1
|
Li J. Exploring the Potential of Adjuvant CDK4/6 Inhibitors in Hormone Receptor-Positive Early Breast Cancer: A Consistent Approach for All. Cancers (Basel) 2025; 17:561. [PMID: 40002156 PMCID: PMC11852482 DOI: 10.3390/cancers17040561] [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: 12/05/2024] [Revised: 01/05/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hormone receptor-positive, HER2-negative breast cancer is the most common subtype, with endocrine therapy as the standard treatment. Despite the advancements in adjuvant endocrine therapy, recurrence remains a challenge, particularly in high-risk patients. Recent trials on cyclin D kinase 4/6 (CDK4/6) inhibitors in adjuvant therapy have shown promise in reducing early recurrence and improving survival. METHODS This review analyzes the clinical evidence supporting the use of CDK4/6 inhibitors, focusing on the NATALEE and monarchE trials, which demonstrate comparable efficacy and manageable safety profiles for ribociclib and abemaciclib. RESULTS AND CONCLUSIONS Ribociclib, with its broader applicability and impact on the decision making for axillary lymph node surgery, may be the preferred option in high-risk populations. The review also addresses unanswered clinical questions and highlights the need for ongoing research to optimize the adjuvant therapy strategies.
Collapse
Affiliation(s)
- Jianbin Li
- Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing 100071, China;
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| |
Collapse
|
2
|
Xu L, Yang L, Zhang D, Wu Y, Shan J, Zhu H, Lian Z, He G, Wang C, Wang Q. Multi-omics analysis reveals the unique landscape of DLD in the breast cancer tumor microenvironment and its implications for immune-related prognosis. Comput Struct Biotechnol J 2024; 23:1201-1213. [PMID: 38545600 PMCID: PMC10966406 DOI: 10.1016/j.csbj.2024.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 11/11/2024] Open
Abstract
Background Cuproptosis, i.e., copper-induced programmed cell death, has potential implications in cancer therapy. However, the impact of the cuproptosis-related gene (CRG) dihydrolipoyl dehydrogenase (DLD) on breast cancer (BC) prognosis remains underexplored. Methods We employed real-time quantitative PCR and multiplexed immunostaining techniques to quantify DLD expression in both BC and the adjacent non-cancerous tissues. Immunofluorescence analysis was employed to assess the influence of DLD on immune cells and immunological checkpoints in the BC microenvironment. DLD knockdown experiments were conducted in BC cell lines MDA-MB-468 and SK-BR-3, with knockdown efficiency validated via western blot. Subsequently, we performed the cell counting kit-8 (CCK-8) assay, clone formation assay, Transwell migration assay, and invasion assay. To construct a prognostic model, we employed a Lasso-Cox regression analysis of immune-related genes associated with DLD. Additionally, we established a competing endogenous RNA network based on CRGs to evaluate potential regulatory pathways. Results Compared to the adjacent tissues, BC tissues exhibited markedly elevated DLD expression levels. In vitro experiments demonstrated that DLD knockdown effectively inhibited BC cell migration, invasion, and proliferation. DLD exhibited positive correlations with CD68+ macrophages and PD-L1 in the tumor, as well as with macrophages and CD4+ T cells in the stroma. Tumor regions with high DLD expression were enriched in PD-L1 and macrophages, while stromal regions with high DLD expression contained CD4+ T cells and macrophages. The AUC values for 1-, 3-, and 5-year overall survival in TCGA-BRCA training set were 0.67, 0.66, and 0.66, respectively. A nomogram with a C-index of 0.715 indicated that risk score, tumor stage, and age could serve as independent prognostic factors for BC. Conclusion Our findings underscore the significant predictive significance of DLD in BC and its influence on the tumor microenvironment. DLD represents a promising diagnostic and prognostic marker for BC, offering novel avenues for the identification of therapeutic targets and the enhancement of immunotherapy in BC.
