016/j.ejca.2023.113358] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Neoadjuvant treatment discriminates responders, but pathologic complete response is uncommon in oestrogen receptor (ER)-positive/HER2-negative early breast cancer. We aimed to assess the prognostic value of Ki-67 index after neoadjuvant endocrine therapy (NET). METHODS We conducted a systematic literature search of PubMed, Embase, CENTRAL, and conference proceedings up to 22nd August 2023 to identify studies reporting the association of Ki-67 index after NET with recurrence-free survival (RFS) and/or overall survival (OS) in women with ER-positive/HER2-negative early breast cancer. We combined RFS and OS hazard ratios (HRs) with 95% confidence intervals (CIs). RESULTS Twelve studies including 7897 patients were analysed. Most studies were clinical trials (n = 7547) including only postmenopausal women (n = 3953) treated with aromatase inhibitor (n = 3359). Three studies evaluated Ki-67 in a preplanned core biopsy at 2-4 weeks of NET (n = 3348), while nine evaluated Ki-67 in the surgical specimen (n = 4549) after 2-24 weeks of NET. Median follow-up ranged between 37 and 95 months for RFS and 62-84 months for OS. High Ki-67 index after NET was significantly associated with worse RFS (HR 2.48, 95% CI 1.86-3.30) and OS (HR 2.66, 95% CI 1.65-4.28). A sensitivity analysis including three studies that measured Ki-67 in a preplanned core biopsy showed similar association with RFS (HR 2.41, 95% CI 1.77-3.30). CONCLUSIONS High Ki-67 after NET is associated with worse survival outcomes, even after a short course of NET, emphasising the prognostic value of this biomarker in women with ER-positive/HER2-negative early breast cancer.
Collapse
Affiliation(s)
- Diogo Martins-Branco
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium.
| | - Guilherme Nader-Marta
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Chiara Molinelli
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Lieveke Ameye
- Data Center, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Marianne Paesmans
- Data Center, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Michail Ignatiadis
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium; Medical Oncology Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Philippe Aftimos
- Medical Oncology Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Roberto Salgado
- Department of Pathology, ZAS-Hospitals, Antwerp, Belgium; Division of Research, Peter Mac Callum Cancer Centre, Melbourne, Australia
| | - Evandro de Azambuja
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium; Medical Oncology Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| |
Collapse
|
41
|
Das K, Paul S, Ghosh A, Gupta S, Mukherjee T, Shankar P, Sharma A, Keshava S, Chauhan SC, Kashyap VK, Parashar D. Extracellular Vesicles in Triple-Negative Breast Cancer: Immune Regulation, Biomarkers, and Immunotherapeutic Potential. Cancers (Basel) 2023; 15:4879. [PMID: 37835573 PMCID: PMC10571545 DOI: 10.3390/cancers15194879] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype accounting for ~10-20% of all human BC and is characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) amplification. Owing to its unique molecular profile and limited targeted therapies, TNBC treatment poses significant challenges. Unlike other BC subtypes, TNBC lacks specific molecular targets, rendering endocrine therapies and HER2-targeted treatments ineffective. The chemotherapeutic regimen is the predominant systemic treatment modality for TNBC in current clinical practice. However, the efficacy of chemotherapy in TNBC is variable, with response rates varying between a wide range of patients, and the emerging resistance further adds to the difficulties. Furthermore, TNBC exhibits a higher mutational burden and is acknowledged as the most immunogenic of all BC subtypes. Consequently, the application of immune checkpoint inhibition has been investigated in TNBC, yielding promising outcomes. Recent evidence identified extracellular vesicles (EVs) as an important contributor in the context of TNBC immunotherapy. In view of the extraordinary ability of EVs to transfer bioactive molecules, such as proteins, lipids, DNA, mRNAs, and small miRNAs, between the cells, EVs are considered a promising diagnostic biomarker and novel drug delivery system among the prospects for immunotherapy. The present review provides an in-depth understanding of how EVs influence TNBC progression, its immune regulation, and their contribution as a predictive biomarker for TNBC. The final part of the review focuses on the recent key advances in immunotherapeutic strategies for better understanding the complex interplay between EVs and the immune system in TNBC and further developing EV-based targeted immunotherapies.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700012, India; (S.P.); (A.G.)
| | - Arnab Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700012, India; (S.P.); (A.G.)
