1
|
Huang X, Hou S, Li Y, Xu G, Xia N, Duan Z, Luo K, Tian B. Targeting lipid metabolism via nanomedicine: A prospective strategy for cancer therapy. Biomaterials 2025; 317:123022. [PMID: 39754967 DOI: 10.1016/j.biomaterials.2024.123022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/20/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025]
Abstract
Lipid metabolism has been increasingly recognized to play an influencing role in tumor initiation, progression, metastasis, and therapeutic drug resistance. Targeting lipid metabolic reprogramming represents a promising therapeutic strategy. Despite their structural complexity and poor targeting efficacy, lipid-metabolizing drugs, either used alone or in combination with chemotherapeutic agents, have been employed in clinical practice. The advent of nanotechnology offers new approaches to enhancing therapeutic effects, includingthe targeted delivery and integration of lipid metabolic reprogramming with chemotherapy, photodynamic therapy (PDT), and immunotherapy. The integrated nanoformulation, nanomedicine, could significantly advance the field of lipid metabolism therapy. In this review, we will briefly introduce the concept of cancer lipid metabolism reprogramming, then elaborate the latest advances in engineered nanomedicine for targeting lipid metabolism during cancer treatment, and finally provide our insights into future perspectives of nanomedicine for interference with lipid metabolism in the tumor microenvironment.
Collapse
Affiliation(s)
- Xing Huang
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shengzhong Hou
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinggang Li
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Xu
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Ning Xia
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.
| | - Kui Luo
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.
| | - Bole Tian
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Huang X, Liu B, Shen S. Lipid Metabolism in Breast Cancer: From Basic Research to Clinical Application. Cancers (Basel) 2025; 17:650. [PMID: 40002245 PMCID: PMC11852908 DOI: 10.3390/cancers17040650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Breast cancer remains the most prevalent cancer among women globally, with significant links to obesity and lipid metabolism abnormalities. This review examines the role of lipid metabolism in breast cancer progression, highlighting its multifaceted contributions to tumor biology. We discuss key metabolic processes, including fatty acid metabolism, triglyceride metabolism, phospholipid metabolism, and cholesterol metabolism, detailing the reprogramming that occurs in these pathways within breast cancer cells. Alterations in lipid metabolism are emphasized for their roles in supporting energy production, membrane biogenesis, and tumor aggressiveness. Furthermore, we examine how lipid metabolism influences immune responses in the tumor microenvironment, affecting immune cell function and therapeutic efficacy. The potential of lipid metabolism as a target for novel therapeutic strategies is also addressed, with a focus on inhibitors of key metabolic enzymes. By integrating lipid metabolism with breast cancer research, this review underscores the importance of lipid metabolism in understanding breast cancer biology and developing treatment approaches aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Xiangyu Huang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China; (X.H.); (B.L.)
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bowen Liu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China; (X.H.); (B.L.)
| | - Songjie Shen
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China; (X.H.); (B.L.)