Collapse
Affiliation(s)
- Lijun Xu
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Lei Yang
- Department of Clinical Biobank & Institute of Oncology, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Dan Zhang
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yunxi Wu
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Jiali Shan
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Huixia Zhu
- Department of Biochemistry, Medical College, Nantong University, Nantong, China
| | - Zhengyi Lian
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Guying He
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Chongyu Wang
- Department of Medicine, Xinglin College, Nantong University, Nantong, China
| | - Qingqing Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
3
|
Keskinkilic M, Arayici ME, Basbinar Y, Ellidokuz H, Yavuzsen T, Oztop I. The efficacy and safety of CDK4/6 inhibitors combined with endocrine therapy versus endocrine therapy alone in the adjuvant treatment of patients with high-risk invasive HR+/HER2-early breast cancer: A comprehensive updated meta-analysis of randomized clinical trials. Breast 2024; 78:103815. [PMID: 39413680 PMCID: PMC11752115 DOI: 10.1016/j.breast.2024.103815] [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: 04/03/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND This paper aimed to evaluate the effectiveness of incorporating CDK 4/6 inhibitors (CDK4/6i) into ET for the adjuvant treatment of HR + HER2-resected early-stage breast cancer (ESBC) patients, employing meta-analysis. METHODS In this paper, we compiled randomized clinical trials focusing on CDK4/6i used in the adjuvant treatment of high-risk invasive HR-positive and HER2-ESBC patients. A meta-analysis was performed in line with the PRISMA guidelines. RESULTS We identified four clinical trials that met our inclusion criteria and were published between 2020 and 2024. These trials involved a combined sample size of 17,749 patients diagnosed with breast cancer. The data obtained from the pooled analysis revealed a remarkable beneficial trend in terms of invasive disease-free survival (iDFS) for the use of ET in combination with CDK4/6i compared to the group receiving ET alone (HR = 0.81, 95 % CI: 0.67-0.98, p = 0.03). Of note, CDK4/6 inhibitors demonstrated a notably beneficial effect in both grade 2 (HR = 0.69, 95 % CI: 0.59-0.81, p < 0.001) and grade 3 (HR = 0.76, 95 % CI: 0.65-0.89, p < 0.001). Significant improvements were noted in terms of distant relapse-free survival (dRFS) in the groups treated with abemaciclib and ribociclib (HR = 0.65, 95 % CI: 0.56-0.76, p < 0.001; HR = 0.72, 95 % CI: 0.58-0.89, p = 0.003, respectively). CDK4/6i didn't yield a statistically significant difference in overall survival (OS) (HR = 0.96, 95 % CI: 0.77-1.19, p = 0.69). The use of CDK4/6i with ET was associated with an increased risk of adverse events, particularly anemia and neutropenia, compared with ET alone (OR = 9.12, 95 % CI = 5.04-16.48, p < 0.001). CONCLUSION The findings of this paper demonstrate a significant improvement in iDFS when ET is combined with CDK4/6i, compared to ET alone. Specifically, treatments with abemaciclib and ribociclib showed notable enhancements in dRFS.
Collapse
Affiliation(s)
- Merve Keskinkilic
- Department of Medical Oncology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkiye.
| | - Mehmet Emin Arayici
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkiye.
| | - Yasemin Basbinar
- Department of Translational Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkiye.
| | - Hulya Ellidokuz
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkiye; Department of Preventive Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkiye.
| | - Tugba Yavuzsen
- Department of Medical Oncology, Institute of Oncology, Dokuz Eylul University, İzmir, Turkiye.
| | - Ilhan Oztop
- Department of Medical Oncology, Institute of Oncology, Dokuz Eylul University, İzmir, Turkiye.
