1
|
Lafci O, Resch D, Santonocito A, Clauser P, Helbich T, Baltzer PAT. Role of imaging based response assesment for adapting neoadjuvant systemic therapy for breast cancer: A systematic review. Eur J Radiol 2025; 187:112105. [PMID: 40252279 DOI: 10.1016/j.ejrad.2025.112105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
PURPOSE The objective of this systematic review is to investigate the role of imaging in response monitoring during neoadjuvant systemic therapy (NST) for breast cancer and assess whether treatment modifications based on imaging response are implemented in clinical practice. METHODS A systematic review was conducted, analyzing five clinical practice guidelines and 147 clinical trial publications involving NST for breast cancer. The snowballing technique was employed, using a "start set" of clinical guidelines to trace relevant trials. Additionally, a PubMed search was conducted to identify trials published between 2023-2024. The review analyzed the use of imaging modalities, timing, and response criteria, and whether escalation, de-escalation, or change of treatment occurred based on imaging response. RESULTS Imaging was utilized in 81 % (119/147) of the trials, with ultrasound, MRI, and mammography being the most frequently employed modalities. Mid-treatment imaging was applied in 56 % (83/147) of the trials. However, only 15 % (22/147) of the trials implemented treatment modifications based on imaging response, highlighting the limited application of imaging response-guided therapy. No standardized imaging protocols or consistent response-guided treatment strategies were identified across the trials or clinical practice guidelines, with considerable variability in imaging methods, timing, and response criteria. CONCLUSION This systematic review underscores the critical need for standardized imaging protocols, response assessment criteria and image-guided treatment decisions. It is therefore evident that imaging for response monitoring during treatment should preferably be performed within clinical trials.
Collapse
Affiliation(s)
- Oguz Lafci
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Daphne Resch
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Ambra Santonocito
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Paola Clauser
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Thomas Helbich
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Pascal A T Baltzer
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
2
|
Zhou S, Zhang Y, Belmar J, Hou C, Zhang Y, Peng C, Meng Y, Li Z, Mughal MJ, Gao Y, Seto E, Shen M, Hall MD, Ma J, Ma CX, Li S, Zhu W. Stabilization of RUNX1 Induced by O-GlcNAcylation Promotes PDGF-BB-Mediated Resistance to CDK4/6 Inhibitors in Breast Cancer. Cancer Res 2025; 85:1708-1724. [PMID: 39937190 DOI: 10.1158/0008-5472.can-24-2492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/01/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4/6) are crucial in regulating cell-cycle progression and cancer development. Targeting CDK4/6 has shown considerable promise in treating various cancers, including breast cancer. Despite significant therapeutic efficacy, resistance to CDK4/6 inhibitors (CDK4/6i), such as palbociclib, remains a substantial hurdle in clinical practice. Using a coculture system, cytokine array, and quantitative high-throughput combinatorial screening, we discovered a mechanism by which the Runt-related transcription factor (RUNX) 1-platelet-derived growth factor (PDGF)-BB axis regulates palbociclib resistance in breast cancer cells. Specifically, RUNX1 functioned as a transcription factor to drive expression of PDGFB, leading to resistance to palbociclib by enhancing the Akt pathway and suppressing senescence. Furthermore, in resistant cells, RUNX1 was O-GlcNAcylated at serine 252 by O-GlcNAc transferase, resulting in the stabilization of RUNX1 by preventing ubiquitin-mediated degradation. Inhibition of the RUNX1-PDGF-BB axis by specific inhibitors overcame palbociclib resistance both in vitro and in vivo. Notably, the RUNX1-PDGF-BB axis was upregulated in resistant patient-derived xenograft lines and in patients with breast cancer following treatment with CDK4/6i. These findings not only unveil O-GlcNAcylation-mediated activation of a RUNX1-PDGF-BB pathway as a driver of palbociclib resistance but also provide clinical evidence supporting the repurposing of FDA-approved PDGFR inhibitors as a therapeutic strategy to treat patients with CDK4/6i-resistant breast cancer. Significance: RUNX1-PDGF-BB signaling drives resistance to CDK4/6 inhibition in breast cancer, providing the foundation to develop approaches to target the RUNX1-PDGF-BB axis to overcome CDK4/6 inhibitor resistance in breast cancer patients.
Collapse
Affiliation(s)
- Shuyan Zhou
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Yi Zhang
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Julie Belmar
- Department of Medicine, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Chunyan Hou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Yaqin Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland
| | - Changmin Peng
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Yunxiao Meng
- Laboratory & Molecular and Genomic Pathology, Department of Laboratory and Transfusion Services, The George Washington University Hospital, Washington, District of Columbia
| | - Zhuqing Li
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Muhammad Jameel Mughal
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Yanjun Gao
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Edward Seto
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Min Shen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland
| | - Matthew D Hall
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland
| | - Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine in St Louis, St. Louis, Missouri
| | - Shunqiang Li
- Department of Medicine, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| |
Collapse
|
3
|
Guo Z, Dong RW, Wu Y, Dong S, Alahari SK. Cyclin-dependent kinase 4 and 6 inhibitors in breast cancer treatment. Oncogene 2025; 44:1135-1152. [PMID: 40200094 DOI: 10.1038/s41388-025-03378-0] [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: 11/13/2024] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Breast cancer is the second largest cancer in the world, and it has highest mortality rate in women worldwide. The aberrant activation of the cyclin-dependent kinase 4 and 6 (CDK4/6) pathway plays an important role in uncontrolled breast cancer cell proliferation. Therefore, targeting CDK4/6 to improve overall survival rates has been a strong interest in breast cancer therapeutics. Till date, four CDK4/6 inhibitors have been developed and approved for hormone receptor-positive and human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer therapies with great success. However, acquired resistance to CDK4/6 inhibitors has emerged and limits their effectiveness in breast cancer. In this review, we systematically discussed the mechanisms of resistance to CDK4/6 inhibitors including the cell cycle-specific and cell cycle-nonspecific mechanisms. Also, we analyzed combination strategies with other signaling inhibitors in clinical and preclinical settings that further expand the clinical application of CDK4/6 inhibitors in future breast cancer therapies.
Collapse
Affiliation(s)
- Zhengfei Guo
- TYK Medicines, Inc., Huzhou, Zhejiang, 313100, China
| | - Richard W Dong
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Yusheng Wu
- TYK Medicines, Inc., Huzhou, Zhejiang, 313100, China
| | - Shengli Dong
- TYK Medicines, Inc., Huzhou, Zhejiang, 313100, China.
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
4
|
Niraula S. Adjuvant CDK4/6 inhibitors in breast cancer: Interpreting trial design, evidence, and uncertainty. Cancer Treat Rev 2025; 136:102944. [PMID: 40262369 DOI: 10.1016/j.ctrv.2025.102944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors have transformed the treatment landscape for metastatic hormone receptor-positive, HER2-negative breast cancer by improving progression-free and Overall Survival (OS). In the adjuvant context, however, results have been discordant and remain immature. The PALLAS and PENELOPE-B trials of palbociclib reported no benefit, while monarchE and NATALEE demonstrated improvements in invasive disease-free survival (iDFS) with abemaciclib and ribociclib, respectively, leading to regulatory approvals despite no demonstrated OS benefit yet. It remains possible that adjuvant CDK4/6 inhibition provides meaningful long-term benefit, but that has not been demonstrated. Concerns related to trial design: risk-enrichment, open-label conduct, high treatment-discontinuation rates, and potential informative censoring complicate interpretation. Although iDFS is a recognized intermediate endpoint with potential psychological validity, it is subject to bias in collection and communication, and has not been validated as a surrogate for OS in this setting. Moreover, early inhibition of CDK4/6 may induce resistance and compromise subsequent efficacy. Reported quality-of-life outcomes were preserved, not improved, which holds limited value considering added toxicity, inconvenience, and cost in a largely curable population. If even half of eligible patients are treated, estimated annual costs in the United States would exceed $7 billion. As these agents are incorporated into clinical guidelines, it is critical to clarify whether they improve long-term outcomes, delay recurrence without affecting survival, or cause unintended harm. Impulse to intervene early is understandable, but emerging data must be carefully assessed to ensure adjuvant CDK4/6 inhibition offers meaningful benefit to patients and health systems.
Collapse
Affiliation(s)
- Saroj Niraula
- University of British Columbia, Medical Oncologist, BC Cancer, British Columbia, Canada.
| |
Collapse
|
5
|
Witkiewicz AK, Wang J, Schultz E, O'Connor TN, O'Connor T, Levine E, Knudsen ES. Using prognostic signatures and machine learning to identify core features associated with response to CDK4/6 inhibitor-based therapy in metastatic breast cancer. Oncogene 2025; 44:1387-1399. [PMID: 40011574 DOI: 10.1038/s41388-025-03308-0] [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: 08/13/2024] [Revised: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
CDK4/6 inhibitors in combination with endocrine therapy are widely used to treat HR+/HER2- metastatic breast cancer leading to improved progression-free survival (PFS) compared to single agent endocrine therapy. Over 300 patients receiving standard-of-care CDK4/6 inhibitor combination therapy for metastatic disease were enrolled at a single institution. Clinical, pathological, and gene expression data were employed to define determinants for PFS duration. Visceral disease (HR 1.55, p = 0.0013), prior endocrine therapy (HR 2.34, p < 0.001), and the type of endocrine therapy (HR 2.16, p < 0.001) were highly associated with PFS duration. Multiple pre-defined gene expression signatures were employed to determine association with response to CDK4/6 inhibitor-based therapy. Random survival forest was applied to define key gene expression and clinical features associated with PFS and develop a predictive model. The time to progression predicted by this model was related to the median PFS observed in PALOMA-2/3 and PEARL studies. Interrogating genes identified as highly significant across all studies indicated common enrichment of gene networks associated with cell cycle and estrogen receptor signaling. These findings indicate that there are common features from real-world use of CDK4/6 inhibitors that could be used to infer time to progression and better inform treatment.
Collapse
Affiliation(s)
- Agnieszka K Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA.
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA.
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Emily Schultz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Thomas N O'Connor
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Tracey O'Connor
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Ellis Levine
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA.
| |
Collapse
|
6
|
Paiva CE, Guimarães VS, Silva ATF, Maia YCDP, Paiva BSR, Reinert T, LeVasseur N. Navigating Endocrine Sensitivity Assessment in Nonmetastatic Breast Cancer Through Early On-Treatment Ki67 Understanding. Clin Breast Cancer 2025:S1526-8209(25)00111-9. [PMID: 40413085 DOI: 10.1016/j.clbc.2025.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 04/04/2025] [Accepted: 04/28/2025] [Indexed: 05/27/2025]
Abstract
In the landscape of breast cancer management, precise assessment of endocrine sensitivity significantly influences therapeutic strategies. The utilization of Ki67, an immunohistochemical marker of tumor proliferation, in tumor rebiopsy holds paramount importance in unraveling breast cancer's response to endocrine therapy. This narrative review aims to elucidate the pivotal role of Ki67 in endocrine sensitivity assessment. We explore the nuanced process of Ki67 evaluation, its timing, and its implications for clinical outcomes in breast cancer patients. From a clinical perspective to pathological response and risk of recurrence, we navigate the spectrum of Ki67's impact. By delving into these facets, we underscore Ki67's potential to guide personalized treatment strategies, shedding light on the intricate interplay between endocrine therapy and clinical outcomes in breast cancer patients.