- Ambulatory Medical Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China
| |
Collapse
|
3
|
Wang X, Li S, Li Z, Lin Z, Wang Z. SRT3025-loaded cell membrane hybrid liposomes (3025@ML) enhanced anti-tumor activity of Oxaliplatin via inhibiting pyruvate kinase M2 and fatty acid synthase. Lipids Health Dis 2025; 24:14. [PMID: 39825408 PMCID: PMC11740399 DOI: 10.1186/s12944-025-02431-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/06/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Bladder cancer is one of the most common malignancies of the urinary system. Despite significant advances in diagnosis and treatment, the compromised therapeutic effect of chemotherapeutic agents, such as Oxaliplatin (OXA), remains a major clinical challenge. Thus, a combination therapy is required to enhance the OXA's therapeutic effectiveness and improve patient outcomes. METHODS The thin film hydration method was used to prepare the liposomes. Drug encapsulation efficiency and loading capacity were determined to investigate the advantages of the SRT3025-loaded cell membrane hybrid liposomes (3025@ML). Bladder cancer cell lines T24 and 5637 were cultured in McCoy's 5 A and RPMI 1640 medium, respectively. The Cell Counting Kit-8 assay was used to determine the cell viability by treating cells with a medium containing either the vehicle solution (control), the cell membrane hybrid liposomes (ML), 3025@ML, or compound 3 K. The antiproliferative activities were investigated after treating cells with OXA + 3025@ML and compound 3 K + OXA. Cell death and apoptosis were quantified by trypan blue and Annexin V-APC/PI apoptosis assay after treating cells with control, OXA, OXA + 3025@ML, and 3025@ML. Western blot analysis was performed after treating cells with 3025@ML, OXA, 3 K, 3025@ML + OXA, and 3 K + OXA to determine the protein levels of pyruvate kinase M2 (PKM2) and fatty acid synthase (FASN), etc. RESULTS: The present study demonstrated that 3025@ML enhances the chemotherapeutic effect of OXA. 3025@ML + OXA treated T24 and 5637 cells showed that combination therapy significantly reduced cell viability and increased cell death rate. Flow cytometry analysis showed that the combination of 3025@ML and OXA significantly increased the percentage of apoptotic cells in T24 cells. 3025@ML and compound 3 K reduced the levels of FASN in T24 and 5637 cells and increased the anti-tumor activity of OXA. Mechanistic studies showed that 3025@ML inhibited the PI3K/AKT/mTOR signaling pathway and reduced the expression of key metabolic regulators PKM2 and FASN. Furthermore, this study demonstrated that targeting lipid metabolism and inhibiting FASN can effectively overcome the compromised therapeutic effect of OXA. CONCLUSION The study demonstrated that 3025@ML significantly enhances the anti-tumor activity of OXA. This novel drug delivery system inhibits key metabolic pathways, which increase DNA damage and tumor cell apoptosis. The results indicate that 3025@ML is a promising therapeutic strategy for overcoming OXA's compromised therapeutic effect and potentially improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Xiaobin Wang
- Department of Urology, Southern University of Science and Technology Hospital, Shenzhen, 518052, China.
| | - Shulin Li
- Department of Urology, Southern University of Science and Technology Hospital, Shenzhen, 518052, China
| | - Zichen Li
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China
| | - Zhuona Lin
- Medical School of Basic Medical Sciences, Shenzhen University, Shenzhen, 518060, China
| | - Zhifeng Wang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450003, China.
| |
Collapse
|
4
|
Li D, Jin P, Cai Y, Wu S, Guo X, Zhang Z, Liu K, Li P, Hu Y, Zhou Y. Clinical significance of lipid pathway-targeted therapy in breast cancer. Front Pharmacol 2025; 15:1514811. [PMID: 39834807 PMCID: PMC11743736 DOI: 10.3389/fphar.2024.1514811] [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: 10/21/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Globally, breast cancer represents the most common cancer and the primary cause of death by cancer in women. Lipids are crucial in human physiology, serving as vital energy reserves, structural elements of biological membranes, and essential signaling molecules. The metabolic reprogramming of lipid pathways has emerged as a critical factor in breast cancer progression, drug resistance, and patient prognosis. In this study, we delve into the clinical implications of lipid pathway-targeted therapy in breast cancer. We highlight key enzymes and potential therapeutic targets involved in lipid metabolism reprogramming, and their associations with cancer progression and treatment outcomes. Furthermore, we detail the clinical trials exploring the anticancer and cancer chemopreventive activity of therapies targeting these molecules. However, the clinical efficacy of these therapies remains controversial, highlighting the urgent need for predictive biomarkers to identify patient subpopulations likely to benefit from such treatment. We propose the Selective Lipid Metabolism Therapy Benefit Hypothesis, emphasizing the importance of personalized medicine in optimizing lipid pathway-targeted therapy for breast cancer patients.