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura 281406, India;
| | - Tanmoy Mukherjee
- School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Prem Shankar
- Department of Neurobiology, The University of Texas Medical Branch, Galveston, TX 77555, USA or
| | - Anshul Sharma
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.C.C.); (V.K.K.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Vivek Kumar Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.C.C.); (V.K.K.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Deepak Parashar
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
42
|
Meneveau MO, Crawford MP, Turkheimer LM, Millard TA, Atkins KA, Showalter SL. The POWER-PAK Score Characterizes Tumor Response to 3 Months of Preoperative Endocrine Therapy. Ann Surg Oncol 2023; 30:6034-6040. [PMID: 37454014 PMCID: PMC10766409 DOI: 10.1245/s10434-023-13892-8] [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: 04/19/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND The Pre-Operative Window of Endocrine Therapy to Inform Radiation Therapy Decisions (POWER, NCT04272801) trial aims to determine whether 3 months of preoperative endocrine therapy (pre-ET) informs adjuvant radiation therapy decisions among older women with early stage, ER-positive breast cancer. We propose the POWER Pathologic Assessment and Ki-67 (POWER-PAK) scoring system to characterize the histologic effects of pre-ET. METHODS Histologic evaluation was performed on core biopsy and lumpectomy specimens from 37 POWER trial participants who completed pre-ET and surgery. The POWER-PAK score consists of tumor regression, decrease in Ki-67 expression, and ER expression, each ranging from 0 to 2. Scores were aggregated to create the POWER-PAK score with a range from 0 to 6. Participants with no residual tumor were labelled 6-NRT. RESULTS ER expression did not decrease after pre-ET. Ki-67 decreased from 13% in biopsy specimens to 5% in the lumpectomy specimens (p < 0.001). Cellularity decreased from 40% to 23% (p < 0.001). There was heterogeneity of POWER-PAK scores ranging from 2 to 6-NRT: score of 2, n = 2 (5.4%); 4, n = 8 (21.6%); 5, n = 4 (10.8%); 6, n = 16 (43.2%); and 6-NRT, n = 7 (18.9%). Participants with a score ≥ 5 were more likely to have smaller tumors after pre-ET compared with those with a score < 5 (p = 0.04). CONCLUSIONS The tumor responses following treatment with pre-ET are heterogenous. We propose that the POWER-PAK scoring system can be used to quantify response to pre-ET. Future studies will explore the use of POWER-PAK to support informed decision-making for adjuvant therapy options for older women with early stage breast cancer.
Collapse
Affiliation(s)
- Max O Meneveau
- Department of Surgery, Division of Breast and Melanoma Surgery, University of Virginia, Charlottesville, VA, USA
| | - Michael P Crawford
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Lena M Turkheimer
- Department of Surgery, Division of Breast and Melanoma Surgery, University of Virginia, Charlottesville, VA, USA
| | - Trish A Millard
- Department of Medicine, Division of Hematology/Oncology, University of Virginia, Charlottesville, VA, USA
| | - Kristen A Atkins
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Shayna L Showalter
- Department of Surgery, Division of Breast and Melanoma Surgery, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
43
|
Chaudhary LN, Jorns JM, Sun Y, Cheng YC, Kamaraju S, Burfeind J, Gonyo MB, Kong AL, Patten C, Yen T, Cortina CS, Carson E, Johnson N, Bergom C, Tsaih SW, Banerjee A, Wang Y, Chervoneva I, Weil E, Chitambar CR, Rui H. Frequent upregulation of HER2 protein in hormone-receptor-positive HER2-negative breast cancer after short-term neoadjuvant endocrine therapy. Breast Cancer Res Treat 2023; 201:387-396. [PMID: 37460683 PMCID: PMC10795510 DOI: 10.1007/s10549-023-07038-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Endocrine resistant metastatic disease develops in ~ 20-25% of hormone-receptor-positive (HR+) breast cancer (BC) patients despite endocrine therapy (ET) use. Upregulation of HER family receptor tyrosine kinases (RTKs) represent escape mechanisms in response to ET in some HR+ tumors. Short-term neoadjuvant ET (NET) offers the opportunity to identify early endocrine escape mechanisms initiated in individual tumors. METHODS This was a single arm, interventional phase II clinical trial evaluating 4 weeks (± 1 week) of NET in patients with early-stage HR+/HER2-negative (HER2-) BC. The primary objective was to assess NET-induced changes in HER1-4 proteins by immunohistochemistry (IHC) score. Protein upregulation was defined as an increase of ≥ 1 in IHC score following NET. RESULTS Thirty-seven patients with cT1-T3, cN0, HR+/HER2- BC were enrolled. In 35 patients with evaluable tumor HER protein after NET, HER2 was upregulated in 48.6% (17/35; p = 0.025), with HER2-positive status (IHC 3+ or FISH-amplified) detected in three patients at surgery, who were recommended adjuvant trastuzumab-based therapy. Downregulation of HER3 and/or HER4 protein was detected in 54.2% of tumors, whereas HER1 protein remained low and unchanged in all cases. While no significant volumetric reduction was detected radiographically after short-term NET, significant reduction in tumor proliferation rates were observed. No significant associations were identified between any clinicopathologic covariates and changes in HER1-4 protein expression on multivariable analysis. CONCLUSION Short-term NET frequently and preferentially upregulates HER2 over other HER family RTKs in early-stage HR+/HER2- BC and may be a promising strategy to identify tumors that utilize HER2 as an early endocrine escape pathway. CLINICAL TRIAL REGISTRY Trial registration number: NCT03219476.
Collapse
Affiliation(s)
- Lubna N Chaudhary
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA.
| | - Julie M Jorns
- Department of Pathology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Yunguang Sun
- Department of Pathology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Yee Chung Cheng
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Sailaja Kamaraju
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - John Burfeind
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Mary Beth Gonyo
- Department of Radiology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Amanda L Kong
- Department of Surgery, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Caitlin Patten
- Department of Surgery, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Tina Yen
- Department of Surgery, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Chandler S Cortina
- Department of Surgery, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Ebony Carson
- Clinical Trials Office, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Nedra Johnson
- Clinical Trials Office, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Shirng-Wern Tsaih
- Department of Obstetrics and Gynecology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Anjishnu Banerjee
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, USA
| | - Yu Wang
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, USA
| | - Inna Chervoneva
- Division of Biostatistics, Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Elizabeth Weil
- Division of Pharmacy, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Christopher R Chitambar
- Division of Hematology and Oncology, Department of Medicine, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Hallgeir Rui
- Department of Pathology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| |
Collapse
|
44
|
Jeong H, Kim SB. Neoadjuvant endocrine therapy in ER-positive breast cancer: evolution, indication, and tailored treatment strategy. Ther Adv Med Oncol 2023; 15:17588359231200457. [PMID: 37786536 PMCID: PMC10541763 DOI: 10.1177/17588359231200457] [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: 11/27/2022] [Accepted: 08/25/2023] [Indexed: 10/04/2023] Open
Abstract
In recent years, endocrine therapy (ET), an effective systemic treatment for the management of estrogen receptor (ER)-positive breast cancers, has regained interest as a neoadjuvant therapy based on evidence that ET can fulfill the aim of neoadjuvant systemic treatment for tumor shrinkage as well as elucidate important clinical information on endocrine sensitivity that enables the prognostication of patients. Moreover, neoadjuvant endocrine therapy (NET) potentially provides an opportunity for early assessment of the clinical efficacy of novel agents. Furthermore, recently reported trials have generated evidence for a more tailored approach for perioperative management of ER-positive breast cancer using clinical and molecular biomarkers, and this has provided a rationale that enables the broadening of clinical indications for NET. This review discusses the current evidence for NET, the evolution of NET trials, clinical indications, and NET-based treatment strategies.