Collapse
Affiliation(s)
- Carlos Eduardo Paiva
- Department of Clinical Oncology, Barretos Cancer Hospital, Barretos, SP, Brazil; 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
| | - Alinne Tatiane Faria Silva
- Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, Barretos, SP, Brazil
| | - Yara Cristina de Paiva Maia
- Nutrition and Molecular Biology Research Group, School of Medicine, Federal University of Uberlandia, Minas Gerais, Uberlandia, Brazil
| | - Bianca Sakamoto Ribeiro Paiva
- Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, Barretos, SP, Brazil; Learning and Research Institute, Barretos Cancer Hospital, Barretos, SP, Brazil
| | - Tomás Reinert
- Centro de Oncologia Família Irineu Boff, Hospital Nora Teixeira, Santa Casa de Porto Alegre, Porto Alegre, RS, Brazil; Grupo Brasileiro de Estudos em Câncer de Mama (GBECAM), Porto Alegre, RS, Brazil
| | - Nathalie LeVasseur
- Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
Jasim SA, Altalbawy FMA, Uthirapathy S, Bishoyi AK, Ballal S, Singh A, Devi A, Yumashev A, Mustafa YF, Abosaoda MK. Regulation of immune-mediated chemoresistance in cancer by lncRNAs: an in-depth review of signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04081-3. [PMID: 40202675 DOI: 10.1007/s00210-025-04081-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Resistance to cancer therapies is increasingly recognized as being influenced by long non-coding RNAs (lncRNAs), which are pivotal in regulating cellular functions and gene expression. Elucidating the intricate relationship between lncRNAs and the mechanisms underlying drug resistance is critical for advancing effective therapeutic strategies. This study offers an in-depth review of the regulatory roles lncRNAs play in various signaling and immunological pathways implicated in cancer chemoresistance. lncRNA-mediated influence on drug resistance-related signaling pathways will be presented, including immune evasion mechanisms and other essential signaling cascades. Furthermore, the interplay between lncRNAs and the immune landscape will be dissected, illustrating their substantial impact on the development of chemoresistance. Overall, the potential of lncRNA-mediated signaling networks as a therapeutic strategy to combat cancer resistance has been highlighted. This review reiterates the fundamental role of lncRNAs in chemoresistance and proposes promising avenues for future research and the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq.
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Mosco, Russia
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
8
|
Iacovacci J, Brough R, Moughari FA, Alexander J, Kemp H, Tutt ANJ, Natrajan R, Lord CJ, Haider S. Proteogenomic discovery of RB1-defective phenocopy in cancer predicts disease outcome, response to treatment, and therapeutic targets. SCIENCE ADVANCES 2025; 11:eadq9495. [PMID: 40138429 PMCID: PMC11939072 DOI: 10.1126/sciadv.adq9495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Genomic defects caused by truncating mutations or deletions in the Retinoblastoma tumor suppressor gene (RB1) are frequently observed in many cancer types leading to dysregulation of the RB pathway. Here, we propose an integrative proteogenomic approach that predicts cancers with dysregulation in the RB pathway. A subset of these cancers, which we term as "RBness," lack RB1 genomic defects and yet phenocopy the transcriptional profile of RB1-defective cancers. We report RBness as a pan-cancer phenomenon, associated with patient outcome and chemotherapy response in multiple cancer types, and predictive of CDK4/6 inhibitor response in estrogen-positive breast cancer. Using RNA interference and a CRISPR-Cas9 screen in isogenic models, we find that RBness cancers also phenocopy synthetic lethal vulnerabilities of cells with RB1 genomic defects. In summary, our findings suggest that dysregulation of the RB pathway in cancers lacking RB1 genomic defects provides a molecular rationale for how these cancers could be treated.
Collapse
Affiliation(s)
- Jacopo Iacovacci
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano 20133, Italy
| | - Rachel Brough
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Fatemeh Ahmadi Moughari
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - John Alexander
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Harriet Kemp
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Andrew N. J. Tutt
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Christopher J. Lord
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
9
|
Luo H, Sun Y, Xu T. Application status and research progress of targeted therapy drugs for hormone receptor-positive breast cancer. Front Med (Lausanne) 2025; 12:1513836. [PMID: 40134916 PMCID: PMC11933059 DOI: 10.3389/fmed.2025.1513836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/10/2025] [Indexed: 03/27/2025] Open
Abstract
Breast cancer (BC) is the most common malignant tumor in women and the leading cause of cancer-related deaths in women. As one of the most common subtypes of breast cancer, patients with hormone receptor-positive (HR+) breast cancer usually experience disease progression over an extended period of time, triggering the search for therapeutic strategies other than endocrine therapy. In recent years, continuous research on various targets has led to dramatic changes in the treatment of hormone receptor-positive breast cancer patients, resulting in prolonged clinical survival. With the redefinition of human epidermal growth factor-2 (HER2) expression, more precise and individualized treatment is possible. This review comprehensively reviews targeted therapies and critical clinical trials for HR+ breast cancer and tracks the latest advances. It also provides valuable insights into the future direction of targeted therapies.
Collapse
Affiliation(s)
- Han Luo
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
| | - Yue Sun
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
| | - Tiefeng Xu
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
10
|
Brumer RP, Angarita FA. Perioperative Considerations of Breast Cancer Neoadjuvant Treatments. Clin Breast Cancer 2025:S1526-8209(25)00050-3. [PMID: 40240236 DOI: 10.1016/j.clbc.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 04/18/2025]
Abstract
Neoadjuvant treatment is increasingly being used in patients with breast cancer. Understanding the effects of neoadjuvant agents in the perioperative setting is crucial as it can affect morbidity. Understanding how these agents should be used to maximize optimal time to surgery is crucial as delays can negatively affective cancer outcomes. This review aims to discuss the perioperative considerations surgeons should be aware of prior to scheduling surgery for patients receiving chemotherapy, endocrine therapy, chemotherapy, targeted therapy, and immunotherapy.
Collapse
Affiliation(s)
- Robert P Brumer
- Department of Surgery, Houston Methodist Hospital, Houston, TX
| | - Fernando A Angarita
- Department of Surgery, Houston Methodist Hospital, Houston, TX; Division of Surgical Oncology and Gastrointestinal Surgery, Department of Surgery, Houston Methodist Hospital, Houston, TX.
| |
Collapse
|
11
|
Zhang L, Liu Y, Yang C, Ma J, Li Y, Luo R, Han J, Wang X, Zhang Z, Ma L, Cai H, Kong X, Wang Z, Zhou X, Shi J, Zhang Y, Wang M, Wang J, Geng C. Clinical efficacy and therapy response prediction of neoadjuvant dalpiciclib plus letrozole in postmenopausal patients with HR+/HER2- stage II-III breast cancer (DARLING 01): a single-arm, open-label, exploratory study. Breast Cancer Res 2025; 27:21. [PMID: 39948624 PMCID: PMC11827216 DOI: 10.1186/s13058-025-01976-0] [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: 12/04/2024] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) breast cancer is the most common subtype of breast cancer, yet its response to traditional chemotherapy remains limited, posing a challenge in achieving optimal therapeutic outcomes. In this study, we aimed to evaluate the clinical efficacy and safety of dalpiciclib, a novel CDK4/6 inhibitor, combined with letrozole as neoadjuvant therapy (NAT) in postmenopausal patients with HR+/HER2- stage II-III breast cancer. Additionally, we explored potential predictive biomarkers for treatment response using gene analysis. METHODS This single-arm, open-label, exploratory phase II trial involved 35 postmenopausal women with HR+/HER2- breast cancer (ClinicalTrials.gov identifier NCT05512780). Patients received four cycles of dalpiciclib (125 mg/day for 3 weeks, followed by 1 week off) plus continuous letrozole (2.5 mg/day). The primary endpoint was objective response rate (ORR), and secondary endpoints included changes in Ki-67 expression, complete cell cycle arrest (CCCA) rate, residual cancer burden (RCB), and safety profiles. Gene expression profiling and least absolute shrinkage and selection operator (LASSO) regression were conducted to identify biomarkers predictive of response to NAT. RESULTS Among the 35 enrolled patients, 31 completed the full treatment course. Of the 29 patients with evaluable response data after 4 cycles, 16 achieved partial response (PR), resulting in an ORR of 55.2%. Following two weeks of treatment, the mean Ki-67 expression significantly decreased from a baseline of 17.5-1.8%, and CCCA was observed in 75% of patients. Grade ≥ 3 treatment-emergent adverse events (TEAEs) were mainly decreased neutrophil count (45.7%), with a median duration of 3 days. The NAT predictive model, developed using gene expression analysis and clinicopathological factors, achieved an area under the curve (AUC) of 0.928, indicating that TFRC, SCUBE2, and MMP11A could serve as novel predictive biomarkers for response to NAT. CONCLUSIONS Dalpiciclib combined with letrozole demonstrated promising antitumor activity and an acceptable safety profile in postmenopausal patients with HR+/HER2- breast cancer. The identification of TFRC, SCUBE2, and MMP11A as predictive biomarkers provides insights into the potential for personalized neoadjuvant treatment strategies.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Hebei, 050011, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Hebei, China
| | - Chao Yang
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Hebei, 050011, China
| | - Jie Ma
- Department of Breast Surgery, TangShan People's Hospital, Tangshan, China
| | - Yuntao Li
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Hebei, 050011, China
| | - Ruizhen Luo
- Department of Breast Surgery, Hebei Province CangZhou Hospital of Integrated Traditional and Western Medicine, Hebei, China
| | - Jianjun Han
- Department of Breast Surgery, Affiliated Hospital of Hebei University of Engineering, Hebei, China
| | - Xiaochun Wang
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Hebei, China
| | - Zhisheng Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Hebei North University, Hebei, China
| | - Li Ma
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Hebei, 050011, China
| | - Haifeng Cai
- Department of Breast Surgery, TangShan People's Hospital, Tangshan, China
| | - Xiangshun Kong
- Department of Breast Surgery, Xingtai People's Hospital, Hebei, China
| | - Zunyi Wang
- Department of Breast Surgery, Cangzhou Central Hospital, Hebei, China
| | - Xinping Zhou
- Department of General surgery, HanDan Central Hospital, Hebei, China
| | - Jiajie Shi
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Hebei, 050011, China
| | - Yanshou Zhang
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Hebei, 050011, China
| | - Meiqi Wang
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Hebei, 050011, China
| | - Jiaxing Wang
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Hebei, 050011, China
| | - Cuizhi Geng
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Hebei, 050011, China.
| |
Collapse
|
12
|
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
|
13
|
Shanabag A, Armand J, Son E, Yang HW. Targeting CDK4/6 in breast cancer. Exp Mol Med 2025; 57:312-322. [PMID: 39930131 PMCID: PMC11873051 DOI: 10.1038/s12276-025-01395-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 03/04/2025] Open
Abstract
Dysregulation of the cell cycle machinery, particularly the overactivation of cyclin-dependent kinases 4 and 6 (CDK4/6), is a hallmark of breast cancer pathogenesis. The introduction of CDK4/6 inhibitors has transformed the treatment landscape for hormone receptor-positive breast cancer by effectively targeting abnormal cell cycle progression. However, despite their initial clinical success, drug resistance remains a significant challenge, with no reliable biomarkers available to predict treatment response or guide strategies for managing resistant populations. Consequently, numerous studies have sought to investigate the mechanisms driving resistance to optimize the therapeutic use of CDK4/6 inhibitors and improve patient outcomes. Here we examine the molecular mechanisms regulating the cell cycle, current clinical applications of CDK4/6 inhibitors in breast cancer, and key mechanisms contributing to drug resistance. Furthermore, we discuss emerging predictive biomarkers and highlight potential directions for overcoming resistance and enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Anusha Shanabag
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Jessica Armand
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Eugene Son
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Hee Won Yang
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
| |
Collapse
|
14
|
Mughal MJ, Zhang Y, Li Z, Zhou S, Peng C, Zhang YQ, Seto E, Shen M, Hall MD, Zhu W. TFAP2C-DDR1 axis regulates resistance to CDK4/6 inhibitor in breast cancer. Cancer Lett 2025; 610:217356. [PMID: 39603379 PMCID: PMC11783577 DOI: 10.1016/j.canlet.2024.217356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Breast cancer is the predominant malignancy with the majority of cases are characterized as HR+/HER2-subtype. Although cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) have shown remarkable efficacy in treating this subtype when combined with endocrine therapy, the development of resistance to these inhibitors remains a significant clinical obstacle. Hence, there is an urgent need to explore innovative therapies and decipher the underlying mechanisms of resistance to CDK4/6i. In this study, we employed quantitative high-throughput combination screening (qHTCS) and genomics/proteomics approaches to uncover the molecular mechanisms driving resistance to CDK4/6i (palbociclib) in breast cancer. The comprehensive analyses revealed DDR1 as a potential factor implicated in mediating resistance to CDK4/6i. Specifically, DDR1 inhibition in combination with palbociclib exhibited remarkable synergistic effects, reducing cell survival signaling and promoting apoptosis in resistant cells. In-vivo xenograft model further validated the synergistic effects, showing a significant reduction in the resistant tumor growth. Exploration into DDR1 activation uncovered TFAP2C as a key transcription factor regulating DDR1 expression in palbociclib resistant cells and inhibition of TFAP2C re-sensitized resistant cells to palbociclib. Gene set enrichment analysis (GSEA) in the NeoPalAna trial demonstrated a significant enrichment of the TFAP2C-DDR1 gene set from patitens after palbociclib treatment, suggesting the possible activation of the TFAP2C-DDR1 axis following palbociclib exposure. Overall, this study provides crucial insights into the novel molecular landscape of palbociclib resistance in breast cancer, suggesting TFAP2C-DDR1 axis inhibition as a promising strategy to overcome resistance.
