1
|
Gao T, Sun Y, Leng P, Liu D, Guo Q, Li J. CDK4/6 inhibitors in breast cancer therapy: mechanisms of drug resistance and strategies for treatment. Front Pharmacol 2025; 16:1549520. [PMID: 40421216 PMCID: PMC12104243 DOI: 10.3389/fphar.2025.1549520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/24/2025] [Indexed: 05/28/2025] Open
Abstract
Dysregulated cell cycle progression is a well-established hallmark of cancer, driving the development of targeted antitumor therapies that intervene at specific phases of the cell cycle. Among these therapeutic targets, cyclin-dependent kinases 4 and 6 (CDK4/6) have emerged as critical regulators of cell cycle progression, with their aberrant activation being strongly implicated in tumorigenesis and cancer progression. Currently, multiple CDK4/6 inhibitors have received clinical approval for hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer, demonstrating dual therapeutic mechanisms through both cell cycle arrest and enhancement of antitumor immunity. However, clinical implementation faces two major challenges: the inevitable development of acquired resistance during prolonged treatment, and the need for optimized combination strategies with other anticancer agents to achieve synergistic efficacy. This review systematically examines the molecular mechanisms underlying CDK4/6 inhibitor function and characterizes currently approved therapeutic agents. Importantly, it synthesizes recent discoveries regarding resistance mechanisms, including dysregulated cell cycle checkpoints, compensatory signaling pathway activation, and tumor microenvironment adaptations. Furthermore, we critically evaluate emerging combination therapeutic approaches targeting these resistance mechanisms. By integrating mechanistic insights with clinical evidence, this analysis aims to provide actionable strategies for overcoming therapeutic resistance and maximizing the clinical potential of CDK4/6 inhibitors in breast cancer management.
Collapse
Affiliation(s)
- Tong Gao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Sun
- Department of Health Management Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ping Leng
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Donghua Liu
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qie Guo
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Li
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Yuan X, Rosen JM. Histone acetylation modulators in breast cancer. Breast Cancer Res 2025; 27:49. [PMID: 40165290 PMCID: PMC11959873 DOI: 10.1186/s13058-025-02006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Breast cancer is the most prevalent cancer in women worldwide. Aberrant epigenetic reprogramming such as dysregulation of histone acetylation has been associated with the development of breast cancer. Histone acetylation modulators have been targeted as potential treatments for breast cancer. This review comprehensively discusses the roles of these modulators and the effects of their inhibitors on breast cancer. In addition, epigenetic reprogramming not only affects breast cancer cells but also the immunosuppressive myeloid cells, which can facilitate breast cancer progression. Therefore, the review also highlights the roles of these immunosuppressive myeloid cells and summarizes how histone acetylation modulators affect their functions and phenotypes. This review provides insights into histone acetylation modulators as potential therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Xueying Yuan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
| |
Collapse
|
3
|
Dai W, Qiao X, Fang Y, Guo R, Bai P, Liu S, Li T, Jiang Y, Wei S, Na Z, Xiao X, Li D. Epigenetics-targeted drugs: current paradigms and future challenges. Signal Transduct Target Ther 2024; 9:332. [PMID: 39592582 PMCID: PMC11627502 DOI: 10.1038/s41392-024-02039-0] [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: 08/02/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Epigenetics governs a chromatin state regulatory system through five key mechanisms: DNA modification, histone modification, RNA modification, chromatin remodeling, and non-coding RNA regulation. These mechanisms and their associated enzymes convey genetic information independently of DNA base sequences, playing essential roles in organismal development and homeostasis. Conversely, disruptions in epigenetic landscapes critically influence the pathogenesis of various human diseases. This understanding has laid a robust theoretical groundwork for developing drugs that target epigenetics-modifying enzymes in pathological conditions. Over the past two decades, a growing array of small molecule drugs targeting epigenetic enzymes such as DNA methyltransferase, histone deacetylase, isocitrate dehydrogenase, and enhancer of zeste homolog 2, have been thoroughly investigated and implemented as therapeutic options, particularly in oncology. Additionally, numerous epigenetics-targeted drugs are undergoing clinical trials, offering promising prospects for clinical benefits. This review delineates the roles of epigenetics in physiological and pathological contexts and underscores pioneering studies on the discovery and clinical implementation of epigenetics-targeted drugs. These include inhibitors, agonists, degraders, and multitarget agents, aiming to identify practical challenges and promising avenues for future research. Ultimately, this review aims to deepen the understanding of epigenetics-oriented therapeutic strategies and their further application in clinical settings.