Collapse
Affiliation(s)
- Hyehyun Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea
| |
Collapse
|
45
|
Litton JK, Regan MM, Pusztai L, Rugo HS, Tolaney SM, Garrett-Mayer E, Amiri-Kordestani L, Basho RK, Best AF, Boileau JF, Denkert C, Foster JC, Harbeck N, Jacene HA, King TA, Mason G, O'Sullivan CC, Prowell TM, Richardson AL, Sepulveda KA, Smith ML, Tjoe JA, Turashvili G, Woodward WA, Butler LP, Schwartz EI, Korde LA. Standardized Definitions for Efficacy End Points in Neoadjuvant Breast Cancer Clinical Trials: NeoSTEEP. J Clin Oncol 2023; 41:4433-4442. [PMID: 37433103 PMCID: PMC10522109 DOI: 10.1200/jco.23.00435] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/25/2023] [Accepted: 06/06/2023] [Indexed: 07/13/2023] Open
Abstract
PURPOSE The Standardized Definitions for Efficacy End Points (STEEP) criteria, established in 2007 and updated in 2021 (STEEP 2.0), provide standardized definitions of adjuvant breast cancer (BC) end points. STEEP 2.0 identified a need to separately address end points for neoadjuvant clinical trials. The multidisciplinary NeoSTEEP working group of experts was convened to critically evaluate and align neoadjuvant BC trial end points. METHODS The NeoSTEEP working group concentrated on neoadjuvant systemic therapy end points in clinical trials with efficacy outcomes-both pathologic and time-to-event survival end points-particularly for registrational intent. Special considerations for subtypes and therapeutic approaches, imaging, nodal staging at surgery, bilateral and multifocal diseases, correlative tissue collection, and US Food and Drug Administration regulatory considerations were contemplated. RESULTS The working group recommends a preferred definition of pathologic complete response (pCR) as the absence of residual invasive cancer in the complete resected breast specimen and all sampled regional lymph nodes (ypT0/Tis ypN0 per AJCC staging). Residual cancer burden should be a secondary end point to facilitate future assessment of its utility. Alternative end points are needed for hormone receptor-positive disease. Time-to-event survival end point definitions should pay particular attention to the measurement starting point. Trials should include end points originating at random assignment (event-free survival and overall survival) to capture presurgery progression and deaths as events. Secondary end points adapted from STEEP 2.0, which are defined from starting at curative-intent surgery, may also be appropriate. Specification and standardization of biopsy protocols, imaging, and pathologic nodal evaluation are also crucial. CONCLUSION End points in addition to pCR should be selected on the basis of clinical and biologic aspects of the tumor and the therapeutic agent investigated. Consistent prespecified definitions and interventions are paramount for clinically meaningful trial results and cross-trial comparison.
Collapse
Affiliation(s)
- Jennifer K. Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Meredith M. Regan
- Division of Biostatistics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Lajos Pusztai
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Hope S. Rugo
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - Sara M. Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - Reva K. Basho
- The Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA
| | - Ana F. Best
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD
| | | | - Carsten Denkert
- Institute of Pathology, Philipps University Marburg and University Hospital Marburg (UKGM), Marburg, Germany
| | - Jared C. Foster
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD
| | - Nadia Harbeck
- The Breast Center, Department of Obstetrics and Gynecology and Comprehensive Cancer Center Munich, LMU University Hospital, Munich, Germany
| | | | - Tari A. King
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA
| | - Ginny Mason
- The Inflammatory Breast Cancer Research Foundation, Broadway, VA
| | | | - Tatiana M. Prowell
- US Food and Drug Administration, Silver Spring, MD
- Women's Malignancies Disease Group, Johns Hopkins Kimmel Comprehensive Cancer Center, Baltimore, MD
| | | | | | | | - Judy A. Tjoe
- Division of Breast Surgery, Department of Surgery, Novant Health, Greensboro, NC
| | - Gulisa Turashvili
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, GA
| | - Wendy A. Woodward
- Department of Breast Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Elena I. Schwartz
- Coordinating Center for Clinical Trials, National Cancer Institute, Rockville, MD
| | - Larissa A. Korde
- Cancer Therapy and Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD
| |
Collapse
|
46
|