Collapse
Affiliation(s)
- Dan Li
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengcheng Jin
- Department of Surgical Oncology, Linhai Branch, The Second Affiliated Hospital, Zhejiang University School of Medicine, Taizhou, Zhejiang, China
| | - Yiqi Cai
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shijie Wu
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianan Guo
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyun Zhang
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kexin Liu
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Panni Li
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Hu
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yunxiang Zhou
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Gu Q, Wang Y, Yi P, Cheng C. Theoretical framework and emerging challenges of lipid metabolism in cancer. Semin Cancer Biol 2025; 108:48-70. [PMID: 39674303 DOI: 10.1016/j.semcancer.2024.12.002] [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: 10/11/2024] [Revised: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Elevated lipid metabolism is one of hallmarks of malignant tumors. Lipids not only serve as essential structural components of biological membranes but also provide energy and substrates for the proliferation of cancer cells and tumor growth. Cancer cells meet their lipid needs by coordinating the processes of lipid absorption, synthesis, transport, storage, and catabolism. As research in this area continues to deepen, numerous new discoveries have emerged, making it crucial for scientists to stay informed about the developments of cancer lipid metabolism. In this review, we first discuss relevant concepts and theories or assumptions that help us understand the lipid metabolism and -based cancer therapies. We then systematically summarize the latest advancements in lipid metabolism including new mechanisms, novel targets, and up-to-date pre-clinical and clinical investigations of anti-cancer treatment with lipid metabolism targeted drugs. Finally, we emphasize emerging research directions and therapeutic strategies, and discuss future prospective and emerging challenges. This review aims to provide the latest insights and guidance for research in the field of cancer lipid metabolism.
Collapse
Affiliation(s)
- Qiuying Gu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Chunming Cheng
- Department of Oncology Science, OU Health Stephenson Cancer Center at University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
6
|
Ye J, Wu S, Quan Q, Ye F, Zhang J, Song C, Fan Y, Cao H, Tang H, Zhao J. Fibroblast Growth Factor Receptor 4 Promotes Triple-Negative Breast Cancer Progression via Regulating Fatty Acid Metabolism Through the AKT/RYR2 Signaling. Cancer Med 2024; 13:e70439. [PMID: 39658878 PMCID: PMC11631837 DOI: 10.1002/cam4.70439] [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: 08/06/2024] [Revised: 10/16/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. Previous studies have found that fibroblast growth factor receptor 4 (FGFR4) plays a crucial role in tumor development and metastasis. However, the potential role and underlying mechanisms of FGFR4 in the progression of TNBC remain unclear. METHODS Statistical analysis of FGFR4 expression data in public databases was used to reveal its role in TNBC. qRT-PCR was used to detect FGFR4 expression levels. The impact of FGFR4 level changes on TNBC cell proliferation was assessed using CCK-8 and colony formation assays, while Transwell invasion assays and JC-1 staining were employed to analyze the effects of FGFR4 level changes on the invasiveness and survival capability of TNBC cells. Differentially expressed genes were subjected to Gene Ontology, KEGG, and GSEA enrichment analyses to identify associated signaling pathways. Additionally, Oil Red O staining, fatty acid metabolite detection, and Western blot analysis were used to investigate the impact of FGFR4 and its inhibitor fisogatinib, as well as the AKT activator SC79, on the metabolic reprogramming of fatty acids in TNBC cells. RESULTS FGFR4 was found to be upregulated in breast cancer and correlated with poorer patient outcomes. Inhibition of FGFR4 resulted in reduced cell growth and invasion in TNBC cells. It also led to increased lipid accumulation, upregulated lipid biosynthesis-related genes, and downregulated lipolysis-related genes. Mechanistically, FGFR4 inhibition suppressed the activation of the AKT/RYR2 signaling pathway. Reactivating the AKT pathway reversed the suppressive effects of FGFR4 inhibition on TNBC progression. CONCLUSION Dysregulated FGFR4 activates the AKT/RYR2 axis, leading to tumor proliferation, invasion, and altered lipid metabolism in TNBC. FGFR4 inhibition could potentially serve as a novel therapeutic strategy for TNBC treatment.