Collapse
Affiliation(s)
- Muhammad Jameel Mughal
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Yi Zhang
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Zhuqing Li
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Shuyan Zhou
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Changmin Peng
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Ya-Qin Zhang
- Early Translation Branch, Division of Preclinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, 20850, USA
| | - Edward Seto
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Min Shen
- Early Translation Branch, Division of Preclinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, 20850, USA
| | - Matthew D Hall
- Early Translation Branch, Division of Preclinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, 20850, USA
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA.
| |
Collapse
|
15
|
Foffano L, Cucciniello L, Nicolò E, Migliaccio I, Noto C, Reduzzi C, Malorni L, Cristofanilli M, Gerratana L, Puglisi F. Cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i): Mechanisms of resistance and where to find them. Breast 2025; 79:103863. [PMID: 39718288 PMCID: PMC11872392 DOI: 10.1016/j.breast.2024.103863] [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: 10/12/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024] Open
Abstract
CDK4/6 inhibitors (CDK4/6i) have significantly impacted on the treatment of HR + HER2 negative (HER2-) metastatic breast cancer (BC) when combined with endocrine therapy. Nonetheless, despite significant research efforts, the mechanisms of de novo and acquired resistance to CDK4/6i have not yet been fully elucidated, highlighting the need for a deeper understanding of these process. Additionally, the importance of dissecting CDK4/6i resistance from endocrine resistance for personalized treatment is increasingly recognized. Liquid biopsy has emerged as a minimally invasive tool for identifying circulating biomarkers of resistance through the integration of multiparametric and dynamic assessments that encompass ctDNA, CTCs, exosomes, and epigenetic ctDNA alterations, representing a promising perspective for the clinical characterization of treatment resistance and guiding post-progression strategies to improve patient outcomes. Aim of this review is summarize potential mechanisms of CDK4/6i resistance, along with the advantages of using liquid biopsy to identify resistance biomarkers in HR+/HER2- MBC patients treated with CDK 4/6 inhibitors.
Collapse
Affiliation(s)
- L Foffano
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - L Cucciniello
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - E Nicolò
- Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
| | - I Migliaccio
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, 59100, Prato, Italy
| | - C Noto
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy; Department of Medicine, University of Udine, Udine, Italy
| | - C Reduzzi
- Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - L Malorni
- "Sandro Pitigliani" Translational Research Unit, Hospital of Prato, 59100, Prato, Italy; "Sandro Pitigliani" Medical Oncology Department, Hospital of Prato, 59100, Prato, Italy
| | - M Cristofanilli
- Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - L Gerratana
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy.
| | - F Puglisi
- Department of Medical Oncology. CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy; Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|
16
|
Kindt CK, Ehmsen S, Traynor S, Policastro B, Nissen N, Jakobsen MK, Hundebøl MF, Johansen LE, Bak M, Arbajian E, Staaf J, Ditzel HJ, Alves CL. RET inhibition overcomes resistance to combined CDK4/6 inhibitor and endocrine therapy in ER+ breast cancer. Front Oncol 2025; 14:1497093. [PMID: 39931212 PMCID: PMC11808005 DOI: 10.3389/fonc.2024.1497093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/18/2024] [Indexed: 02/13/2025] Open
Abstract
Background Combined CDK4/6 inhibitor (CDK4/6i) and endocrine therapy significantly improve the outcome of patients with advanced estrogen receptor-positive (ER+) breast cancer. However, resistance to this treatment and disease progression remains a major clinical challenge. High expression of the receptor tyrosine kinase REarranged during Transfection (RET) has been associated with resistance to endocrine therapy in breast cancer, but the role of RET in CDK4/6i treatment response/resistance remains unexplored. Methods To identify gene expression alterations associated with resistance to combined endocrine therapy and CDK4/6i, we performed RNA sequencing of two ER+ breast cancer cell models resistant to this combined therapy. The functional role of RET was assessed by siRNA-mediated RET silencing and targeted inhibition with the FDA/EMA-approved RET-selective inhibitor selpercatinib in resistant breast cancer cells and patient-derived organoids (PDOs). RET silencing was evaluated mechanistically using global gene expression and pathway analysis. The clinical relevance of RET expression in ER+ breast cancer was investigated by gene array analysis of primary tumors treated with endocrine therapy and by immunohistochemical scoring of metastatic lesions from patients who received combined CDK4/6i and endocrine therapy. Results We show that RET is upregulated in ER+ breast cancer cell lines resistant to combined CDK4/6i and fulvestrant compared to isogenic cells resistant to fulvestrant alone. siRNA-mediated silence of RET in high RET-expressing, combined CDK4/6i- and fulvestrant-resistant cells reduced their growth partially by affecting cell cycle regulators of the G2-M phase and E2F targets. Notably, targeting RET with selpercatinib in combination with CDK4/6i inhibited the growth of CDK4/6i-resistant cell lines and resensitized ER+ breast cancer patient-derived organoids resistant to CDK4/6i. Finally, analysis of RET expression in ER+ breast cancer patients treated with endocrine therapy showed that high RET expression correlated with poor clinical outcomes. We further observed a shorter median survival to combined CDK4/6i and endocrine therapy in patients with RET-positive compared to RET-negative tumors, but this difference did not reach statistical significance. Conclusions Our findings show that RET is overexpressed in ER+ metastatic breast cancer resistant to combined CDK4/6i and endocrine therapy, rendering RET inhibition a promising therapeutic approach for patients who experience disease progression on combined CDK4/6i and endocrine therapy.
Collapse
Affiliation(s)
- Charlotte K. Kindt
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sidse Ehmsen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Sofie Traynor
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Benedetta Policastro
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Nikoline Nissen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Mie K. Jakobsen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Monique F. Hundebøl
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lene E. Johansen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Martin Bak
- Department of Pathology, Sydvestjysk Sygehus, Esbjerg, Denmark
| | - Elsa Arbajian
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Henrik J. Ditzel
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Carla L. Alves
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
17
|
Raghavendra AS, Kettner NM, Kwiatkowski D, Damodaran S, Wang Y, Ramirez D, Gombos DS, Hunt KK, Shen Y, Keyomarsi K, Tripathy D. Phase I trial of hydroxychloroquine to enhance palbociclib and letrozole efficacy in ER+/HER2- breast cancer. NPJ Breast Cancer 2025; 11:7. [PMID: 39865083 PMCID: PMC11770068 DOI: 10.1038/s41523-025-00722-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/13/2025] [Indexed: 01/28/2025] Open
Abstract
Endocrine therapy with CDK4/6 inhibitors is standard for estrogen receptor-positive, HER2-negative metastatic breast cancer (ER+/HER2- MBC), yet clinical resistance develops. Previously, we demonstrated that low doses of palbociclib activate autophagy, reversing initial G1 cell cycle arrest, while high concentrations induce off-target senescence. The autophagy inhibitor hydroxychloroquine (HCQ) induced on-target senescence at lower palbociclib doses. We conducted a phase I trial (NCT03774472 registered in ClinicalTrials.gov on 8/20/2018) of HCQ (400, 600, 800 mg/day) with palbociclib (75 mg/day continuous) and letrozole, using a 3 + 3 design. Primary objectives included safety, tolerability, and determining the recommended phase 2 dose (RP2D) of HCQ. Secondary objectives included tumor response and biomarker analysis. Fourteen ER+/HER2- MBC patients were evaluable [400 mg (n = 4), 600 mg (n = 4), 800 mg (n = 6)]. Grade 3 adverse events (AEs) included hematological (3 at 800 mg), skin rash (2 at 600 mg), and anorexia (1 at 400 mg), with no serious AEs. The best responses were partial (2), stable (11), and progression (1). Tumor reductions ranged from 11% to 30%, with one 55% increase. The two partial responders sustained tumor size reductions of 30% to 55% over an extended treatment period, lasting nearly 300 days. Biomarker analysis in responders demonstrated significant decreases in Ki67, Rb, and nuclear cyclin E levels and increases in autophagy markers p62 and LAMP1, suggesting a correlation between these biomarkers and treatment response. This phase I study demonstrated that HCQ is safe and well-tolerated and the RP2D was established at 800 mg/day with continuous low-dose palbociclib (75 mg/day) and letrozole (2.5 mg/day). These findings suggest that adding HCQ could potentially enhance the efficacy of low-dose palbociclib and standard letrozole therapy, pending verification in larger randomized studies.
Collapse
Affiliation(s)
| | - Nicole M Kettner
- Department of Experimental Radiation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Danielle Kwiatkowski
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Senthil Damodaran
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yan Wang
- Department of Experimental Radiation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Ramirez
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dan S Gombos
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly K Hunt
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yu Shen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Khandan Keyomarsi
- Department of Experimental Radiation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
18
|
Pellarin I, Dall'Acqua A, Favero A, Segatto I, Rossi V, Crestan N, Karimbayli J, Belletti B, Baldassarre G. Cyclin-dependent protein kinases and cell cycle regulation in biology and disease. Signal Transduct Target Ther 2025; 10:11. [PMID: 39800748 PMCID: PMC11734941 DOI: 10.1038/s41392-024-02080-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/16/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025] Open
Abstract
Cyclin Dependent Kinases (CDKs) are closely connected to the regulation of cell cycle progression, having been first identified as the kinases able to drive cell division. In reality, the human genome contains 20 different CDKs, which can be divided in at least three different sub-family with different functions, mechanisms of regulation, expression patterns and subcellular localization. Most of these kinases play fundamental roles the normal physiology of eucaryotic cells; therefore, their deregulation is associated with the onset and/or progression of multiple human disease including but not limited to neoplastic and neurodegenerative conditions. Here, we describe the functions of CDKs, categorized into the three main functional groups in which they are classified, highlighting the most relevant pathways that drive their expression and functions. We then discuss the potential roles and deregulation of CDKs in human pathologies, with a particular focus on cancer, the human disease in which CDKs have been most extensively studied and explored as therapeutic targets. Finally, we discuss how CDKs inhibitors have become standard therapies in selected human cancers and propose novel ways of investigation to export their targeting from cancer to other relevant chronic diseases. We hope that the effort we made in collecting all available information on both the prominent and lesser-known CDK family members will help in identify and develop novel areas of research to improve the lives of patients affected by debilitating chronic diseases.