Collapse
Affiliation(s)
- Wanlin Dai
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinbo Qiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Fang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Renhao Guo
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Bai
- Department of Forensic Genetics, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Shuang Liu
- Shenyang Maternity and Child Health Hospital, Shenyang, China
| | - Tingting Li
- Department of General Internal Medicine VIP Ward, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yutao Jiang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuang Wei
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhijing Na
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
| | - Xue Xiao
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China.
| | - Da Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China.
| |
Collapse
|
4
|
Wu S, Xu J, Ma Y, Liang G, Wang J, Sun T. Advances in the mechanism of CDK4/6 inhibitor resistance in HR+/HER2- breast cancer. Ther Adv Med Oncol 2024; 16:17588359241282499. [PMID: 39371618 PMCID: PMC11450575 DOI: 10.1177/17588359241282499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/24/2024] [Indexed: 10/08/2024] Open
Abstract
Among women, breast cancer is the most prevalent form of a malignant tumour. Among the subtypes of breast cancer, hormone receptor (HR) positive and human epidermal growth factor receptor (HER2) negative kinds make up the biggest proportion. The advent of cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors, which are dependent on cell cycle proteins, has greatly enhanced the prognosis of patients with advanced HR+/HER2- breast cancer. This is a specific treatment that stops the growth of cancer cells by preventing them from dividing. Nevertheless, the drug resistance of the disease unavoidably impacts the effectiveness of treatment and the prognosis of patients. This report provides a thorough analysis of the current research advancements about the resistance mechanism of CDK4/6 inhibitors in HR+/HER2- breast cancer. It presents an in-depth discussion from numerous viewpoints, such as aberrant cell cycle regulation and changes in signalling pathways. In response to the drug resistance problem, subsequent treatment strategies are also being explored, including switching to other CDK4/6 inhibitor drugs, a combination of novel endocrine therapeutic agents, an optimal combination of targeted therapies and switching to chemotherapy. An in-depth study of the resistance mechanism can assist in identifying creative tactics that can overcome or postpone drug resistance, alleviate the problem of restricted treatment strategies following drug resistance and enhance the prognosis of patients.
Collapse
Affiliation(s)
- Sijia Wu
- Breast Medicine Section One, Liaoning Cancer Hospital, Shenyang, Liaoning, China
| | - Junnan Xu
- Breast Medicine Section One, Liaoning Cancer Hospital, Shenyang, Liaoning, China
| | - Yiwen Ma
- Breast Medicine Section One, Liaoning Cancer Hospital, Shenyang, Liaoning, China
| | - Guilian Liang
- Breast Medicine Section One, Liaoning Cancer Hospital, Shenyang, Liaoning, China
| | - Jiaxing Wang
- Breast Medicine Section One, Liaoning Cancer Hospital, Shenyang, Liaoning, China
| | - Tao Sun
- Breast Medicine Section One, Liaoning Cancer Hospital, Shenyang, Liaoning 110000, China
| |
Collapse
|
5
|
Xu T, Xiong W, Zhang L, Yuan Y. The Effects of Anlotinib Combined with Chemotherapy following Progression on Cyclin-Dependent Kinase 4/6 Inhibitor in Hormone Receptor-Positive Metastatic Breast Cancer. Breast J 2024; 2024:5396107. [PMID: 39742382 PMCID: PMC11315966 DOI: 10.1155/2024/5396107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/20/2024] [Accepted: 07/20/2024] [Indexed: 01/03/2025]
Abstract
Purpose Endocrine therapy combined with cyclin-dependent kinase (CDK) 4/6 inhibitors (CDK4/6i) is the preferred treatment for hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (MBC). However, there are currently no recommendations for therapeutic strategies after progression on CDK4/6i-based treatment. This study aimed to examine the efficacy and safety of anlotinib plus chemotherapy in HR+/HER2- MBC after progression on CDK4/6 inhibitors. Methods We collected data from 32 patients with HR+/HER2- MBC treated with anlotinib plus chemotherapy after progressing on CDK4/6i at Jiangsu Cancer Hospital from March 2020 to October 2023. The median follow-up was 9.1 months (range, 2.0-19.7 months) as of the data cutoff date in October 2023. The primary endpoint was median progression-free survival (PFS); secondary endpoints included objective response rate (ORR), disease control rate (DCR), and adverse events. Results The median PFS (mPFS) of all patients was 7.6 months (95% confidence interval (CI), 5.75-9.45). There was no significant difference in mPFS between patients who responded to prior CDK4/6i treatment and those who did not (8.3 months vs. 6.8 months, p=0.580). Besides, the ORR was 34.4% and DCR was 93.8%. The most frequently observed adverse events were anemia (50.0%), neutropenia (40.6%), thrombocytopenia (34.4%), and epistaxis (34.4%). Dose interruption or reductions due to adverse events occurred in 2 (6.3%) and 5 (15.6%) patients, respectively. Conclusions The study preliminarily demonstrates that anlotinib combined with chemotherapy may be an optional recommendation for patients with HR+/HER2- metastatic breast cancer who have progressed after CDK4/6i.