| |
Collapse
|
4
|
Lișcu HD, Verga N, Atasiei DI, Badiu DC, Dumitru AV, Ultimescu F, Pavel C, Stefan RE, Manole DC, Ionescu AI. Biomarkers in Colorectal Cancer: Actual and Future Perspectives. Int J Mol Sci 2024; 25:11535. [PMID: 39519088 PMCID: PMC11546354 DOI: 10.3390/ijms252111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Biomarkers in colorectal cancer (CRC) are of great interest in the current literature due to improvements in techniques such as liquid biopsy and next-generation sequencing (NGS). However, screening methods vary globally, with multi-target stool DNA (mt-sDNA) predominantly used in the USA and, more recently, the Cologuard Plus; biomarkers such as the Galectins family and septins show promise in early detection. Gut microbiome assessments, such as Fusobacterium nucleatum, are under intense exploration. Diagnostic tests, such as circulating DNA analysis via NGS, exhibit effectiveness and are being increasingly adopted. Circulating tumor cells emerge as potential alternatives to traditional methods in terms of diagnosis and prognosis. Predictive biomarkers are well established in guidelines; nonetheless, with the aid of machine learning and artificial intelligence, these biomarkers may be improved. This review critically explores the actual dynamic landscape of CRC biomarkers and future, promising biomarkers involved in screening, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Horia-Dan Lișcu
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Nicolae Verga
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Dimitrie-Ionuț Atasiei
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
| | - Dumitru-Cristinel Badiu
- Department of Surgery, Bagdasar Arseni Clinical Emergency Hospital, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Adrian Vasile Dumitru
- Department of Pathology, University Emergency Hospital Bucharest, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Flavia Ultimescu
- Department of Pathology, Institute of Oncology Alexandru Trestioreanu, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Christopher Pavel
- Department of Gastroenterology, Floreasca Emergency Hospital Bucharest, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Roxana-Elena Stefan
- General Surgery Department, Clinic of General and Esophageal Surgery, Sf. Maria Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Diandra-Carmen Manole
- Department of Endocrinology, Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Andreea-Iuliana Ionescu
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| |
Collapse
|
5
|
Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, Luo P, Cheng Q, Xu H, Han X. Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduct Target Ther 2024; 9:270. [PMID: 39389953 PMCID: PMC11467208 DOI: 10.1038/s41392-024-01955-5] [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: 02/24/2024] [Revised: 07/18/2024] [Accepted: 08/24/2024] [Indexed: 10/12/2024] Open
Abstract
The cascade of metastasis in tumor cells, exhibiting organ-specific tendencies, may occur at numerous phases of the disease and progress under intense evolutionary pressures. Organ-specific metastasis relies on the formation of pre-metastatic niche (PMN), with diverse cell types and complex cell interactions contributing to this concept, adding a new dimension to the traditional metastasis cascade. Prior to metastatic dissemination, as orchestrators of PMN formation, primary tumor-derived extracellular vesicles prepare a fertile microenvironment for the settlement and colonization of circulating tumor cells at distant secondary sites, significantly impacting cancer progression and outcomes. Obviously, solely intervening in cancer metastatic sites passively after macrometastasis is often insufficient. Early prediction of metastasis and holistic, macro-level control represent the future directions in cancer therapy. This review emphasizes the dynamic and intricate systematic alterations that occur as cancer progresses, illustrates the immunological landscape of organ-specific PMN creation, and deepens understanding of treatment modalities pertinent to metastasis, thereby identifying some prognostic and predictive biomarkers favorable to early predict the occurrence of metastasis and design appropriate treatment combinations.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingqi Chen
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|
6
|
Henze M, Stuckey BGA. Endocrine consequences of breast cancer therapy and survivorship. Climacteric 2024; 27:333-339. [PMID: 38867405 DOI: 10.1080/13697137.2024.2354725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/01/2024] [Accepted: 04/28/2024] [Indexed: 06/14/2024]
Abstract
Breast cancer survivorship is increasing, due to earlier diagnosis of the disease and more effective therapies. Long-term endocrine sequelae, including early menopause, bone health, fertility implications and menopausal symptoms, are important survivorship issues. Ovarian failure is common with chemotherapy and options for preserving fertility in young women include ovarian suppression during chemotherapy and oocyte or embryo cryopreservation before chemotherapy. Tamoxifen as adjunct therapy in premenopausal women leads to ovarian stimulation, sometimes ovulation and occasionally pregnancy with important teratogenic implications. Aromatase inhibitor therapy with or without gonadotrophin releasing hormone (GnRH) agonist leads to profound bone loss and anti-resorptive therapy is advised to prevent fracture. Tamoxifen acts to preserve bone in postmenopausal women but not premenopausal women. Pregnancy is not discouraged in young women with early breast cancer, even to the point of pausing adjunct therapy in order to conceive. However, menopausal hormone therapy is discouraged even years later. Non-hormonal therapy for menopausal symptoms in breast cancer survivors is available but, in some cases, estrogen-containing therapy may be worthy of consideration for quality of life in the informed patient.