Collapse
Affiliation(s)
- Ilenia Pellarin
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Alessandra Dall'Acqua
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Andrea Favero
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Ilenia Segatto
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Valentina Rossi
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Nicole Crestan
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Javad Karimbayli
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Barbara Belletti
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy.
| |
Collapse
|
19
|
Yu S, Si Y, Xu M, Wang Y, Liu C, Bi C, Sun M, Sun H. Downregulation of the splicing regulator NSRP1 confers resistance to CDK4/6 inhibitors via activation of interferon signaling in breast cancer. J Biol Chem 2025; 301:108070. [PMID: 39667501 PMCID: PMC11750474 DOI: 10.1016/j.jbc.2024.108070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/16/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024] Open
Abstract
The combination of CDK4/6 inhibitors (CDK4/6i) and endocrine therapy is the first-line therapy for ER+/Her2-breast cancer; however, the development of drug resistance limited the efficacy of the agents. Although activation of the IFN signaling pathway has been identified as a critical driver of intrinsic and acquired CDK4/6i resistance, it remains unknown how the IFN signaling pathway was activated in resistant cells. Here, we report that NSRP1, a regulator of alternative mRNA splicing is downregulated in CDK4/6i resistant breast cancer cells and contributes to CDK4/6i resistance by mediating alternative splicing of NSD2 mRNA and activation of the IFN signaling pathway. Knockdown of NSRP1 reduces CDK4/6i (palbociclib) sensitivity of MCF7 cells while overexpression of NSRP1 sensitizes MCF7-PalR cells towards palbociclib treatment. Mechanistically, RNA sequencing suggests that NSRP1 knockdown strongly activates the IFN signaling pathway in MCF7 cells and elevates the expression of most of the "IFN-related palbociclib-resistance Signature" (IRPS) genes. NSRP1 also regulates numerous alternative splicing (AS) events in breast cancer cells, several of which are key regulators of the IFN signaling pathway. Among them, the inclusion of NSD2 exon 2 is elevated upon NSRP1 knockdown. Our data further show that the inclusion of NSD2 exon 2 is increased in breast cancer and associated with the poor prognosis of patients. In addition, including NSD2 exon 2 elevates NSD2 protein expression to activate the IFN signaling pathway. This study unveils the critical role of NSRP1 in regulating the IFN signaling pathway and the CDK4/6i resistance, which could be a promising biomarker for predicting therapy response.
Collapse
Affiliation(s)
- Shiyi Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yue Si
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Miao Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Wang
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Chengxu Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Caili Bi
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China
| | - Maoqiu Sun
- Department of Obstetrics and Gynecology, Haian Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nantong, Jiangsu, China.
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
20
|
Migliaccio I, Guarducci C, Malorni L. CDK4/6 Inhibitor Resistance in ER+ Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:475-493. [PMID: 39821039 DOI: 10.1007/978-3-031-70875-6_23] [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
The cyclin-dependent kinases 4 and 6 inhibitors are the mainstay of treatment for patients with hormone receptor-positive and HER2-negative breast cancer. The ability of these drugs to improve the outcome of patients both in the metastatic and the early setting has been largely demonstrated. However, resistance, either de novo or acquired, represents a major clinical challenge. In the past years, efforts have been made to identify biomarkers that might help in a better selection of patients or to unravel the mechanisms leading to resistance in order to develop new therapeutic strategies to overcome it. Alterations of cell cycle-related genes and proteins are among the best characterized markers of resistance, and pathways impacting the cell cycle, including nuclear and growth factor receptors signaling, have been thoroughly investigated. Despite this, to date, cyclin-dependent kinases 4 and 6 inhibitors are administered based only on the hormone receptor and HER2 status of the tumor, and patients progressing on therapy are managed with currently available treatments. Here we summarize present knowledge on the cyclin-dependent kinases 4 and 6 inhibitors' mechanisms of action, efficacy data, and mechanisms of resistance.
Collapse
Affiliation(s)
- Ilenia Migliaccio
- Translational Research Unit, Hospital of Prato, AUSL Toscana Centro, Prato, Italy
| | - Cristina Guarducci
- Translational Research Unit, Hospital of Prato, AUSL Toscana Centro, Prato, Italy
| | - Luca Malorni
- Translational Research Unit, Hospital of Prato, AUSL Toscana Centro, Prato, Italy.
| |
Collapse
|
21
|
Cui J, Sun J, Zhou X, Li Y, Zhao J, Shen G. Association between cyclin-dependent kinase 4/6 inhibitors and nephrotoxicity in patients with breast cancer: A Systematic Review and meta-analysis. iScience 2024; 27:111370. [PMID: 39640578 PMCID: PMC11618030 DOI: 10.1016/j.isci.2024.111370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/20/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
Nephrotoxic adverse events (AEs) have been observed in patients with breast cancer receiving cyclin-dependent kinase (CDK) 4/6 inhibitors. This study aimed to evaluate the risk of nephrotoxicity associated with these inhibitors through a meta-analysis of 17 randomized controlled trials involving 19,638 patients. The results indicate a significant increase in all-grade nephrotoxic AEs, including elevated blood creatinine levels, acute kidney injury, and renal impairment (RR = 3.12, 95% CI [2.11, 4.63]). The incidence of grade 3 or higher nephrotoxicity was also more prevalent among treated patients (RR = 3.12, 95% CI [1.74, 5.58]). Subgroup analyses revealed varying risks among 4 different CDK 4/6 inhibitors. Furthermore, analysis of FDA Adverse Event Reporting System data corroborated these findings, emphasizing the occurrence of nephrotoxicity in real-world settings. Clinicians should remain vigilant in monitoring renal function indicators when prescribing CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Jiayong Cui
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Xining, China
| | - Jinquan Sun
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Xining, China
| | - Xueying Zhou
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Xining, China
| | - Yi Li
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Xining, China
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Xining, China
| | - Guoshuang Shen
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Xining, China
| |
Collapse
|
22
|
Neven P, Stahl N, Vidal M, Martín M, Kaufman PA, Harbeck N, Hunt KK, Carter S, Bidard FC, Fasching PA, Aftimos P, Wheatley D, Hamilton E, Aft R, Kulkarni S, Schmid P, Bhave M, Ismail-Khan R, Karacsonyi C, Estrem ST, Nguyen B, Ozbek U, Yuen E, Rodrik-Outmezguine V, Ciruelos E. A Preoperative Window-of-Opportunity Study of Oral SERD, Imlunestrant, in Newly Diagnosed ER-Positive, HER2-Negative Early Breast Cancer: Results from the EMBER-2 Study. Clin Cancer Res 2024; 30:5304-5313. [PMID: 39377773 PMCID: PMC11609621 DOI: 10.1158/1078-0432.ccr-24-2113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE Imlunestrant is an oral selective estrogen receptor degrader with favorable safety and preliminary efficacy in patients with advanced breast cancer. Pharmacodynamic (PD) biomarker data can optimize drug dosing; in this study, we present PD data from the EMBER-2 study. PATIENTS AND METHODS Postmenopausal women with untreated, operable estrogen receptor (ER)-positive, HER2-negative early breast cancer were randomized to 400 versus 800 mg of imlunestrant daily for ∼2 weeks before surgery. A single arm study tested a daily dose of 200 mg. PD biomarker changes (ER, progesterone receptor, Ki-67 by IHC, and mRNA expression of ER-related genes) were evaluated in paired tumor samples (pre-/posttreatment). Safety and pharmacokinetics were also assessed. RESULTS Among evaluable paired samples (n = 75), PD profiles demonstrated consistent ER targeting between 400- and 800-mg doses, with less toxicity at the 400-mg dose. Although inducing the lowest rate of complete cell-cycle arrest, PD and pharmacokinetic results were similar for the 200-mg dose. CONCLUSIONS EMBER-2 combined with existing phase I data has identified 400 mg as the optimal imlunestrant dose.
Collapse
Affiliation(s)
- Patrick Neven
- Leuven Cancer Institute, Universitaire Ziekenhuizen (UZ), Leuven, Belgium
| | | | - Maria Vidal
- Department of Medical Oncology, Hospital Clinic, Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Miguel Martín
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | | | - Nadia Harbeck
- Breast Center, Department of Obstetrics and Gynecology, and Comprehensive Cancer Center CCCLMU, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Kelly K. Hunt
- Department of Breast Surgical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Stacey Carter
- Division of Surgical Oncology, Baylor College of Medicine, Lester and Sue Smith Breast Center, Dan L. Duncan Comprehensive Cancer Center, Houston, Texas
| | | | - Peter A. Fasching
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Philippe Aftimos
- Department of Medical Oncology, Institute Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | - Rebecca Aft
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Swati Kulkarni
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Peter Schmid
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Manali Bhave
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | | | | | | | - Bastien Nguyen
- Loxo@Lilly, Lilly Corporate Center, Indianapolis, Indiana
| | - Umut Ozbek
- Eli Lilly and Company, Indianapolis, Indiana
| | - Eunice Yuen
- Eli Lilly and Company, Indianapolis, Indiana
| | | | - Eva Ciruelos
- Medical Oncology Department, Breast Cancer Unit, University Hospital 12 de Octubre, Madrid, Spain
- Medical Oncology Department, Breast Cancer Unit, HM Hospitales, Madrid, Spain
| |
Collapse
|
23
|
Riis MLH. The Challenges of Lobular Carcinomas from a Surgeon's Point of View. Clin Breast Cancer 2024; 24:e645-e654. [PMID: 39033066 DOI: 10.1016/j.clbc.2024.06.007] [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: 05/08/2024] [Accepted: 06/13/2024] [Indexed: 07/23/2024]
Abstract
Invasive lobular breast cancer (ILC) presents unique challenges and considerations in the realm of surgical management. Characterized by its distinct histological features, including the loss of E-cadherin expression and dys-cohesive growth pattern, ILC often poses diagnostic and therapeutic dilemmas for clinicians. This abstract explores the surgical landscape of ILC, focusing on its epidemiology, clinical presentation, diagnostic modalities, and surgical interventions. Emphasizing the importance of individualized treatment strategies, this narrative delves into the nuances of surgical decision-making, including the role of breast-conserving surgery versus mastectomy, axillary staging, and the significance of margin status. Additionally, advancements in surgical techniques, such as oncoplastic approaches and sentinel lymph node biopsy, are examined in the context of optimizing oncologic outcomes and preserving cosmesis. Through a comprehensive review of current literature and clinical guidelines, this overview aims to provide a nuanced understanding of the surgical considerations inherent to the management of invasive lobular breast cancer.
Collapse
Affiliation(s)
- Margit L H Riis
- Department of Breast and Endocrine Surgery, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
24
|
Pernas S, Sanfeliu E, Villacampa G, Salvador J, Perelló A, González X, Jiménez B, Merino M, Palacios P, Pascual T, Alba E, Villanueva L, Chillara S, Ferrero-Cafiero JM, Galvan P, Prat A, Ciruelos E. Palbociclib and letrozole for hormone receptor-positive HER2-negative breast cancer with residual disease after neoadjuvant chemotherapy. NPJ Breast Cancer 2024; 10:101. [PMID: 39592624 PMCID: PMC11599376 DOI: 10.1038/s41523-024-00710-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
With the incorporation of cyclin-dependent kinase inhibitors in early breast cancer (BC), a better identification of biomarkers is needed. The PROMETEO II trial aimed to evaluate the antitumor activity of palbociclib plus letrozole and to identify response biomarkers in patients with operable HR+/HER2- BC and residual disease after neoadjuvant chemotherapy (NAC). The primary endpoint was the rate of complete cell cycle arrest (CCCA), centrally determined by Ki67 ≤ 2.7% at surgery. A comprehensive translational analysis was conducted. At surgery, the CCCA rate was 59.1%, with a 44.2% decrease in Ki67 from the end of NAC. Changes in intrinsic subtypes occurred in 48% of patients, with proliferation genes suppressed, and immune genes more upregulated in tumors with CCCA. Overall, 14% of tumors were classified as PD-L1+ after palbociclib. Nine patients experienced grade 3 adverse events (AEs). Palbociclib showed an anti-proliferative effect, with increased immune infiltration in residual tumors with CCCA.Trial registration: Palbociclib Plus Letrozole in Hormone Receptor Positive Residual Disease After Neoadjuvant Chemotherapy (PROMETEO II) ClinicalTrial.gov number NCT04130152. Study registration; October 17, 2019.
Collapse
Affiliation(s)
- Sonia Pernas
- SOLTI Cancer Research Group, Barcelona, Spain.