Collapse
Affiliation(s)
- Ting Xu
- Department of OncologyThe Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Weili Xiong
- Department of OncologyThe Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Lili Zhang
- Department of ChemotherapyJiangsu Cancer HospitalJiangsu Institute of Cancer ResearchThe Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan Yuan
- Department of ChemotherapyJiangsu Cancer HospitalJiangsu Institute of Cancer ResearchThe Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
Suo J, Zhu K, Zhuang C, Zhong X, Bravaccini S, Maltoni R, Bertucci F, Zheng H, Luo T. Efficacy and safety of tucidinostat in patients with advanced hormone receptor-positive human epidermal growth factor receptor 2-negative breast cancer: real-world insights. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:409. [PMID: 38213803 PMCID: PMC10777213 DOI: 10.21037/atm-23-1913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024]
Abstract
Background Tucidinostat, which is a subtype-selective histone deacetylase inhibitor, has been approved in China for the treatment of hormone receptor-positive (HR+) human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer (ABC). However, existing evidence mainly stemmed from randomized controlled trials, and might have limitations in representing the complexities of clinical practice and diverse patient populations. Therefore, there is a need to explore the efficacy and optimal therapeutic modality for tucidinostat in real-world clinical settings. Methods The objective of this real-world study was to analyze the clinical data of 47 patients with HR+/HER2- ABC who received tucidinostat treatment at West China Hospital, Sichuan University, between August 2020 and May 2023. The primary outcomes were progression-free survival (PFS) and clinical benefit rate [CBR; defined as partial response (PR) and stable disease (SD) for ≥6 months on clinical evaluation]. Results A total of 47 patients were included, and the median follow-up time was 18.20 months. The median line of tucidinostat therapy was 3 (range, 1-9). In all, 52.17% patients were treated with tucidinostat plus fulvestrant, while 38.30% were treated with tucidinostat plus aromatase inhibitors. Notably, 10.64% of the patients with rapidly progressing visceral metastases received tucidinostat plus endocrine therapy as maintenance treatment after achieving disease control with chemotherapy. The median PFS was 4.43 months [95% confidence interval (CI), 2.77-10.53], and the median overall survival was 19.57 months (95% CI, 12.83-not reached). The 6-month CBR for the overall population was 41.86%. Patients undergoing maintenance therapy demonstrated a significantly longer PFS than did those who did not receive it as maintenance therapy (14.13 vs. 3.93 months; P=0.01). Univariate Cox regression analysis showed that use of tucidinostat in lines 1-2, use of tucidinostat plus fulvestrant, presence of one metastatic site, and lack of brain metastasis were favorable factors for PFS. Thrombocytopenia was the most frequently reported adverse event, with an incidence rate of 31.91% at all grades and 14.89% at grade ≥3. Four (8.51%) patients discontinued the treatment. Conclusions For patients with HR+/HER2- ABC, tucidinostat combination therapy offers certain survival benefits with controllable safety. Furthermore, compared with non-maintenance therapy, maintenance therapy after chemotherapy may have promising efficacy.