Collapse
Affiliation(s)
- Meg Henze
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Bronwyn G A Stuckey
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Keogh Institute for Medical Research, Nedlands, WA, Australia
- Medical School, University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
7
|
Pan J, Tong F, Ren N, Ren L, Yang Y, Gao F, Xu Q. Role of N 6‑methyladenosine in the pathogenesis, diagnosis and treatment of prostate cancer (Review). Oncol Rep 2024; 51:88. [PMID: 38757383 PMCID: PMC11110010 DOI: 10.3892/or.2024.8747] [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: 11/02/2023] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Prostate cancer (PCa) affects males of all racial and ethnic groups, and leads to higher rates of mortality in those belonging to a lower socioeconomic status due to the late detection of the disease. PCa affects middle‑aged males between the ages of 45 and 60 years, and is the highest cause of cancer‑associated mortality in Western countries. As the most abundant and common mRNA modification in higher eukaryotes, N6‑methyladenosine (m6A) is widely distributed in mammalian cells and influences various aspects of mRNA metabolism. Recent studies have found that abnormal expression levels of various m6A regulators significantly affect the development and progression of various types of cancer, including PCa. The present review discusses the influence of m6A regulatory factors on the pathogenesis and progression of PCa through mRNA modification based on the current state of research on m6A methylation modification in PCa. It is considered that the treatment of PCa with micro‑molecular drugs that target the epigenetics of the m6A regulator to correct abnormal m6A modifications is a direction for future research into current diagnostic and therapeutic approaches for PCa.
Collapse
Affiliation(s)
- Junjie Pan
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310051, P.R. China
| | - Fei Tong
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Ning Ren
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310051, P.R. China
| | - Lanqi Ren
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310051, P.R. China
| | - Yibei Yang
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310051, P.R. China
| | - Feng Gao
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, P.R. China
| | - Qiaoping Xu
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
8
|
Wang H, Wang R, Shen K, Huang R, Wang Z. Biological Roles and Clinical Applications of Exosomes in Breast Cancer: A Brief Review. Int J Mol Sci 2024; 25:4620. [PMID: 38731840 PMCID: PMC11083446 DOI: 10.3390/ijms25094620] [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: 03/14/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Breast cancer (BC) is a global health risk for women and has a high prevalence rate. The drug resistance, recurrence, and metastasis of BC affect patient prognosis, thus posing a challenge to scientists. Exosomes are extracellular vesicles (EVs) that originate from various cells; they have a double-layered lipid membrane structure and contain rich biological information. They mediate intercellular communication and have pivotal roles in tumor development, progression, and metastasis and drug resistance. Exosomes are important cell communication mediators in the tumor microenvironment (TME). Exosomes are utilized as diagnostic and prognostic biomarkers for estimating the treatment efficacy of BC and have the potential to function as tools to enable the targeted delivery of antitumor drugs. This review introduces recent progress in research on how exosomes influence tumor development and the TME. We also present the research progress on the application of exosomes as prognostic and diagnostic biomarkers and drug delivery tools.
Collapse
Affiliation(s)
| | | | | | - Renhong Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (H.W.); (R.W.); (K.S.)
| | - Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (H.W.); (R.W.); (K.S.)