Collapse
Affiliation(s)
- Jinhui Ye
- Research Center of Cancer Diagnosis and Therapy, Department of OncologyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- Department of Breast OncologyThe First People's Hospital of ZhaoqingZhaoqingChina
| | - Song Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Qiang Quan
- Research Center of Cancer Diagnosis and Therapy, Department of OncologyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Feng Ye
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Jinhui Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Cailu Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Yidan Fan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Huijiao Cao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Jianfu Zhao
- Research Center of Cancer Diagnosis and Therapy, Department of OncologyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| |
Collapse
|
7
|
Fan S, Guo J, Nie H, Xiong H, Xia Y. Aberrant Energy Metabolism in Tumors and Potential Therapeutic Targets. Genes Chromosomes Cancer 2024; 63:e70008. [PMID: 39584783 PMCID: PMC11587691 DOI: 10.1002/gcc.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Energy metabolic reprogramming is frequently observed during tumor progression as tumor cells necessitate adequate energy production for rapid proliferation. Although current medical research shows promising prospects in studying the characteristics of tumor energy metabolism and developing anti-tumor drugs targeting energy metabolism, there is a lack of systematic compendiums and comprehensive reviews in this field. The objective of this study is to conduct a systematic review on the characteristics of tumor cells' energy metabolism, with a specific focus on comparing abnormalities between tumor and normal cells, as well as summarizing potential targets for tumor therapy. Additionally, this review also elucidates the aberrant mechanisms underlying four major energy metabolic pathways (glucose, lipid, glutamine, and mitochondria-dependent) during carcinogenesis and tumor progression. Through the utilization of graphical representations, we have identified anomalies in crucial energy metabolism pathways, encompassing transporter proteins (glucose transporter, CD36, and ASCT2), signaling molecules (Ras, AMPK, and PTEN), as well as transcription factors (Myc, HIF-1α, CREB-1, and p53). The key molecules responsible for aberrant energy metabolism in tumors may serve as potential targets for cancer therapy. Therefore, this review provides an overview of the distinct energy-generating pathways within tumor cells, laying the groundwork for developing innovative strategies for precise cancer treatment.
Collapse
Affiliation(s)
- Shuhao Fan
- Shandong First Medical UniversityJinanShandongPeople's Republic of China
- College of Medical EngineeringJining Medical UniversityJiningShandongPeople's Republic of China
| | - Jianhua Guo
- College of Medical EngineeringJining Medical UniversityJiningShandongPeople's Republic of China
| | - Hui Nie
- Shandong First Medical UniversityJinanShandongPeople's Republic of China
- College of Medical EngineeringJining Medical UniversityJiningShandongPeople's Republic of China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJiningShandongPeople's Republic of China
| | - Yong Xia
- Shandong First Medical UniversityJinanShandongPeople's Republic of China
- College of Medical EngineeringJining Medical UniversityJiningShandongPeople's Republic of China
| |
Collapse
|
8
|
Chiu CF, Guerrero JJG, Regalado RRH, Zhou J, Notarte KI, Lu YW, Encarnacion PC, Carles CDD, Octavo EM, Limbaroc DCI, Saengboonmee C, Huang SY. Insights into Metabolic Reprogramming in Tumor Evolution and Therapy. Cancers (Basel) 2024; 16:3513. [PMID: 39456607 PMCID: PMC11506062 DOI: 10.3390/cancers16203513] [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: 09/21/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Cancer remains a global health challenge, characterized not just by uncontrolled cell proliferation but also by the complex metabolic reprogramming that underlies its development and progression. Objectives: This review delves into the intricate relationship between cancer and its metabolic alterations, drawing an innovative comparison with the cosmological concepts of dark matter and dark energy to highlight the pivotal yet often overlooked role of metabolic reprogramming in tumor evolution. Methods: It scrutinizes the Warburg effect and other metabolic adaptations, such as shifts in lipid synthesis, amino acid turnover, and mitochondrial function, driven by mutations in key regulatory genes. Results: This review emphasizes the significance of targeting these metabolic pathways for therapeutic intervention, outlining the potential to disrupt cancer's energy supply and signaling mechanisms. It calls for an interdisciplinary research approach to fully understand and exploit the intricacies of cancer metabolism, pointing toward metabolic reprogramming as a promising frontier for developing more effective cancer treatments. Conclusion: By equating cancer's metabolic complexity with the enigmatic nature of dark matter and energy, this review underscores the critical need for innovative strategies in oncology, highlighting the importance of unveiling and targeting the "dark energy" within cancer cells to revolutionize future therapy and research.