Collapse
Affiliation(s)
- Jiaojiao Suo
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kunrui Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chunying Zhuang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaorong Zhong
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Center, West China Hospital, Sichuan University, Chengdu, China
| | - Sara Bravaccini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Roberta Maltoni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - François Bertucci
- Department of Medical Oncology, Cancer Research Center of Marseille, Paoli-Calmettes Institute, Aix-Marseille University, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Hong Zheng
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Luo
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Breast Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Neven P, Dullens L, Han S, Deblander A, Van Herck Y, Van Houdt M, Wildiers H. Navigating next-generation HR +/HER2 - metastatic breast cancer therapies: a critical commentary on abemaciclib vs. tucidinostat after palbociclib progression. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2023; 4:31. [PMID: 38751459 PMCID: PMC11093079 DOI: 10.21037/tbcr-23-41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/11/2023] [Indexed: 05/18/2024]
Affiliation(s)
- Patrick Neven
- Department of Oncology, KU Leuven-University of Leuven, Leuven, Belgium
- Department of Gynaecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Lieke Dullens
- Master Student of Medicine, KU Leuven-University of Leuven, Leuven, Belgium
| | - Sileny Han
- Department of Oncology, KU Leuven-University of Leuven, Leuven, Belgium
- Department of Gynaecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Anne Deblander
- Department of Gynaecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Yannick Van Herck
- Laboratory of Experimental Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Maxime Van Houdt
- Department of Gynaecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Hans Wildiers
- Department of Oncology, KU Leuven-University of Leuven, Leuven, Belgium
- Department of Gynaecological Oncology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Gomes I, Abreu C, Costa L, Casimiro S. The Evolving Pathways of the Efficacy of and Resistance to CDK4/6 Inhibitors in Breast Cancer. Cancers (Basel) 2023; 15:4835. [PMID: 37835528 PMCID: PMC10571967 DOI: 10.3390/cancers15194835] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
The approval of cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) in combination with endocrine therapy (ET) has remarkably improved the survival outcomes of patients with advanced hormone receptor-positive (HR+) breast cancer (BC), becoming the new standard of care treatment in these patients. Despite the efficacy of this therapeutic combination, intrinsic and acquired resistance inevitably occurs and represents a major clinical challenge. Several mechanisms associated with resistance to CDK4/6i have been identified, including both cell cycle-related and cell cycle-nonspecific mechanisms. This review discusses new insights underlying the mechanisms of action of CDK4/6i, which are more far-reaching than initially thought, and the currently available evidence of the mechanisms of resistance to CDK4/6i in BC. Finally, it highlights possible treatment strategies to improve CDK4/6i efficacy, summarizing the most relevant clinical data on novel combination therapies involving CDK4/6i.
Collapse
Affiliation(s)
- Inês Gomes
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Catarina Abreu
- Oncology Division, Hospital de Santa Maria—Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal;
| | - Luis Costa
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
- Oncology Division, Hospital de Santa Maria—Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal;
| | - Sandra Casimiro
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| |
Collapse
|
9
|
Lee RS, Sad K, Fawwal DV, Spangle JM. Emerging Role of Epigenetic Modifiers in Breast Cancer Pathogenesis and Therapeutic Response. Cancers (Basel) 2023; 15:4005. [PMID: 37568822 PMCID: PMC10417282 DOI: 10.3390/cancers15154005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
Breast cancer pathogenesis, treatment, and patient outcomes are shaped by tumor-intrinsic genomic alterations that divide breast tumors into molecular subtypes. These molecular subtypes often dictate viable therapeutic interventions and, ultimately, patient outcomes. However, heterogeneity in therapeutic response may be a result of underlying epigenetic features that may further stratify breast cancer patient outcomes. In this review, we examine non-genetic mechanisms that drive functional changes to chromatin in breast cancer to contribute to cell and tumor fitness and highlight how epigenetic activity may inform the therapeutic response. We conclude by providing perspectives on the future of therapeutic targeting of epigenetic enzymes, an approach that holds untapped potential to improve breast cancer patient outcomes.
Collapse
Affiliation(s)
- Richard Sean Lee
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (R.S.L.); (K.S.); (D.V.F.)
- Department of Biology, Emory College, Atlanta, GA 30322, USA
| | - Kirti Sad
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (R.S.L.); (K.S.); (D.V.F.)
| | - Dorelle V. Fawwal
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (R.S.L.); (K.S.); (D.V.F.)