Fukui R, Watanabe T, Morimoto K, Fujimoto Y, Nagahashi M, Ishikawa E, Hirota S, Miyoshi Y. An increase in tumor-infiltrating lymphocytes after treatment is significantly associated with a poor response to neoadjuvant endocrine therapy for estrogen receptor-positive/HER2-negative breast cancers. Breast Cancer 2023; 30:703-713. [PMID: 37115435 PMCID: PMC10404203 DOI: 10.1007/s12282-023-01462-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND The reason for the poor prognosis of estrogen receptor (ER) + /human epidermal growth factor receptor 2 (HER2)- breast cancer patients with high levels of tumor-infiltrating lymphocytes (TILs) is poorly understood. The association between TILs and response to neoadjuvant endocrine therapy (NET) was examined. METHODS We recruited 170 patients with ER + /HER2- breast cancer who were treated with preoperative endocrine monotherapy. TILs were evaluated before and after NET, and their changes were noted. Furthermore, T cell subtypes were examined using CD8 and FOXP3 immunohistochemical analyses. Neutrophil and lymphocyte counts in the peripheral blood were analyzed with reference to TIL levels or changes. Responders were defined as Ki67 expression levels ≤ 2.7% after treatment. RESULTS Post-treatment (p = 0.016), but not pre-treatment (p = 0.464), TIL levels were significantly associated with the response to NET. TIL levels increased significantly after treatment among non-responders (p = 0.001). FOXP3 + T cell counts increased significantly after treatment in patients with increased TILs (p = 0.035), but not in those without increased TILs (p = 0.281). Neutrophil counts decreased significantly after treatment in patients without increased TILs (p = 0.026), but not in patients with increased TILs (p = 0.312). CONCLUSION An increase in TILs after NET was significantly associated with a poor response to NET. Given that FOXP3 + T-cell counts increased, and neutrophil counts did not decrease in patients with increased TILs after NET, the induction of an immunosuppressive microenvironment was speculated to play a role in the inferior efficacy. These data might partially indicate the involvement of the immune response in the efficacy of endocrine therapy.
Collapse
Affiliation(s)
- Reiko Fukui
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, Mukogawa-Cho 1-1, Nishinomiya City, Hyogo, 663-8501, Japan
| | - Takahiro Watanabe
- Department of Clinical Pathology, Chibune General Hospital, Osaka, Japan
| | - Koji Morimoto
- Department of Nutrition, College of Nutrition, Koshien University, Takarazuka, Hyogo, Japan
| | - Yukie Fujimoto
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, Mukogawa-Cho 1-1, Nishinomiya City, Hyogo, 663-8501, Japan
| | - Masayuki Nagahashi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, Mukogawa-Cho 1-1, Nishinomiya City, Hyogo, 663-8501, Japan
| | - Eri Ishikawa
- Department of Surgical Pathology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Yasuo Miyoshi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, Mukogawa-Cho 1-1, Nishinomiya City, Hyogo, 663-8501, Japan.
| |
Collapse
|
47
|
Huppert LA, Gumusay O, Idossa D, Rugo HS. Systemic therapy for hormone receptor-positive/human epidermal growth factor receptor 2-negative early stage and metastatic breast cancer. CA Cancer J Clin 2023; 73:480-515. [PMID: 36939293 DOI: 10.3322/caac.21777] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 01/09/2023] [Accepted: 02/01/2023] [Indexed: 03/21/2023] Open
Abstract
Hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative breast cancer is defined by the presence of the estrogen receptor and/or the progesterone receptor and the absence of HER2 gene amplification. HR-positive/HER2-negative breast cancer accounts for 65%-70% of all breast cancers, and incidence increases with increasing age. Treatment varies by stage, and endocrine therapy is the mainstay of treatment in both early stage and late-stage disease. Combinations with cyclin-dependent kinase 4/6 inhibitors have reduced distant recurrence in the early stage setting and improved overall survival in the metastatic setting. Chemotherapy is used based on stage and tumor biology in the early stage setting and after endocrine resistance for advanced disease. New therapies, including novel endocrine agents and antibody-drug conjugates, are now changing the treatment landscape. With the availability of new treatment options, it is important to define the optimal sequence of treatment to maximize clinical benefit while minimizing toxicity. In this review, the authors first discuss the pathologic and molecular features of HR-positive/HER2-negative breast cancer and mechanisms of endocrine resistance. Then, they discuss current and emerging therapies for both early stage and metastatic HR-positive/HER2-negative breast cancer, including treatment algorithms based on current data.