Collapse
Affiliation(s)
- Ching-Feng Chiu
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan; (J.J.G.G.); (Y.-W.L.); (P.C.E.)
- Taipei Medical University Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Jonathan Jaime G. Guerrero
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan; (J.J.G.G.); (Y.-W.L.); (P.C.E.)
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
- College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
| | - Ric Ryan H. Regalado
- National Institute of Molecular Biology and Biotechnology, College of Science, University of the Philippines Diliman, Quezon City 1101, Philippines;
| | - Jiayan Zhou
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Kin Israel Notarte
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
| | - Yu-Wei Lu
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan; (J.J.G.G.); (Y.-W.L.); (P.C.E.)
| | - Paolo C. Encarnacion
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan; (J.J.G.G.); (Y.-W.L.); (P.C.E.)
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
- College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
- Department of Industrial Engineering and Management, Yuan Ze University, 135 Yuan-Tung Road, Chung-Li 32003, Taiwan
| | - Cidne Danielle D. Carles
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
- College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
| | - Edrian M. Octavo
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
| | - Dan Christopher I. Limbaroc
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
- College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
| | - Charupong Saengboonmee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan
| |
Collapse
|
9
|
Rosell R, Jantus-Lewintre E, Cao P, Cai X, Xing B, Ito M, Gomez-Vazquez JL, Marco-Jordán M, Calabuig-Fariñas S, Cardona AF, Codony-Servat J, Gonzalez J, València-Clua K, Aguilar A, Pedraz-Valdunciel C, Dantes Z, Jain A, Chandan S, Molina-Vila MA, Arrieta O, Ferrero M, Camps C, González-Cao M. KRAS-mutant non-small cell lung cancer (NSCLC) therapy based on tepotinib and omeprazole combination. Cell Commun Signal 2024; 22:324. [PMID: 38867255 PMCID: PMC11167791 DOI: 10.1186/s12964-024-01667-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/17/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND KRAS-mutant non-small cell lung cancer (NSCLC) shows a relatively low response rate to chemotherapy, immunotherapy and KRAS-G12C selective inhibitors, leading to short median progression-free survival, and overall survival. The MET receptor tyrosine kinase (c-MET), the cognate receptor of hepatocyte growth factor (HGF), was reported to be overexpressed in KRAS-mutant lung cancer cells leading to tumor-growth in anchorage-independent conditions. METHODS Cell viability assay and synergy analysis were carried out in native, sotorasib and trametinib-resistant KRAS-mutant NSCLC cell lines. Colony formation assays and Western blot analysis were also performed. RNA isolation from tumors of KRAS-mutant NSCLC patients was performed and KRAS and MET mRNA expression was determined by real-time RT-qPCR. In vivo studies were conducted in NSCLC (NCI-H358) cell-derived tumor xenograft model. RESULTS Our research has shown promising activity of omeprazole, a V-ATPase-driven proton pump inhibitor with potential anti-cancer properties, in combination with the MET inhibitor tepotinib in KRAS-mutant G12C and non-G12C NSCLC cell lines, as well as in G12C inhibitor (AMG510, sotorasib) and MEK inhibitor (trametinib)-resistant cell lines. Moreover, in a xenograft mouse model, combination of omeprazole plus tepotinib caused tumor growth regression. We observed that the combination of these two drugs downregulates phosphorylation of the glycolytic enzyme enolase 1 (ENO1) and the low-density lipoprotein receptor-related protein (LRP) 5/6 in the H358 KRAS G12C cell line, but not in the H358 sotorasib resistant, indicating that the effect of the combination could be independent of ENO1. In addition, we examined the probability of recurrence-free survival and overall survival in 40 early lung adenocarcinoma patients with KRAS G12C mutation stratified by KRAS and MET mRNA levels. Significant differences were observed in recurrence-free survival according to high levels of KRAS mRNA expression. Hazard ratio (HR) of recurrence-free survival was 7.291 (p = 0.014) for high levels of KRAS mRNA expression and 3.742 (p = 0.052) for high MET mRNA expression. CONCLUSIONS We posit that the combination of the V-ATPase inhibitor omeprazole plus tepotinib warrants further assessment in KRAS-mutant G12C and non G12C cell lines, including those resistant to the covalent KRAS G12C inhibitors.