- Institut Catala d'Oncologia-IDIBELL; L'Hospitalet, Barcelona, Spain.
| | - Esther Sanfeliu
- SOLTI Cancer Research Group, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Pathology Department Hospital Clinic de Barcelona, Barcelona, Spain
| | - Guillermo Villacampa
- SOLTI Cancer Research Group, Barcelona, Spain
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | | | - Xavier González
- SOLTI Cancer Research Group, Barcelona, Spain
- Institut Oncològic Dr. Rosell, Barcelona, Spain
- Hospital General de Catalunya, Sant Cugat del Vallès, Spain
- Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain
| | - Begoña Jiménez
- Hospitales Regional y Universitario Virgen de la Victoria, Málaga, Spain
| | - María Merino
- Hospital Universitario Infanta Sofía, Madrid, Spain
| | - Patricia Palacios
- Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | | | - Emilio Alba
- Hospital Clínico Universitario Virgen de la Victoria, Málaga. Instituto de Investigación Biomédica de Málaga, IBIMA, Málaga, Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain
| | | | | | | | - Patricia Galvan
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Aleix Prat
- SOLTI Cancer Research Group, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Hospital Clínic de Barcelona, Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Eva Ciruelos
- SOLTI Cancer Research Group, Barcelona, Spain.
- Hospital 12 de Octubre, Madrid, Spain.
| |
Collapse
|
25
|
Kudo R, Safonov A, Jones C, Moiso E, Dry JR, Shao H, Nag S, da Silva EM, Yildirim SY, Li Q, O'Connell E, Patel P, Will M, Fushimi A, Benitez M, Bradic M, Fan L, Nakshatri H, Sudhan DR, Denz CR, Huerga Sanchez I, Reis-Filho JS, Goel S, Koff A, Weigelt B, Khan QJ, Razavi P, Chandarlapaty S. Long-term breast cancer response to CDK4/6 inhibition defined by TP53-mediated geroconversion. Cancer Cell 2024; 42:1919-1935.e9. [PMID: 39393354 DOI: 10.1016/j.ccell.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 07/02/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
Inhibition of CDK4/6 kinases has led to improved outcomes in breast cancer. Nevertheless, only a minority of patients experience long-term disease control. Using a large, clinically annotated cohort of patients with metastatic hormone receptor-positive (HR+) breast cancer, we identify TP53 loss (27.6%) and MDM2 amplification (6.4%) to be associated with lack of long-term disease control. Human breast cancer models reveal that p53 loss does not alter CDK4/6 activity or G1 blockade but instead promotes drug-insensitive p130 phosphorylation by CDK2. The persistence of phospho-p130 prevents DREAM complex assembly, enabling cell-cycle re-entry and tumor progression. Inhibitors of CDK2 can overcome p53 loss, leading to geroconversion and manifestation of senescence phenotypes. Complete inhibition of both CDK4/6 and CDK2 kinases appears to be necessary to facilitate long-term response across genomically diverse HR+ breast cancers.
Collapse
Affiliation(s)
- Rei Kudo
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Department of Surgery, The Jikei University School of Medicine, Tokyo 1058461, Japan
| | - Anton Safonov
- Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Catherine Jones
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Enrico Moiso
- Department of Medicine, MSK, New York, NY 10065, USA; Department of Epidemiology and Biostatistics, MSK, New York, NY 10065, USA
| | | | - Hong Shao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Sharanya Nag
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Edaise M da Silva
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Selma Yeni Yildirim
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Qing Li
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Elizabeth O'Connell
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Payal Patel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Marie Will
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Clinical Genetics Service, Department of Medicine, MSK, New York, NY 10065, USA
| | - Atsushi Fushimi
- Department of Surgery, The Jikei University School of Medicine, Tokyo 1058461, Japan
| | - Marimar Benitez
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Martina Bradic
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Shom Goel
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Andrew Koff
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Qamar J Khan
- Division of Medical Oncology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Pedram Razavi
- Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
26
|
Manjunath GK, Sharma S, Nashier D, Vasanthaiah S, Jha S, Bage S, Mitra T, Goyal P, Neerathilingam M, Kumar A. Breast cancer genomic analyses reveal genes, mutations, and signaling networks. Funct Integr Genomics 2024; 24:206. [PMID: 39496981 DOI: 10.1007/s10142-024-01484-y] [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: 08/26/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/06/2024]
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer and the predominant cause of death in women. BC is a complex disorder, and the exploration of several types of BC omic data, highlighting genes, perturbations, signaling and cellular mechanisms, is needed. We collected mutational data from 9,555 BC samples using cBioPortal. We classified 1174 BC genes (mutated ≥ 40 samples) into five tiers (BCtier_I-V) and subjected them to pathway and protein‒protein network analyses using EnrichR and STRING 11, respectively. BCtier_I possesses 12 BC genes with mutational frequencies > 5%, with only 5 genes possessing > 10% frequencies, namely, PIK3CA (35.7%), TP53 (34.3%), GATA3 (11.5%), CDH1 (11.4%) and MUC16 (11%), and the next seven BC genes are KMT2C (8.8%), TTN (8%), MAP3K1 (8%), SYNE1 (7.2%), AHNAK2 (7%), USH2A (5.5%), and RYR2 (5.4%). Our pathway analyses revealed that the five top BC pathways were the PI3K-AKT, TP53, NOTCH, HIPPO, and RAS pathways. We found that BC panels share only seven genes. These findings show that BC arises from genetic disruptions evident in BC signaling and protein networks.
Collapse
Affiliation(s)
- Gowrang Kasaba Manjunath
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore, 560066, Karnataka, India
| | - Srihari Sharma
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore, 560066, Karnataka, India
| | - Disha Nashier
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore, 560066, Karnataka, India
| | - Shruthi Vasanthaiah
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore, 560066, Karnataka, India
| | - Spriha Jha
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore, 560066, Karnataka, India
| | - Saloni Bage
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Tamoghna Mitra
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore, 560066, Karnataka, India
| | - Pankaj Goyal
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Muniasamy Neerathilingam
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore, 560066, Karnataka, India
| | - Abhishek Kumar
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India.
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore, 560066, Karnataka, India.
| |
Collapse
|
27
|
Song G, Liu J, Tang X, Zhong J, Zeng Y, Zhang X, Zhou J, Zhou J, Cao L, Zhang Q, Li Y. Cell cycle checkpoint revolution: targeted therapies in the fight against malignant tumors. Front Pharmacol 2024; 15:1459057. [PMID: 39464635 PMCID: PMC11505109 DOI: 10.3389/fphar.2024.1459057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Malignant tumors are among the most important causes of death worldwide. The pathogenesis of a malignant tumor is complex and has not been fully elucidated. Studies have shown that such pathogenesis is related to abnormal cell cycle progression. The expression levels of cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors as well as functions of the cell cycle checkpoints determine whether the cell cycle progression is smooth. Cell-cycle-targeting drugs have the advantages of high specificity, low toxicity, low side effects, and low drug resistance. Identifying drugs that target the cell cycle and applying them in clinical treatments are expected to promote chemotherapeutic developments against malignant tumors. This article aims to review drugs targeted against the cell cycle and their action mechanisms.
Collapse
Affiliation(s)
- Guangming Song
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jue Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, The affiliated Zhuzhou hospital Xiangya medical college, Central South University, Zhuzhou, Hunan, China
| | - Jie Zhong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuhuan Zeng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaodi Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jianbin Zhou
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jie Zhou
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lu Cao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qunfeng Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, The affiliated Zhuzhou hospital Xiangya medical college, Central South University, Zhuzhou, Hunan, China
| |
Collapse
|
28
|
Guerrero-Zotano Á, Pérez-García JM, Ruiz-Borrego M, Bermejo B, Gil-Gil M, de la Haba J, Alba Conejo E, Quiroga V, Carañana V, Urruticoechea A, Morales S, Bellet M, Antón A, Fernández-Abad M, Sánchez-Rovira P, Calabuig L, Pérez-Escuredo J, Sampayo-Cordero M, Cortés J, Llombart-Cussac A. Neoadjuvant letrozole and palbociclib in patients with HR-positive/HER2-negative early breast cancer and Oncotype DX Recurrence Score ≥18: DxCARTES study. ESMO Open 2024; 9:103733. [PMID: 39353215 PMCID: PMC11472226 DOI: 10.1016/j.esmoop.2024.103733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/25/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The effect of the addition of cyclin-dependent kinases 4 and 6 inhibitors to endocrine therapy in terms of molecular downstaging remains undetermined. Switching from a high-risk to a low risk Recurrence Score (RS) group could provide useful information to identify patients who might not require chemotherapy. The purpose of this study was to assess the biological and clinical activity of letrozole plus palbociclib as neoadjuvant treatment for patients with hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative early breast cancer with an initial Oncotype DX RS ≥18. PATIENTS AND METHODS Participants were women aged ≥18 years with HR-positive/HER2-negative, Ki67 ≥ 20%, stage II-IIIB early breast cancer with a baseline RS ≥18. Eligible patients with a pretreatment RS 18-25 (cohort A) and 26-100 (cohort B) received six 28-day cycles of letrozole (2.5 mg per day; plus goserelin if pre- or perimenopausal) plus palbociclib (125 mg per day; 3/1 schedule) before surgery. The primary endpoint for both cohorts was the proportion of patients who achieved an RS ≤25 at surgery or a pathological complete response (pCR). RESULTS A total of 67 patients were enrolled, among which 65 were assessable for the primary endpoint (32 patients in cohort A and 33 in cohort B). At surgery, 22 (68.8%) patients in cohort A and 18 (54.5%) patients in cohort B had an RS ≤25 or a pCR [only 1 (3.0%) patient in cohort B], meeting the primary endpoint in cohort B (P < 0.01), but not in cohort A (P = 0.98). No new safety signals were identified. CONCLUSIONS The efficacy of neoadjuvant treatment with letrozole plus palbociclib does not seem to depend on pretreatment RS for patients with RS ≥18. However, around half of patients with HR-positive/HER2-negative early breast cancer with an RS 26-100 at baseline achieved molecular downstaging with this regimen.
Collapse
Affiliation(s)
| | - J M Pérez-García
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil)
| | - M Ruiz-Borrego
- Hospital For Universitario Virgen del Rocío, Sevilla, Spain
| | - B Bermejo
- Hospital Clínico Universitario de Valencia, Valencia, Spain; Biomedical Research Institute INCLIVA, Valencia, Spain
| | - M Gil-Gil
- Institut Català d'Oncologia L'Hospitalet, Hospitalet de Llobregat, Barcelona, Spain
| | - J de la Haba
- Hospital Universitario Reina Sofía, Cordoba, Spain
| | - E Alba Conejo
- Unidad de Gestión Clínica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, Malaga, Spain; Faculty of Medicine, The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), University of Málaga, Malaga, Spain; Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), Madrid, Spain
| | - V Quiroga
- Hospital Clínico Universitario Virgen de la Victoria; Instituto de Investigación Biomédica de Málaga, IBIMA; GEICAM, Spanish Breast Cancer Group, Málaga, Spain
| | - V Carañana
- Hospital Arnau de Vilanova de Valencia, Valencia, Spain
| | - A Urruticoechea
- Gipuzkoa Cancer Unit - BioGipuzkoa, Osakidetza, Donostia, Spain
| | - S Morales
- Hospital Arnau de Vilanova de Lleida, Lleida, Spain
| | - M Bellet
- Hospital Universitari Vall d'Hebrón, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - A Antón
- Hospital Universitario Miguel Servet, Zaragoza, Spain
| | | | | | - L Calabuig
- Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil)
| | - J Pérez-Escuredo
- Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil)
| | - M Sampayo-Cordero
- Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil)
| | - J Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil); Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain; IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | - A Llombart-Cussac
- Medica Scientia Innovation Research (MEDSIR) - Oncoclínicas&Co, Jersey City (New Jersey, USA), Sao Paulo (Brazil); Hospital Arnau de Vilanova, Valencia, Spain; Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain.
| |
Collapse
|
29
|
Chen A, Kim BJ, Mitra A, Vollert CT, Lei JT, Fandino D, Anurag M, Holt MV, Gou X, Pilcher JB, Goetz MP, Northfelt DW, Hilsenbeck SG, Marshall CG, Hyer ML, Papp R, Yin SY, De Angelis C, Schiff R, Fuqua SAW, Ma CX, Foulds CE, Ellis MJ. PKMYT1 Is a Marker of Treatment Response and a Therapeutic Target for CDK4/6 Inhibitor-Resistance in ER+ Breast Cancer. Mol Cancer Ther 2024; 23:1494-1510. [PMID: 38781103 PMCID: PMC11443213 DOI: 10.1158/1535-7163.mct-23-0564] [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: 08/26/2023] [Revised: 03/25/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Endocrine therapies (ET) with cyclin-dependent kinase 4/6 (CDK4/6) inhibition are the standard treatment for estrogen receptor-α-positive (ER+) breast cancer, however drug resistance is common. In this study, proteogenomic analyses of patient-derived xenografts (PDXs) from patients with 22 ER+ breast cancer demonstrated that protein kinase, membrane-associated tyrosine/threonine one (PKMYT1), a WEE1 homolog, is estradiol (E2) regulated in E2-dependent PDXs and constitutively expressed when growth is E2-independent. In clinical samples, high PKMYT1 mRNA levels associated with resistance to both ET and CDK4/6 inhibition. The PKMYT1 inhibitor lunresertib (RP-6306) with gemcitabine selectively and synergistically reduced the viability of ET and palbociclib-resistant ER+ breast cancer cells without functional p53. In vitro the combination increased DNA damage and apoptosis. In palbociclib-resistant, TP53 mutant PDX-derived organoids and PDXs, RP-6306 with low-dose gemcitabine induced greater tumor volume reduction compared to treatment with either single agent. Our study demonstrates the clinical potential of RP-6306 in combination with gemcitabine for ET and CDK4/6 inhibitor resistant TP53 mutant ER+ breast cancer.