Collapse
Affiliation(s)
- Laura A Huppert
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Ozge Gumusay
- Department of Medical Oncology, Acibadem University, School of Medicine, Istanbul, Turkey
| | - Dame Idossa
- Masonic Comprehensive Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hope S Rugo
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| |
Collapse
|
48
|
Curigliano G, Dent R, Llombart-Cussac A, Pegram M, Pusztai L, Turner N, Viale G. Incorporating clinicopathological and molecular risk prediction tools to improve outcomes in early HR+/HER2- breast cancer. NPJ Breast Cancer 2023; 9:56. [PMID: 37380659 PMCID: PMC10307886 DOI: 10.1038/s41523-023-00560-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/06/2023] [Indexed: 06/30/2023] Open
Abstract
Stratification of recurrence risk is a cornerstone of early breast cancer diagnosis that informs a patient's optimal treatment pathway. Several tools exist that combine clinicopathological and molecular information, including multigene assays, which can estimate risk of recurrence and quantify the potential benefit of different adjuvant treatment modalities. While the tools endorsed by treatment guidelines are supported by level I and II evidence and provide similar prognostic accuracy at the population level, they can yield discordant risk prediction at the individual patient level. This review examines the evidence for these tools in clinical practice and offers a perspective of potential future risk stratification strategies. Experience from clinical trials with cyclin D kinase 4/6 (CDK4/6) inhibitors in the setting of hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) early breast cancer is provided as an illustrative example of risk stratification.
Collapse
Affiliation(s)
- Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy.
| | | | | | | | | | | | - Giuseppe Viale
- European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| |
Collapse
|
49
|
Serrano D, Gandini S, Thomas P, Crew KD, Kumar NB, Vornik LA, Lee JJ, Veronesi P, Viale G, Guerrieri-Gonzaga A, Lazzeroni M, Johansson H, D’Amico M, Guasone F, Spinaci S, Bertelsen BE, Mellgren G, Bedrosian I, Weber D, Castile T, Dimond E, Heckman-Stoddard BM, Szabo E, Brown PH, DeCensi A, Bonanni B. Efficacy of Alternative Dose Regimens of Exemestane in Postmenopausal Women With Stage 0 to II Estrogen Receptor-Positive Breast Cancer: A Randomized Clinical Trial. JAMA Oncol 2023; 9:664-672. [PMID: 36951827 PMCID: PMC10037202 DOI: 10.1001/jamaoncol.2023.0089] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/06/2022] [Indexed: 03/24/2023]
Abstract
Importance Successful therapeutic cancer prevention requires definition of the minimal effective dose. Aromatase inhibitors decrease breast cancer incidence in high-risk women, but use in prevention and compliance in adjuvant settings are hampered by adverse events. Objective To compare the noninferiority percentage change of estradiol in postmenopausal women with estrogen receptor-positive breast cancer given exemestane, 25 mg, 3 times weekly or once weekly vs a standard daily dose with a noninferiority margin of -6%. Design, Setting, and Participants This multicenter, presurgical, double-blind phase 2b randomized clinical trial evaluated 2 alternative dosing schedules of exemestane. Postmenopausal women with estrogen receptor-positive breast cancer who were candidates for breast surgery were screened from February 1, 2017, to August 31, 2019. Blood samples were collected at baseline and final visit; tissue biomarker changes were assessed from diagnostic biopsy and surgical specimen. Biomarkers were measured in different laboratories between April 2020 and December 2021. Interventions Exemestane, 25 mg, once daily, 3 times weekly, or once weekly for 4 to 6 weeks before surgery. Main Outcomes and Measures Serum estradiol concentrations were measured by solid-phase extraction followed by liquid chromatography-tandem mass spectrometry detection. Toxic effects were evaluated using the National Cancer Institute terminology criteria, and Ki-67 was assessed by immunohistochemistry. Results A total of 180 women were randomized into 1 of the 3 arms; median (IQR) age was 66 (60-71) years, 63 (60-69) years, and 65 (61-70) years in the once-daily, 3-times-weekly, and once-weekly arms, respectively. In the intention-to-treat population (n = 171), the least square mean percentage change of serum estradiol was -89%, -85%, and -60% for exemestane once daily (n = 55), 3 times