| |
Collapse
|
9
|
Nedyalkova M, Robeva R, Romanova J, Yovcheva K, Lattuada M, Simeonov V. In silico screening of potential agonists of a glucagon-like peptide-1 receptor among female sex hormone derivatives. J Biomol Struct Dyn 2024:1-12. [PMID: 38587907 DOI: 10.1080/07391102.2024.2330714] [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: 08/16/2023] [Accepted: 03/09/2024] [Indexed: 04/10/2024]
Abstract
Glucagon-like peptide-1 (GLP-1) is an intestinal hormone that exerts its pleiotropic effects through a specific GLP-1 receptor (GLP-1R). The hormone-receptor complex might regulate glucose-dependent insulin secretion, and energy homeostasis; moreover, it could decrease inflammation and provide cardio- and neuroprotection. Additionally, the beneficial influence of GLP-1 on obesity in women might lead to improvement of their ovarian function. The links between metabolism and reproduction are tightly connected, and it is not surprising that different estrogen derivatives, estrogen-receptor modulator (SERM) and progestins used for gonadal and oncological disorders might influence carbohydrate and lipid metabolism. However, their possible influence on the GLP-1R has not been studied. The docking scores and top-ranked poses of raloxifene were much higher than those observed for other investigated SERMs and estradiol per se. Among different studied progestins, drospirenone showed slightly higher affinity to GLP-1R. Herein, the same data set of the drugs is evaluated by molecular dynamics (MD) simulations and compared with the obtained docking result. Notably, it is demonstrated that the used docking protocol and the applied MD calculations ranked the same ligand (raloxifene) as the best one. In the present study, raloxifene might exert an allosteric influence on GLP-1R signaling, which might contribute to potential beneficial effects on metabolism and weight regulation. However, further experimental and clinical studies are needed to reveal if the GLP-1R modulation has a real biological impact.
Collapse
Affiliation(s)
- Miroslava Nedyalkova
- Department of Chemistry, Fribourg University, Fribourg, Switzerland
- Department of Inorganic Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia 'St. Kl. Ohridski', Sofia, Bulgaria
- Swiss National Center for Competence in Research (NCCR) Bio-inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Ralitsa Robeva
- Department of Endocrinology, Faculty of Medicine, Medical University-Sofia, Sofia, Bulgaria
| | - Julia Romanova
- Department of Inorganic Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia 'St. Kl. Ohridski', Sofia, Bulgaria
| | - Kirila Yovcheva
- Department of Inorganic Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia 'St. Kl. Ohridski', Sofia, Bulgaria
| | - Marco Lattuada
- Department of Chemistry, Fribourg University, Fribourg, Switzerland
| | - Vasil Simeonov
- Department of Inorganic Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia 'St. Kl. Ohridski', Sofia, Bulgaria
| |
Collapse
|
10
|
Pescia C, Guerini-Rocco E, Viale G, Fusco N. Advances in Early Breast Cancer Risk Profiling: From Histopathology to Molecular Technologies. Cancers (Basel) 2023; 15:5430. [PMID: 38001690 PMCID: PMC10670146 DOI: 10.3390/cancers15225430] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Early breast cancer (BC) is the definition applied to breast-confined tumors with or without limited involvement of locoregional lymph nodes. While risk stratification is essential for guiding clinical decisions, it can be a complex endeavor in these patients due to the absence of comprehensive guidelines. Histopathological analysis and biomarker assessment play a pivotal role in defining patient outcomes. Traditional histological criteria such as tumor size, lymph node involvement, histological type and grade, lymphovascular invasion, and immune cell infiltration are significant prognostic indicators. In addition to the hormone receptor, HER2, and-in specific scenarios-BRCA1/2 testing, molecular subtyping through gene expression profiling provides valuable insights to tailor clinical decision-making. The emergence of "omics" technologies, applicable to both tissue and liquid biopsy samples, has broadened our arsenal for evaluating the risk of early BC. However, a pressing need remains for standardized methodologies and integrated pathological models that encompass multiple analytical dimensions. In this study, we provide a detailed examination of the existing strategies for early BC risk stratification, intending to serve as a practical guide for histopathologists and molecular pathologists.