Collapse
Affiliation(s)
- Anran Chen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, Texas
- Repare Therapeutics, Cambridge, Massachusetts
| | - Beom-Jun Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Aparna Mitra
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Craig T Vollert
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Employee of Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas
| | - Diana Fandino
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Matthew V Holt
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Xuxu Gou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Jacob B Pilcher
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | | | - Donald W Northfelt
- Division of Hematology and Medical Oncology at Mayo Clinic, Phoenix, Arizona
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | | | - Marc L Hyer
- Repare Therapeutics, Cambridge, Massachusetts
| | - Robert Papp
- Repare Therapeutics, Saint-Laurent, Quebec, Canada
| | - Shou-Yun Yin
- Repare Therapeutics, Saint-Laurent, Quebec, Canada
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Suzanne A W Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Charles E Foulds
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
30
|
Hirmas N, Holtschmidt J, Loibl S. Shifting the Paradigm: The Transformative Role of Neoadjuvant Therapy in Early Breast Cancer. Cancers (Basel) 2024; 16:3236. [PMID: 39335206 PMCID: PMC11430607 DOI: 10.3390/cancers16183236] [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: 09/05/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
The use of neoadjuvant systemic therapy (NST) has become increasingly important in the treatment of breast cancer because of its various advantages. These include the ability to downstage tumors without compromising locoregional control and the potential to obtain valuable information about clinical and biological response to therapy with implications for individual prognoses. Surgical response assessment paves the way for response-adapted therapy, and pathological complete response (pCR; defined as ypT0/is ypN0) serves as an additional endpoint for drug development trials. Recommended NST regimens commonly consist of anthracyclines and taxane, with dose-dense anthracyclines and weekly paclitaxel often preferred, whenever feasible. For patients with human epidermal growth factor receptor-2 (HER2)-positive tumors, dual anti-HER2 therapy (trastuzumab and pertuzumab) is indicated together with NST in case of elevated risk of recurrence. For patients with triple-negative breast cancer (TNBC), adding carboplatin to NST correlates with improved pCR and survival rates, as does the addition of immune checkpoint inhibitors. For hormone receptor (HR)-positive/HER2-negative cancers, emerging data on NST including immune checkpoint inhibitors may elevate the significance of NST in high-risk luminal breast cancer. Here, we present a synthesis of the results from neoadjuvant clinical trials that aim at optimizing treatment options for patients with high-risk breast cancer.
Collapse
Affiliation(s)
- Nader Hirmas
- German Breast Group, 63263 Neu-Isenburg, Germany
| | | | - Sibylle Loibl
- German Breast Group, 63263 Neu-Isenburg, Germany
- Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| |
Collapse
|
31
|
He W, Demas DM, Kraikivski P, Shajahan-Haq AN, Baumann WT. WEE1 inhibition delays resistance to CDK4/6 inhibitor and antiestrogen treatment in estrogen receptor-positive breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613122. [PMID: 39345487 PMCID: PMC11429701 DOI: 10.1101/2024.09.15.613122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Although endocrine therapies and Cdk4/6 inhibitors have produced significantly improved outcomes for patients with estrogen receptor positive (ER+) breast cancer, continuous application of these drugs often results in resistance. We hypothesized that cancer cells acquiring drug resistance might increase their dependency on negative regulators of the cell cycle. Therefore, we investigated the effect of inhibiting WEE1 on delaying the development of resistance to palbociclib and fulvestrant. We treated ER+ MCF7 breast cancer cells with palbociclib alternating with a combination of fulvestrant and a WEE1 inhibitor AZD1775 for 12 months. We found that the alternating treatment prevented the development of drug resistance to palbociclib and fulvestrant compared to monotherapies. Furthermore, we developed a mathematical model that can simulate cell proliferation under monotherapy, combination or alternating drug treatments. Finally, we showed that the mathematical model can be used to minimize the number of fulvestrant plus AZD1775 treatment periods while maintaining its efficacy.
Collapse
Affiliation(s)
- Wei He
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Diane M Demas
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Pavel Kraikivski
- Division of Systems Biology, Academy of Integrated Science, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Ayesha N Shajahan-Haq
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - William T Baumann
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| |
Collapse
|
32
|
Zhang Z, Zhao X, Chen J. Adjuvant and neoadjuvant therapy with or without CDK4/6 inhibitors in HR+/HER2- early breast cancer: a systematic review and meta-analysis. Front Pharmacol 2024; 15:1438288. [PMID: 39329126 PMCID: PMC11424878 DOI: 10.3389/fphar.2024.1438288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Background The combination of cyclin-dependent kinases 4/6 (CDK4/6) inhibitors and endocrine therapy is the standard treatment for patients with hormone receptor-positive (HR+)/HER2-negative (HER2-) advanced breast cancer. However, the role of CDK4/6 inhibitors in early breast cancer remains controversial. Methods This study aimed to evaluate the efficacy and safety of CDK4/6 inhibitors combined with endocrine therapy versus endocrine therapy alone in patients with HR+, HER2- early breast cancer. A systematic review of Cochrane, PubMed and EMBASE databases was conducted. The efficacy endpoints of adjuvant therapy were invasive disease-free survival (IDFS), overall survival (OS) and distant relapse-free survival (DRFS). The efficacy endpoint included complete cell cycle arrest (CCCA) and complete pathologic response (PCR) with neoadjuvant therapy. Grade 3/4 adverse events (AEs) were assessed as safety outcomes. Results Eight randomized controlled trials (RCTs) were included in the study. CDK4/6 inhibitors combined with endocrine therapy showed a significant improvement in IDFS (hazard ratio (HR) = 0.81, 95% confidence interval (CI) = 0.68-0.97, P = 0.024), but not DRFS (HR = 0.84, 95% CI = 0.56-1.29, P = 0.106) or OS (HR = 0.96, 95% CI = 0.77-1.19, P = 0.692) in adjuvant therapy. In the neoadjuvant therapy setting, CDK4/6 inhibitors improved CCCA compared with the control group (RR = 2.08, 95% CI = 1.33-3.26, P = 0.001). The risk of 3/4 grade AEs increased significantly with the addition of CDK4/6 inhibitors to endocrine therapy. Conclusion The addition of CDK4/6 inhibitors in HR+/HER2- early breast cancer patients significantly improved IDFS in adjuvant therapy and CCCA in neoadjuvant. However, CDK4/6 inhibitors also showed significant toxicities during therapy. Systematic Review Registration Identifier CRD42024530704.
Collapse
Affiliation(s)
| | | | - Jie Chen
- Breast Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Schmiester L, Brasó-Maristany F, González-Farré B, Pascual T, Gavilá J, Tekpli X, Geisler J, Kristensen VN, Frigessi A, Prat A, Köhn-Luque A. Computational Model Predicts Patient Outcomes in Luminal B Breast Cancer Treated with Endocrine Therapy and CDK4/6 Inhibition. Clin Cancer Res 2024; 30:3779-3787. [PMID: 38922642 DOI: 10.1158/1078-0432.ccr-24-0244] [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: 01/28/2024] [Revised: 04/12/2024] [Accepted: 06/24/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE Development of a computational biomarker to predict, prior to treatment, the response to CDK4/6 inhibition (CDK4/6i) in combination with endocrine therapy in patients with breast cancer. EXPERIMENTAL DESIGN A mechanistic mathematical model that accounts for protein signaling and drug mechanisms of action was developed and trained on extensive, publicly available data from breast cancer cell lines. The model was built to provide a patient-specific response score based on the expression of six genes (CCND1, CCNE1, ESR1, RB1, MYC, and CDKN1A). The model was validated in five independent cohorts of 148 patients in total with early-stage or advanced breast cancer treated with endocrine therapy and CDK4/6i. Response was measured either by evaluating Ki67 levels and PAM50 risk of relapse (ROR) after neoadjuvant treatment or by evaluating progression-free survival (PFS). RESULTS The model showed significant association with patient's outcomes in all five cohorts. The model predicted high Ki67 [area under the curve; AUC (95% confidence interval, CI) of 0.80 (0.64-0.92), 0.81 (0.60-1.00) and 0.80 (0.65-0.93)] and high PAM50 ROR [AUC of 0.78 (0.64-0.89)]. This observation was not obtained in patients treated with chemotherapy. In the other cohorts, patient stratification based on the model prediction was significantly associated with PFS [hazard ratio (HR) = 2.92 (95% CI, 1.08-7.86), P = 0.034 and HR = 2.16 (1.02 4.55), P = 0.043]. CONCLUSIONS A mathematical modeling approach accurately predicts patient outcome following CDK4/6i plus endocrine therapy that marks a step toward more personalized treatments in patients with Luminal B breast cancer.
Collapse
Affiliation(s)
- Leonard Schmiester
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Blanca González-Farré
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Tomás Pascual
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Joaquín Gavilá
- SOLTI Cancer Research Group, Barcelona, Spain
- Department of Medical Oncology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Xavier Tekpli
- Department of Medical Genetics, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Jürgen Geisler
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Akershus University Hospital, Oslo, Norway
| | - Vessela N Kristensen
- Department of Medical Genetics, Oslo University Hospital, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Arnoldo Frigessi
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Alvaro Köhn-Luque
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
34
|
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
|
35
|
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
|
36
|
Lorman-Carbó N, Martínez-Sáez O, Fernandez-Martinez A, Galván P, Chic N, Garcia-Fructuoso I, Rodríguez A, Gómez-Bravo R, Schettini F, Blasco P, Castillo O, González-Farré B, Adamo B, Vidal M, Muñoz M, Perou CM, Malumbres M, Gavilá J, Pascual T, Prat A, Brasó-Maristany F. Comparative biological activity of palbociclib and ribociclib in hormone receptor-positive breast cancer. Sci Rep 2024; 14:16030. [PMID: 38992220 PMCID: PMC11239654 DOI: 10.1038/s41598-024-67126-2] [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: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
This study examines the biological effects of palbociclib and ribociclib in hormone receptor-positive breast cancer, pivotal to the HARMONIA prospective phase III clinical trial. We explore the downstream impacts of these CDK4/6 inhibitors, focusing on cell lines and patient-derived tumor samples. We treated HR+ breast cancer cell lines (T47D, MCF7, and BT474) with palbociclib or ribociclib (100 nM or 500 nM), alone or combined with fulvestrant (1 nM), over periods of 24, 72, or 144 h. Our assessments included PAM50 gene expression, RB1 phosphorylation, Lamin-B1 protein levels, and senescence-associated β-galactosidase activity. We further analyzed PAM50 gene signatures from the CORALLEEN and NeoPalAna phase II trials. Both CDK4/6 inhibitors similarly inhibited proliferation across the cell lines. At 100 nM, both drugs partially reduced p-RB1, with further decreases at 500 nM over 144 h. Treatment led to reduced Lamin-B1 expression and increased senescence-associated β-galactosidase activity. Both drugs enhanced Luminal A and reduced Luminal B and proliferation signatures at both doses. However, the HER2-enriched signature significantly diminished only at the higher dose of 500 nM. Corresponding changes were observed in tumor samples from the CORALLEEN and NeoPalAna studies. At 2 weeks of treatment, both drugs significantly reduced the HER2-enriched signature, but at surgery, this reduction was consistent only with ribociclib. Our findings suggest that while both CDK4/6 inhibitors effectively modulate key biological pathways in HR+/HER2- breast cancer, nuances in their impact, particularly on the HER2-enriched signature, are dose-dependent, influenced by the addition of fulvestrant and warrant further investigation.