- Biochemistry, Cell & Developmental Biology Graduate Program, Emory University School of Medicine, Atlanta, GA 30311, USA
| | - Jennifer Marie Spangle
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (R.S.L.); (K.S.); (D.V.F.)
| |
Collapse
|
10
|
Zhao M, Hanson KA, Zhang Y, Zhou A, Cha-Silva AS. Place in Therapy of Cyclin-Dependent Kinase 4/6 Inhibitors in Breast Cancer: A Targeted Literature Review. Target Oncol 2023; 18:327-358. [PMID: 37074594 DOI: 10.1007/s11523-023-00957-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2023] [Indexed: 04/20/2023]
Abstract
Cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) are the preferred regimen for patients with hormone receptor-positive and human epidermal growth factor receptor 2-negative (HR+/HER2-) advanced or metastatic breast cancer. However, the optimal treatment sequencing for CDK4/6i with other available therapeutic options is unclear. We conducted a targeted literature review to identify the current evidence on CDK4/6i treatment patterns in patients with breast cancer. The search was initially conducted in October 2021 and subsequently updated in October 2022. Biomedical databases and gray literature were searched, and bibliographies of included reviews were screened for relevant studies. The search identified ten reviews published since 2021 and 87 clinical trials or observational studies published since 2015. The included reviews discussed CDK4/6i usage with or without endocrine therapy (ET) in first-line and second-line treatment for patients with HR+/HER2- advanced or metastatic breast cancer, followed by ET, chemotherapy, or targeted therapy with ET. Clinical studies reported similar treatment sequences consisting of ET, chemotherapy, or targeted therapy with ET prior to CDK4/6i with ET, followed by ET monotherapy, chemotherapy, targeted therapy with ET, or continued CDK4/6i with ET. Current evidence suggests CDK4/6i are effective for HR+/HER2- advanced or metastatic breast cancer in earlier lines of therapy. Efficacy of CDK4/6i as measured by progression-free survival and overall survival was similar within a line of therapy regardless of the type of prior therapy. Survival on different post-CDK4/6i treatments was also similar within the same line of therapy. Additional research is needed to investigate the optimal place in therapy of CDK4/6i and the sequencing of treatments following progression on CDK4/6i.
Collapse
Affiliation(s)
- Melody Zhao
- EVERSANA, 113-3228 South Service Road, Burlington, ON, L9N 3H8, Canada.
| | | | - Yixie Zhang
- EVERSANA, 113-3228 South Service Road, Burlington, ON, L9N 3H8, Canada
| | - Anna Zhou
- EVERSANA, 113-3228 South Service Road, Burlington, ON, L9N 3H8, Canada
| | | |
Collapse
|
11
|
Lian B, Chen X, Shen K. Inhibition of histone deacetylases attenuates tumor progression and improves immunotherapy in breast cancer. Front Immunol 2023; 14:1164514. [PMID: 36969235 PMCID: PMC10034161 DOI: 10.3389/fimmu.2023.1164514] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Breast cancer is one of the common malignancies with poor prognosis worldwide. The treatment of breast cancer patients includes surgery, radiation, hormone therapy, chemotherapy, targeted drug therapy and immunotherapy. In recent years, immunotherapy has potentiated the survival of certain breast cancer patients; however, primary resistance or acquired resistance attenuate the therapeutic outcomes. Histone acetyltransferases induce histone acetylation on lysine residues, which can be reversed by histone deacetylases (HDACs). Dysregulation of HDACs via mutation and abnormal expression contributes to tumorigenesis and tumor progression. Numerous HDAC inhibitors have been developed and exhibited the potent anti-tumor activity in a variety of cancers, including breast cancer. HDAC inhibitors ameliorated immunotherapeutic efficacy in cancer patients. In this review, we discuss the anti-tumor activity of HDAC inhibitors in breast cancer, including dacinostat, belinostat, abexinostat, mocetinotat, panobinostat, romidepsin, entinostat, vorinostat, pracinostat, tubastatin A, trichostatin A, and tucidinostat. Moreover, we uncover the mechanisms of HDAC inhibitors in improving immunotherapy in breast cancer. Furthermore, we highlight that HDAC inhibitors might be potent agents to potentiate immunotherapy in breast cancer.
Collapse
Affiliation(s)
| | | | - Kunwei Shen
- *Correspondence: Xiaosong Chen, ; Kunwei Shen,
| |
Collapse
|