Collapse
Affiliation(s)
- Carlo Pescia
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- School of Pathology, University of Milan, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| | - Giuseppe Viale
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| |
Collapse
|
11
|
Cucciniello L, Garufi G, Di Rienzo R, Martinelli C, Pavone G, Giuliano M, Arpino G, Montemurro F, Del Mastro L, De Laurentiis M, Puglisi F. Estrogen deprivation effects of endocrine therapy in breast cancer patients: Incidence, management and outcome. Cancer Treat Rev 2023; 120:102624. [PMID: 37751658 DOI: 10.1016/j.ctrv.2023.102624] [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: 06/23/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
Endocrine therapy is one of the standard adjuvant treatments to reduce the risk of recurrence and mortality in patients with hormone receptor positive early breast cancer. Despite its proven efficacy, ET side effects, which persist over time even if low grade, may deteriorate quality of life. During follow-up visits, emphasis is generally placed on the risk of disease recurrence, while the topic of ET side effects is commonly neglected and discussed only briefly. This could lead to poor adherence to therapy and early treatment discontinuation, resulting in worse survival outcomes. The aim of this review is to provide an overview of the available evidence on the incidence and reporting of ET-related side effects (including vasomotor symptoms, musculoskeletal disorders and genitourinary syndrome of menopause, as well as fatigue, psychological and ocular disorders, dysmetabolic effects and loss of bone density) and of the pharmacological and non-pharmacological strategies available to mitigate symptom burden.
Collapse
Affiliation(s)
- Linda Cucciniello
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| | - Giovanna Garufi
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy.
| | - Rossana Di Rienzo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Claudia Martinelli
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Giuliana Pavone
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico - San Marco", Catania, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy.
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | | | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy; Breast Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | | | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| |
Collapse
|
12
|
Targeting Breast Cancer: An Overlook on Current Strategies. Int J Mol Sci 2023; 24:ijms24043643. [PMID: 36835056 PMCID: PMC9959993 DOI: 10.3390/ijms24043643] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Breast cancer (BC) is one of the most widely diagnosed cancers and a leading cause of cancer death among women worldwide. Globally, BC is the second most frequent cancer and first most frequent gynecological one, affecting women with a relatively low case-mortality rate. Surgery, radiotherapy, and chemotherapy are the main treatments for BC, even though the latter are often not aways successful because of the common side effects and the damage caused to healthy tissues and organs. Aggressive and metastatic BCs are difficult to treat, thus new studies are needed in order to find new therapies and strategies for managing these diseases. In this review, we intend to give an overview of studies in this field, presenting the data from the literature concerning the classification of BCs and the drugs used in therapy for the treatment of BCs, along with drugs in clinical studies.
Collapse
|
13
|
Liu Z, Zou H, Dang Q, Xu H, Liu L, Zhang Y, Lv J, Li H, Zhou Z, Han X. Biological and pharmacological roles of m 6A modifications in cancer drug resistance. Mol Cancer 2022; 21:220. [PMID: 36517820 PMCID: PMC9749187 DOI: 10.1186/s12943-022-01680-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/11/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer drug resistance represents the main obstacle in cancer treatment. Drug-resistant cancers exhibit complex molecular mechanisms to hit back therapy under pharmacological pressure. As a reversible epigenetic modification, N6-methyladenosine (m6A) RNA modification was regarded to be the most common epigenetic RNA modification. RNA methyltransferases (writers), demethylases (erasers), and m6A-binding proteins (readers) are frequently disordered in several tumors, thus regulating the expression of oncoproteins, enhancing tumorigenesis, cancer proliferation, development, and metastasis. The review elucidated the underlying role of m6A in therapy resistance. Alteration of the m6A modification affected drug efficacy by restructuring multidrug efflux transporters, drug-metabolizing enzymes, and anticancer drug targets. Furthermore, the variation resulted in resistance by regulating DNA damage repair, downstream adaptive response (apoptosis, autophagy, and oncogenic bypass signaling), cell stemness, tumor immune microenvironment, and exosomal non-coding RNA. It is highlighted that several small molecules targeting m6A regulators have shown significant potential for overcoming drug resistance in different cancer categories. Further inhibitors and activators of RNA m6A-modified proteins are expected to provide novel anticancer drugs, delivering the therapeutic potential for addressing the challenge of resistance in clinical resistance.