Collapse
Affiliation(s)
- Rafael Rosell
- Germans Trias i Pujol Research Institute, Badalona (IGTP), Barcelona, Spain.
- IOR, Hospital Quiron-Dexeus Barcelona, Barcelona, Spain.
- Laboratory of Molecular Biology, Germans Trias i Pujol Health Sciences Institute and Hospital (IGTP), Camí de les Escoles, s/n, 08916, Badalona, Barcelona, Spain.
| | - Eloisa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain.
- Trial Mixed Unit, Centro Investigación Príncipe Felipe-Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain.
- Department of Biotechnology, Universitat Politècnica de València, Camí de Vera s/n, Valencia, 46022, Spain.
- Joint Unit: Nanomedicine, Centro Investigación Príncipe Felipe-Universitat Politècnica de Valencia, Valencia, Spain.
| | - Peng Cao
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China.
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou Peoples Hospital, Quzhou, China.
- Shandong Academy of Chinese Medicine, Jinan, China.
| | - Xueting Cai
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Baojuan Xing
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Masaoki Ito
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Jose Luis Gomez-Vazquez
- Germans Trias i Pujol Research Institute, Badalona (IGTP), Barcelona, Spain
- Hospital Universitari de Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain
- Trial Mixed Unit, Centro Investigación Príncipe Felipe-Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Department of Pathology, Universitat de Valéncia, Valencia, Spain
| | - Andrés Felipe Cardona
- Institute of Research and Education, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center - CTIC, Bogotá, Colombia
| | - Jordi Codony-Servat
- Germans Trias i Pujol Research Institute, Badalona (IGTP), Barcelona, Spain
- Pangaea Oncology, Hospital Quiron-Dexeus Barcelona, Barcelona, Spain
| | - Jessica Gonzalez
- Germans Trias i Pujol Research Institute, Badalona (IGTP), Barcelona, Spain
| | | | | | | | | | - Anisha Jain
- Department of Microbiology, JSS Academy of Higher Education & Research, Mysuru, India
| | - S Chandan
- Department of Microbiology, JSS Academy of Higher Education & Research, Mysuru, India
| | | | - Oscar Arrieta
- National Institute of Cancerology (INCAN), Mexico City, Mexico
| | - Macarena Ferrero
- Trial Mixed Unit, Centro Investigación Príncipe Felipe-Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Carlos Camps
- Trial Mixed Unit, Centro Investigación Príncipe Felipe-Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Medical Oncology Department, General University Hospital of Valencia, Valencia, Spain
| | | |
Collapse
|
10
|
Valenzuela R, Walbaum B, Farias C, Acevedo F, Vargas C, Bennett JT, Bravo ML, Pinto MP, Medina L, Merino T, Ibañez C, Parada A, Sanchez C. High linoleic acid levels in red blood cells predict a poor response to neoadjuvant chemotherapy in human epidermal growth factor receptor type 2-positive breast cancer patients. Nutrition 2024; 121:112357. [PMID: 38430738 DOI: 10.1016/j.nut.2024.112357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Polyunsaturated fatty acids are categorized as ω-3 or ⍵-6. Previous studies demonstrate that breast cancers display a high expression of fatty acid synthase and high fatty acid levels. Our study sought to determine if changes in plasma or red blood cell membrane fatty acid levels were associated with the response to preoperative (neoadjuvant) chemotherapy in non-metastatic breast cancer patients. METHODS Our prospective study assessed fatty acid levels in plasma and red blood cell membrane. Response to neoadjuvant chemotherapy was evaluated by the presence or absence of pathologic complete response and/or residual cancer burden. RESULTS A total of 28 patients were included. First, patients who achieved pathologic complete response had significantly higher neutrophil-to-lymphocyte ratio versus no pathologic complete response (P = 0.003). Second, total red blood cell membrane polyunsaturated fatty acids were higher in the absence of pathologic complete response (P = 0.0028). Third, total red blood cell membrane ⍵-6 polyunsaturated fatty acids were also higher in no pathologic complete response (P < 0.01). Among ⍵-6 polyunsaturated fatty acids, red blood cell membrane linoleic acid was higher in the absence of pathologic complete response (P < 0.01). Notably, plasma polyunsaturated fatty acid, ⍵-6, and linoleic acid levels did not have significant differences. A multivariate analysis confirmed red blood cell membrane linoleic acid was associated with no pathologic complete response; this was further confirmed by receiver operating characteristic analysis (specificity = 92.3%, sensitivity = 76.9%, and area under the curve = 0.855). CONCLUSIONS Pending further validation, red blood cell membrane linoleic acid might serve as a predictor biomarker of poorer response to neoadjuvant chemotherapy in non-metastatic human epidermal growth factor receptor type 2-positive breast cancer. Measuring fatty acids in red blood cell membrane could offer a convenient, minimally invasive strategy to identifying patients more likely to respond or those with chemoresistance.