weekly (n = 56), and once weekly (n = 60), respectively. The difference in estradiol percentage change between the once-daily and 3-times-weekly arms was -3.6% (P for noninferiority = .37), whereas in compliant participants (n = 153), it was 2.0% (97.5% lower confidence limit, -5.6%; P for noninferiority = .02). Among secondary end points, Ki-67 and progesterone receptor were reduced in all arms, with median absolute percentage changes of -7.5%, -5.0%, and -4.0% for Ki-67 in the once-daily, 3-times-weekly, and once-weekly arms, respectively (once daily vs 3 times weekly, P = .31; once daily vs once weekly, P = .06), and -17.0%, -9.0%, and -7.0% for progesterone receptor, respectively. Sex hormone-binding globulin and high-density lipoprotein cholesterol had a better profile among participants in the 3-times-weekly arm compared with once-daily arm. Adverse events were similar in all arms. Conclusions and Relevance In this randomized clinical trial, exemestane, 25 mg, given 3 times weekly in compliant patients was noninferior to the once-daily dosage in decreasing serum estradiol. This new schedule should be further studied in prevention studies and in women who do not tolerate the daily dose in the adjuvant setting. Trial Registration ClinicalTrials.gov Identifier: NCT02598557; EudraCT: 2015-005063-16.
Collapse
Affiliation(s)
| | - Sara Gandini
- European Institute of Oncology IRCCS, Milan, Italy
| | | | | | - Nagi B. Kumar
- Moffitt Cancer Center, University of South Florida, Tampa
| | - Lana A. Vornik
- The University of Texas MD Anderson Cancer Center, Houston
| | - J. Jack Lee
- The University of Texas MD Anderson Cancer Center, Houston
| | | | | | | | | | | | | | | | | | - Bjørn-Erik Bertelsen
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Diane Weber
- The University of Texas MD Anderson Cancer Center, Houston
| | - Tawana Castile
- The University of Texas MD Anderson Cancer Center, Houston
| | - Eileen Dimond
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | | | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Powel H. Brown
- The University of Texas MD Anderson Cancer Center, Houston
| | - Andrea DeCensi
- Ospedali Galliera, Genoa, Italy
- Wolfson Institute of Population Health, Queen Mary University of London, London, England, United Kingdom
| | | |
Collapse
|
50
|
Elayoubi J, Chi J, Mahmoud AA, Alloghbi A, Assad H, Shekhar M, Simon MS. A Review of Endocrine Therapy in Early-stage Breast Cancer: The Journey From Crudeness to Precision. Am J Clin Oncol 2023; 46:225-230. [PMID: 36856249 DOI: 10.1097/coc.0000000000000993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Endocrine therapy (ET) is the standard of care for hormone receptor-positive early-stage breast cancer in the adjuvant setting. However, response to ET can vary across patient subgroups. Historically, hormone receptor expression and clinical stage are the main predictors of the benefit of ET. A "window of opportunity" trials has raised significant interest in recent years as a means of assessing the sensitivity of a patient's cancer to short-term neoadjuvant ET, which provides important prognostic information, and helps in decision-making regarding treatment options in a time-efficient and cost-efficient manner. In the era of genomics, molecular profiling has led to the discovery and evaluation of the prognostic and predictive abilities of new molecular profiles. To realize the goal of personalized medicine, we are in urgent need to explore reliable biomarkers or genomic signatures to accurately predict the clinical response and long-term outcomes associated with ET. Validation of these biomarkers as reliable surrogate endpoints can also lead to a revolution in the clinical trial designs, and potentially avoid the need for repeated tissue biopsies in the surveillance of disease response. The clinical potential of tumor genomic profiling marks the beginning of a new era of precision medicine in breast cancer treatment.
Collapse
Affiliation(s)
- Jailan Elayoubi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Jie Chi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Amr A Mahmoud
- Department of Clinical Oncology, Kafr Elshiekh University, Egypt
| | - Abdurahman Alloghbi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Hadeel Assad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Malathy Shekhar
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Michael S Simon
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI
| |
Collapse
|