Collapse
Affiliation(s)
- Natàlia Lorman-Carbó
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Olga Martínez-Sáez
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Aranzazu Fernandez-Martinez
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Patricia Galván
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
| | - Nuria Chic
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Isabel Garcia-Fructuoso
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Adela Rodríguez
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Raquel Gómez-Bravo
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Francesco Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Paula Blasco
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
| | - Oleguer Castillo
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
| | - Blanca González-Farré
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Pathology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Barbara Adamo
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Maria Vidal
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- SOLTI Cooperative Group, Barcelona, Spain
- Institute of Oncology-Hospital Quirónsalud, Barcelona, Spain
| | - Montserrat Muñoz
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- SOLTI Cooperative Group, Barcelona, Spain
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Marcos Malumbres
- Cancer Cell Cycle Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- ICREA, Barcelona, Spain
| | - Joaquín Gavilá
- SOLTI Cooperative Group, Barcelona, Spain
- Department of Medical Oncology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Tomás Pascual
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- SOLTI Cooperative Group, Barcelona, Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Institute of Oncology-Hospital Quirónsalud, Barcelona, Spain
- Reveal Genomics, S.L, Barcelona, Spain
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Carrer de Casanova, 143, 08036, Barcelona, Spain.
- Reveal Genomics, S.L, Barcelona, Spain.
| |
Collapse
|
37
|
Zhang Y, Zhou S, Kai Y, Zhang YQ, Peng C, Li Z, Mughal MJ, Julie B, Zheng X, Ma J, Ma CX, Shen M, Hall MD, Li S, Zhu W. O-GlcNAcylation of MITF regulates its activity and CDK4/6 inhibitor resistance in breast cancer. Nat Commun 2024; 15:5597. [PMID: 38961064 PMCID: PMC11222436 DOI: 10.1038/s41467-024-49875-w] [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: 09/22/2023] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4/6) play a pivotal role in cell cycle and cancer development. Targeting CDK4/6 has demonstrated promising effects against breast cancer. However, resistance to CDK4/6 inhibitors (CDK4/6i), such as palbociclib, remains a substantial challenge in clinical settings. Using high-throughput combinatorial drug screening and genomic sequencing, we find that the microphthalmia-associated transcription factor (MITF) is activated via O-GlcNAcylation by O-GlcNAc transferase (OGT) in palbociclib-resistant breast cancer cells and tumors. Mechanistically, O-GlcNAcylation of MITF at Serine 49 enhances its interaction with importin α/β, thus promoting its translocation to nuclei, where it suppresses palbociclib-induced senescence. Inhibition of MITF or its O-GlcNAcylation re-sensitizes resistant cells to palbociclib. Moreover, clinical studies confirm the activation of MITF in tumors from patients who are palbociclib-resistant or undergoing palbociclib treatment. Collectively, our studies shed light on the mechanism regulating palbociclib resistance and present clinical evidence for developing therapeutic approaches to treat CDK4/6i-resistant breast cancer patients.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Shuyan Zhou
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Yan Kai
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Ya-Qin Zhang
- Division of Preclinical Innovation (Intramural), National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD, USA
| | - Changmin Peng
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Zhuqing Li
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Muhammad Jameel Mughal
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Belmar Julie
- Department of Medicine, Washington University School of Medicine in St Louis, Siteman Cancer Center, St Louis, MO, USA
| | - Xiaoyan Zheng
- Department of Anatomy and Cell Biology, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Min Shen
- Division of Preclinical Innovation (Intramural), National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD, USA
| | - Matthew D Hall
- Division of Preclinical Innovation (Intramural), National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD, USA
| | - Shunqiang Li
- Department of Medicine, Washington University School of Medicine in St Louis, Siteman Cancer Center, St Louis, MO, USA.
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
38
|
Poulet S, Dai M, Wang N, Yan G, Boudreault J, Daliah G, Guillevin A, Nguyen H, Galal S, Ali S, Lebrun JJ. Genome-wide in vivo CRISPR screen identifies TGFβ3 as actionable biomarker of palbociclib resistance in triple negative breast cancer. Mol Cancer 2024; 23:118. [PMID: 38831405 PMCID: PMC11145857 DOI: 10.1186/s12943-024-02029-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 05/22/2024] [Indexed: 06/05/2024] Open
Abstract
Triple negative breast cancer (TNBC) remains exceptionally challenging to treat. While CDK4/6 inhibitors have revolutionized HR + breast cancer therapy, there is limited understanding of their efficacy in TNBC and meaningful predictors of response and resistance to these drugs remain scarce. We conducted an in vivo genome-wide CRISPR screen using palbociclib as a selection pressure in TNBC. Hits were prioritized using microarray data from a large panel of breast cancer cell lines to identify top palbociclib sensitizers. Our study defines TGFβ3 as an actionable determinant of palbociclib sensitivity that potentiates its anti-tumor effects. Mechanistically, we show that chronic palbociclib exposure depletes p21 levels, contributing to acquired resistance, and that TGFβ3 treatment can overcome this. This study defines TGFβ3 as an actionable biomarker that can be used to improve patient stratification for palbociclib treatment and exploits the synergistic interaction between CDK4/6 and TGFβ3 to propose a new combinatorial treatment for TNBC.
Collapse
Affiliation(s)
- Sophie Poulet
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Meiou Dai
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Ni Wang
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Gang Yan
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Julien Boudreault
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Girija Daliah
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Alan Guillevin
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Huong Nguyen
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Soaad Galal
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Suhad Ali
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada
| | - Jean-Jacques Lebrun
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
39
|
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
|
40
|
Corti C, Batra-Sharma H, Kelsten M, Shatsky RA, Garrido-Castro AC, Gradishar WJ. Systemic Therapy in Breast Cancer. Am Soc Clin Oncol Educ Book 2024; 44:e432442. [PMID: 39013124 DOI: 10.1200/edbk_432442] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Therapeutic advances in breast cancer have significantly improved outcomes in recent decades. In the early setting, there has been a gradual shift from adjuvant-only to neoadjuvant strategies, with a growing focus on customizing post-neoadjuvant treatments through escalation and de-escalation based on pathologic response. At the same time, the transition from a pre-genomic to a post-genomic era, utilizing specific assays in the adjuvant setting and targeted sequencing in the advanced stage, has deepened our understanding of disease biology and aided in identifying molecular markers associated with treatment benefit. Finally, the introduction of new drug classes such as antibody-drug conjugates, and the incorporation in the (neo)adjuvant setting of therapies previously investigated in the advanced stage, like immunotherapy and CDK4-6 inhibitors, poses new challenges in treatment sequencing.
Collapse
Affiliation(s)
- Chiara Corti
- Dana Farber Cancer Institute, Harvard University, Boston, MA
| | | | - Max Kelsten
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | | | - William J Gradishar
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| |
Collapse
|
41
|
van de Loo ME, Andour L, van Heesewijk AE, Oosterkamp HM, Liefers GJ, Straver ME. Neoadjuvant endocrine treatment in hormone receptor-positive breast cancer: Does it result in more breast-conserving surgery? Breast Cancer Res Treat 2024; 205:5-16. [PMID: 38265568 DOI: 10.1007/s10549-023-07222-5] [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: 06/30/2023] [Accepted: 12/10/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Patients with locally advanced endocrine positive tumors who will not benefit from chemotherapy can be treated by either primary surgery or neoadjuvant endocrine therapy (NET). How often does NET result in breast-conserving surgery (BCS)? METHODS We conducted a literature search in PubMed and Embase, to identify articles on surgical treatment after NET. RESULTS In 19 studies the pathological complete response (pCR) rate was reported after NET; an overall pCR rate of 1% was found. Compared with neoadjuvant chemotherapy (NCT), the BCS rate was significantly higher after NET (OR 0.60; 95% CI, 0.51-0.69; P < 0.00001). The surgical conversion rate was reported in eight studies [4-75.9%], with a mean of 30.2%. CONCLUSION This review found that one out of three patients becomes eligible for BCS after treatment with NET.
Collapse
Affiliation(s)
- Merel E van de Loo
- Department of Surgery, Medical Center Haaglanden, The Hague, The Netherlands
| | - Layla Andour
- Department of Surgery, Medical Center Haaglanden, The Hague, The Netherlands
| | | | | | - Gerrit-Jan Liefers
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke E Straver
- Department of Surgery, Medical Center Haaglanden, The Hague, The Netherlands.
| |
Collapse
|
42
|
Ploumaki I, Triantafyllou E, Koumprentziotis IA, Karampinos K, Drougkas K, Karavolias I, Kotteas E. Cyclin-Dependent Kinase 4/6 Inhibitors as Neoadjuvant Therapy of Hormone Receptor-Positive/HER2-Negative Early Breast Cancer: What do we Know so Far? Clin Breast Cancer 2024; 24:e177-e185. [PMID: 38320891 DOI: 10.1016/j.clbc.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/04/2024] [Accepted: 01/07/2024] [Indexed: 02/08/2024]
Abstract
The introduction of cyclin-dependent kinase 4/6 (CDK4/6) inhibitors to the treatment of hormone receptor-positive and human epidermal growth factor receptor 2-negative (HR+/HER2-) metastatic breast cancer is regarded as one of the greatest achievements of the last decades in breast oncology. To date, palbociclib, abemaciclib and ribociclib are the 3 approved CDK4/6 inhibitors that combined with endocrine therapy are now considered as the standard first-line treatment of metastatic HR+/HER2- breast cancer. The great success of these drugs in the setting of metastatic disease and the need to combat the high risk of recurrence have paved the way for a number of clinical trials to explore the use of CDK4/6 inhibitors in the neoadjuvant treatment of early breast cancer. In this review, we summarize the main findings of clinical trials that examined the use of CDK4/6 inhibitors in combination with hormone therapy or chemotherapy as neoadjuvant treatment of hormone receptor-positive and HER2-negative breast cancer. Active clinical trials that investigate different treatment schemes are also briefly presented and current limitations and future goals are discussed.