Collapse
Affiliation(s)
- Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Benjamín Walbaum
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Farias
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Francisco Acevedo
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Vargas
- Department of Surgical Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José Tomas Bennett
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M Loreto Bravo
- Support Team for Oncological Research and Medicine (STORM), Santiago, Chile
| | - Mauricio P Pinto
- Support Team for Oncological Research and Medicine (STORM), Santiago, Chile
| | - Lidia Medina
- Centro del Cáncer Nuestra Señora de la Esperanza, UC CHRISTUS Healthcare Network, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tomas Merino
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Ibañez
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Parada
- Department of Health Sciences. School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cesar Sanchez
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
11
|
Barlow L, Josephraj S, Gu B, Dong Z, Zhang JT. FASN negatively regulates p65 expression by reducing its stability via Thr 254 phosphorylation and isomerization by Pin1. J Lipid Res 2024; 65:100529. [PMID: 38467328 PMCID: PMC11017288 DOI: 10.1016/j.jlr.2024.100529] [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: 07/03/2023] [Revised: 02/18/2024] [Accepted: 02/20/2024] [Indexed: 03/13/2024] Open
Abstract
FASN, the sole cytosolic enzyme responsible for de novo palmitate synthesis in mammalian cells, has been associated with poor prognosis in cancer and shown to cause drug and radiation resistance by upregulating DNA damage repair via suppression of p65 expression. Targeting FASN by repurposing proton pump inhibitors has generated impressive outcomes in triple-negative breast cancer patients. While p65 regulation of DNA damage repair was thought to be due to its suppression of poly(ADP-ribose) polymerase 1 gene transcription, the mechanism of FASN regulation of p65 expression was unknown. In this study, we show that FASN regulates p65 stability by controlling its phosphorylation at Thr254, which recruits the peptidyl-prolyl cis/trans isomerase Pin1 that is known to stabilize many proteins in the nucleus. This regulation is mediated by palmitate, the FASN catalytic product, not by FASN protein per se. This finding of FASN regulation of p65 stability via phosphorylation of Thr254 and isomerization by Pin1 implicates that FASN and its catalytic product palmitate may play an important role in regulating protein stability in general and p65 more specifically.