Collapse
Affiliation(s)
- Zaoqu Liu
- grid.412633.10000 0004 1799 0733Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China ,grid.207374.50000 0001 2189 3846Interventional Institute of Zhengzhou University, Zhengzhou, 450052 Henan China ,grid.412633.10000 0004 1799 0733Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, 450052 Henan China
| | - Haijiao Zou
- grid.412633.10000 0004 1799 0733Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Qin Dang
- grid.412633.10000 0004 1799 0733Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Hui Xu
- grid.412633.10000 0004 1799 0733Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Long Liu
- grid.412633.10000 0004 1799 0733Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yuyuan Zhang
- grid.412633.10000 0004 1799 0733Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Jinxiang Lv
- grid.412633.10000 0004 1799 0733Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Huanyun Li
- grid.412633.10000 0004 1799 0733Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Zhaokai Zhou
- grid.412633.10000 0004 1799 0733Department of Pediatric Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xinwei Han
- grid.412633.10000 0004 1799 0733Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China ,grid.207374.50000 0001 2189 3846Interventional Institute of Zhengzhou University, Zhengzhou, 450052 Henan China ,grid.412633.10000 0004 1799 0733Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, 450052 Henan China
| |
Collapse
|
14
|
Buono G, Arpino G, Del Mastro L, Fabi A, Generali D, Puglisi F, Zambelli A, Cinieri S, Nuzzo F, Di Lauro V, Vigneri P, Bianchini G, Montemurro F, Gennari A, De Laurentiis M. Extended adjuvant endocrine treatment for premenopausal women: A Delphi approach to guide clinical practice. Front Oncol 2022; 12:1032166. [PMID: 36387212 PMCID: PMC9645191 DOI: 10.3389/fonc.2022.1032166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/22/2022] [Indexed: 10/02/2023] Open
Abstract
The use of an aromatase inhibitor (AI) in combination with ovarian function suppression (OFS) has become the mainstay of adjuvant endocrine therapy in high-risk premenopausal patients with hormone receptor-positive breast cancer. Although five years of such therapy effectively reduces recurrence rates, a substantial risk of late recurrence remains in this setting. Multiple trials have shown that extending AI treatment beyond five years could offer further protection. However, as these studies comprised only postmenopausal patients, no direct evidence currently exists to inform about the potential benefits and/or side effects of extended AI + OFS therapies in premenopausal women. Given these grey areas, we conducted a Delphi survey to report on the opinion of experts in breast cancer treatment and summarize a consensus on the discussed topics. A total of 44 items were identified, all centred around two main themes: 1) defining reliable prognostic factors to pinpoint premenopausal patients eligible for endocrine therapy extension; 2) designing how such therapy should optimally be administered in terms of treatment combinations and duration based on patients' menopausal status. Each item was separately discussed and anonymously voted by 12 experts representing oncological institutes spread across Italy. The consensus threshold was reached in 36 out of 44 items (82%). Herein, we discuss the levels of agreement/disagreement achieved by each item in relation to the current body of literature. In the absence of randomized trials to guide the tailoring of extended AI treatment in premenopausal women, conclusions from our study provide a framework to assist routine clinical practice.
Collapse
Affiliation(s)
- Giuseppe Buono
- Department of Breast and Thoracic Oncology, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples “Federico II”, Naples, Italy
| | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
- Department of Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “San Martino” General Hospital, Genoa, Italy
| | - Alessandra Fabi
- Precision Medicine in Breast Cancer, Scientific Directorate, Department of Women Child and Public Health, Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Daniele Generali
- Breast Cancer Unit, Azienda Socio-Sanitaria Territoriale di Cremona, Cremona, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy
- Department of Medical Oncology, Centro di Riferimento Oncologico (CRO) Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Alberto Zambelli
- Medical Oncology, “Papa Giovanni XXIII” Hospital, Bergamo, Italy
| | - Saverio Cinieri
- Medical Oncology Division and Breast Unit, “Senatore Antonio Perrino” Hospital, Brindisi, Italy
| | - Francesco Nuzzo
- Department of Breast and Thoracic Oncology, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| | - Vincenzo Di Lauro
- Department of Breast and Thoracic Oncology, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “San Raffaele” Hospital, Milan, Italy
| | - Filippo Montemurro
- Breast Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia - Istituto di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Candiolo, Italy
| | - Alessandra Gennari
- Medical Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Michelino De Laurentiis
- Department of Breast and Thoracic Oncology, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| |
Collapse
|