Collapse
Affiliation(s)
- Ioanna Ploumaki
- Oncology Unit, 3rd Department of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Αthens, Greece.
| | - Efthymios Triantafyllou
- Oncology Unit, 3rd Department of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Αthens, Greece
| | - Ioannis-Alexios Koumprentziotis
- Oncology Unit, 3rd Department of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Αthens, Greece
| | - Konstantinos Karampinos
- Oncology Unit, 3rd Department of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Αthens, Greece
| | - Konstantinos Drougkas
- Oncology Unit, 3rd Department of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Αthens, Greece
| | - Ioannis Karavolias
- Oncology Unit, 3rd Department of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Αthens, Greece
| | - Elias Kotteas
- Oncology Unit, 3rd Department of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Αthens, Greece
| |
Collapse
|
43
|
Park S, Ryu WJ, Kim TY, Hwang Y, Han HJ, Lee JD, Kim GM, Sohn J, Kim SK, Kim MH, Kim J. Overcoming BRAF and CDK4/6 inhibitor resistance by inhibiting MAP3K3-dependent protection against YAP lysosomal degradation. Exp Mol Med 2024; 56:987-1000. [PMID: 38622197 PMCID: PMC11059244 DOI: 10.1038/s12276-024-01210-5] [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: 01/05/2023] [Revised: 11/09/2023] [Accepted: 02/01/2024] [Indexed: 04/17/2024] Open
Abstract
Transcriptional programs governed by YAP play key roles in conferring resistance to various molecular-targeted anticancer agents. Strategies aimed at inhibiting YAP activity have garnered substantial interest as a means to overcome drug resistance. However, despite extensive research into the canonical Hippo-YAP pathway, few clinical agents are currently available to counteract YAP-associated drug resistance. Here, we present a novel mechanism of YAP stability regulation by MAP3K3 that is independent of Hippo kinases. Furthermore, we identified MAP3K3 as a target for overcoming anticancer drug resistance. Depletion of MAP3K3 led to a substantial reduction in the YAP protein level in melanoma and breast cancer cells. Mass spectrometry analysis revealed that MAP3K3 phosphorylates YAP at serine 405. This MAP3K3-mediated phosphorylation event hindered the binding of the E3 ubiquitin ligase FBXW7 to YAP, thereby preventing its p62-mediated lysosomal degradation. Robust YAP activation was observed in CDK4/6 inhibitor-resistant luminal breast cancer cells. Knockdown or pharmacological inhibition of MAP3K3 effectively suppressed YAP activity and restored CDK4/6 inhibitor sensitivity. Similarly, elevated MAP3K3 expression supported the prosurvival activity of YAP in BRAF inhibitor-resistant melanoma cells. Inhibition of MAP3K3 decreased YAP-dependent cell proliferation and successfully restored BRAF inhibitor sensitivity. In conclusion, our study reveals a previously unrecognized mechanism for the regulation of YAP stability, suggesting MAP3K3 inhibition as a promising strategy for overcoming resistance to CDK4/6 and BRAF inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Sanghyun Park
- Department of Dermatology, Chonnam National University Medical School, Gwangju, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Won-Ji Ryu
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Tae Yeong Kim
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Yumi Hwang
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Ju Han
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong Dong Lee
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Gun Min Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Kyum Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea.
| | - Min Hwan Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| | - Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.
| |
Collapse
|
44
|
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
|
45
|
Pascual T, Fernandez-Martinez A, Agrawal Y, Pfefferle AD, Chic N, Brasó-Maristany F, Gonzàlez-Farré B, Paré L, Villacampa G, Saura C, Hernando C, Muñoz M, Galván P, Gonzàlez-Farré X, Oliveira M, Gil-Gil M, Ciruelos E, Villagrasa P, Gavilá J, Prat A, Perou CM. Cell-cycle inhibition and immune microenvironment in breast cancer treated with ribociclib and letrozole or chemotherapy. NPJ Breast Cancer 2024; 10:20. [PMID: 38448600 PMCID: PMC10918094 DOI: 10.1038/s41523-024-00625-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/22/2024] [Indexed: 03/08/2024] Open
Abstract
In this study, we performed genomic analyses of cell cycle and tumor microenvironment changes during and after ribociclib and letrozole or chemotherapy in the CORALLEEN trial. 106 women with untreated PAM50-defined Luminal B early breast cancers were randomly assigned to receive neoadjuvant ribociclib and letrozole or standard-of-care chemotherapy. Ki67 immunohistochemistry, tumor-infiltrating lymphocytes quantification, and RNA sequencing were obtained from tissue biopsies pre-treatment, on day 14 of treatment, and tumor specimens from surgical resection. Results showed that at surgery, Ki67 and the PAM50 proliferation scores were lower after ribociclib compared to chemotherapy. However, consistent reactivation of tumor cell proliferation from day 14 to surgery was only observed in the ribociclib arm. In tumors with complete cell cycle arrest (CCCA) at surgery, PAM50 proliferation scores were lower in the ribociclib arm compared to chemotherapy (p < 0.001), whereas the opposite was observed with tumor cellularity (p = 0.002). Gene expression signatures (GES) associated with antigen-presenting cells (APCs) and innate immune system activity showed increased expression post-chemotherapy but decreased expression post-ribociclib. Interferon-associated GES had decreased expression with CCCA and increased expression with non-CCCA. Our findings suggest that while both treatment strategies decreased proliferation, the depth and the patterns over time differed by treatment arm. Immunologically, ribociclib was associated with downregulated GES associated with APCs and the innate immune system in Luminal B tumors, contrary to existing preclinical data. Further studies are needed to understand the effect of CDK4/6 inhibition on the tumor cells and microenvironment, an effect which may vary according to tumor subtypes.
Collapse
Affiliation(s)
- Tomás Pascual
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Aranzazu Fernandez-Martinez
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Yash Agrawal
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Adam D Pfefferle
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nuria Chic
- Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Blanca Gonzàlez-Farré
- SOLTI Cancer Research Group, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Pathology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Laia Paré
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | - Cristina Saura
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Breast Cancer Program, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Cristina Hernando
- Medical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Breast Cancer Biology Research Group, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Montserrat Muñoz
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Patricia Galván
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Xavier Gonzàlez-Farré
- SOLTI Cancer Research Group, Barcelona, Spain
- Breast Cancer Unit, Hospital Universitari General de Catalunya, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Mafalda Oliveira
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Breast Cancer Program, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Miguel Gil-Gil
- IDIBELL, L'Hospitalet, Barcelona, Spain
- Department of Medical Oncology, Multidisciplinary Breast Cancer Unit, Institut Català d'Oncologia Medical Oncology, Barcelona, Spain
| | - Eva Ciruelos
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital 12 de Octubre, Madrid, Spain
- Medical Oncology Department, HM Hospitales Madrid, Madrid, Spain
| | | | - Joaquín Gavilá
- SOLTI Cancer Research Group, Barcelona, Spain
- Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Aleix Prat
- SOLTI Cancer Research Group, Barcelona, Spain.
- Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, Spain.
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
- Department of Medicine, University of Barcelona, Barcelona, Spain.
- Breast Cancer Unit, IOB-Quirón Salud, Barcelona, Spain.
| | - Charles M Perou
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
46
|
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
|
47
|
Yu Z, Deng P, Chen Y, Lin D, Liu S, Hong J, Guan P, Chen J, Zhong ME, Chen J, Chen X, Sun Y, Wang Y, Wang P, Cai Z, Chan JY, Huang Y, Xiao R, Guo Y, Zeng X, Wang W, Zou Y, Yu Q, Lan P, Teh BT, Wu X, Tan J. Pharmacological modulation of RB1 activity mitigates resistance to neoadjuvant chemotherapy in locally advanced rectal cancer. Proc Natl Acad Sci U S A 2024; 121:e2304619121. [PMID: 38289962 PMCID: PMC10861914 DOI: 10.1073/pnas.2304619121] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 11/29/2023] [Indexed: 02/01/2024] Open
Abstract
Resistance to neoadjuvant chemotherapy leads to poor prognosis of locally advanced rectal cancer (LARC), representing an unmet clinical need that demands further exploration of therapeutic strategies to improve clinical outcomes. Here, we identified a noncanonical role of RB1 for modulating chromatin activity that contributes to oxaliplatin resistance in colorectal cancer (CRC). We demonstrate that oxaliplatin induces RB1 phosphorylation, which is associated with the resistance to neoadjuvant oxaliplatin-based chemotherapy in LARC. Inhibition of RB1 phosphorylation by CDK4/6 inhibitor results in vulnerability to oxaliplatin in both intrinsic and acquired chemoresistant CRC. Mechanistically, we show that RB1 modulates chromatin activity through the TEAD4/HDAC1 complex to epigenetically suppress the expression of DNA repair genes. Antagonizing RB1 phosphorylation through CDK4/6 inhibition enforces RB1/TEAD4/HDAC1 repressor activity, leading to DNA repair defects, thus sensitizing oxaliplatin treatment in LARC. Our study identifies a RB1 function in regulating chromatin activity through TEAD4/HDAC1. It also provides the combination of CDK4/6 inhibitor with oxaliplatin as a potential synthetic lethality strategy to mitigate oxaliplatin resistance in LARC, whereby phosphorylated RB1/TEAD4 can serve as potential biomarkers to guide the patient stratification.
Collapse
Affiliation(s)
- Zhaoliang Yu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Peng Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Yufeng Chen
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Dezheng Lin
- Department of Endoscopic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510060, People’s Republic of China
| | - Shini Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Jinghan Hong
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
| | - Peiyong Guan
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore138672, Singapore
| | - Jianfeng Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Min-er Zhong
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Jinghong Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Xiaochuan Chen
- Department of Obstetrics and Gynecology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Yichen Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Yali Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Peili Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Zerong Cai
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Jason Yongsheng Chan
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore169610, Singapore
| | - Yulin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Rong Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Yaoyu Guo
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Xian Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
| | - Wenyu Wang
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Yifeng Zou
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Qiang Yu
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore138672, Singapore
| | - Ping Lan
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Bin Tean Teh
- Cancer and Stem Cell Biology Program, Duke–National University of Singapore Medical School, Singapore169857, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore169610, Singapore
| | - Xiaojian Wu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong510655, People’s Republic of China
| | - Jing Tan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong510060, People’s Republic of China
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore169610, Singapore
| |
Collapse
|
48
|
Morrison L, Loibl S, Turner NC. The CDK4/6 inhibitor revolution - a game-changing era for breast cancer treatment. Nat Rev Clin Oncol 2024; 21:89-105. [PMID: 38082107 DOI: 10.1038/s41571-023-00840-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/27/2024]
Abstract
Cyclin-dependent kinase (CDK) 4/6 inhibition in combination with endocrine therapy is the standard-of-care treatment for patients with advanced-stage hormone receptor-positive, HER2 non-amplified (HR+HER2-) breast cancer. These agents can also be administered as adjuvant therapy to patients with higher-risk early stage disease. Nonetheless, the clinical success of these agents has created several challenges, such as how to address acquired resistance, identifying which patients are most likely to benefit from therapy prior to treatment, and understanding the optimal timing of administration and sequencing of these agents. In this Review, we describe the rationale for targeting CDK4/6 in patients with breast cancer, including a summary of updated clinical evidence and how this should inform clinical practice. We also discuss ongoing research efforts that are attempting to address the various challenges created by the widespread implementation of these agents.
Collapse
Affiliation(s)
- Laura Morrison
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
- Breast Unit, The Royal Marsden Hospital, London, UK
| | - Sibylle Loibl
- German Breast Group, Goethe University, Frankfurt, Germany
| | - Nicholas C Turner
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK.
- Breast Unit, The Royal Marsden Hospital, London, UK.
| |
Collapse
|
49
|
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: 13] [Impact Index Per Article: 13.0] [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
|
50
|
Ebrahimnejad P, Mohammadi Z, Babaei A, Ahmadi M, Amirkhanloo S, Asare-Addo K, Nokhodchid A. Novel Strategies Using Sagacious Targeting for Site-Specific Drug Delivery in Breast Cancer Treatment: Clinical Potential and Applications. Crit Rev Ther Drug Carrier Syst 2024; 41:35-84. [PMID: 37824418 DOI: 10.1615/critrevtherdrugcarriersyst.v41.i1.20] [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: 10/14/2023]
Abstract
For more than a decade, researchers have been working to achieve new strategies and smart targeting drug delivery techniques and technologies to treat breast cancer (BC). Nanotechnology presents a hopeful strategy for targeted drug delivery into the building of new therapeutics using the properties of nanomaterials. Nanoparticles are of high regard in the field of diagnosis and the treatment of cancer. The use of these nanoparticles as an encouraging approach in the treatment of various cancers has drawn the interest of researchers in recent years. In order to achieve the maximum therapeutic effectiveness in the treatment of BC, combination therapy has also been adopted, leading to minimal side effects and thus an enhancement in the quality of life for patients. This review article compares, discusses and criticizes the approaches to treat BC using novel design strategies and smart targeting of site-specific drug delivery systems.
Collapse
Affiliation(s)
- Pedram Ebrahimnejad
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Mohammadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amirhossein Babaei
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Melika Ahmadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shervin Amirkhanloo
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kofi Asare-Addo
- Department of Pharmacy, University of Huddersfield, Huddersfield, UK
| | - Ali Nokhodchid
- Lupin Pharmaceutical Research Center, Coral Springs, Florida, USA; Pharmaceutics Research Lab, Arundel Building, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|