Collapse
Affiliation(s)
- Lincoln Barlow
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sophia Josephraj
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Boqing Gu
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Zizheng Dong
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Jian-Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| |
Collapse
|
12
|
Cui Y, Man S, Tao J, Liu Y, Ma L, Guo L, Huang L, Liu C, Gao W. The lipid droplet in cancer: From being a tumor-supporting hallmark to clinical therapy. Acta Physiol (Oxf) 2024; 240:e14087. [PMID: 38247395 DOI: 10.1111/apha.14087] [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: 08/27/2023] [Revised: 10/18/2023] [Accepted: 01/01/2024] [Indexed: 01/23/2024]
Abstract
INTRODUCTION Abnormal lipid metabolism, one of the hallmarks in cancer, has gradually emerged as a novel target for cancer treatment. As organelles that store and release excess lipids, lipid droplets (LDs) resemble "gears" and facilitate cancer development in the body. AIM This review discusses the life cycle of LDs, the relationship between abnormal LDs and cancer hallmarks, and the application of LDs in theragnostic and clinical contexts to provide a contemporary understanding of the role of LDs in cancer. METHODS A systematic literature search was conducted in PubMed and SPORTDiscus. Retrieve and summarize clinical trials of drugs that target proteins associated with LD formation using the Clinical Trials website. Create a schematic diagram of lipid droplets in the tumor microenvironment using Adobe Illustrator. CONCLUSION As one of the top ten hallmarks of cancer, abnormal lipid metabolism caused by excessive generation of LDs interrelates with other hallmarks. The crosstalk between excessive LDs and intracellular free fatty acids (FFAs) promotes an inflammatory environment that supports tumor growth. Moreover, LDs contribute to cancer metastasis and cell death resistance in vivo. Statins, as HMGCR inhibitors, are promising to be the pioneering commercially available anti-cancer drugs that target LD formation.
Collapse
Affiliation(s)
- Yingfang Cui
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Jiejing Tao
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yu Liu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Changxiao Liu
- State Key Laboratory of Drug Release Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research Co and Ltd., Tianjin, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
13
|
Liu Z, Yu B, Su M, Yuan C, Liu C, Wang X, Song X, Li C, Wang F, Ma J, Wu M, Chen D, Yu J, Yu Z. Construction of a risk stratification model integrating ctDNA to predict response and survival in neoadjuvant-treated breast cancer. BMC Med 2023; 21:493. [PMID: 38087296 PMCID: PMC10717175 DOI: 10.1186/s12916-023-03163-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The pathological complete response (pCR) to neoadjuvant chemotherapy (NAC) of breast cancer is closely related to a better prognosis. However, there are no reliable indicators to accurately identify which patients will achieve pCR before surgery, and a model for predicting pCR to NAC is required. METHODS A total of 269 breast cancer patients in Shandong Cancer Hospital and Liaocheng People's Hospital receiving anthracycline and taxane-based NAC were prospectively enrolled. Expression profiling using a 457 cancer-related gene sequencing panel (DNA sequencing) covering genes recurrently mutated in breast cancer was carried out on 243 formalin-fixed paraffin-embedded tumor biopsies samples before NAC from 243 patients. The unique personalized panel of nine individual somatic mutation genes from the constructed model was used to detect and analyze ctDNA on 216 blood samples. Blood samples were collected at indicated time points including before chemotherapy initiation, after the 1st NAC and before the 2nd NAC cycle, during intermediate evaluation, and prior to surgery. In this study, we characterized the value of gene profile mutation and circulating tumor DNA (ctDNA) in combination with clinical characteristics in the prediction of pCR before surgery and investigated the prognostic prediction. The median follow-up time for survival analysis was 898 days. RESULTS Firstly, we constructed a predictive NAC response model including five single nucleotide variant (SNV) mutations (TP53, SETBP1, PIK3CA, NOTCH4 and MSH2) and four copy number variation (CNV) mutations (FOXP1-gain, EGFR-gain, IL7R-gain, and NFKB1A-gain) in the breast tumor, combined with three clinical factors (luminal A, Her2 and Ki67 status). The tumor prediction model showed good discrimination of chemotherapy sensitivity for pCR and non-pCR with an AUC of 0.871 (95% CI, 0.797-0.927) in the training set, 0.771 (95% CI, 0.649-0.883) in the test set, and 0.726 (95% CI, 0.556-0.865) in an extra test set. This tumor prediction model can also effectively predict the prognosis of disease-free survival (DFS) with an AUC of 0.749 at 1 year and 0.830 at 3 years. We further screened the genes from the tumor prediction model to establish a unique personalized panel consisting of 9 individual somatic mutation genes to detect and analyze ctDNA. It was found that ctDNA positivity decreased with the passage of time during NAC, and ctDNA status can predict NAC response and metastasis recurrence. Finally, we constructed the chemotherapy prediction model combined with the tumor prediction model and pretreatment ctDNA levels, which has a better prediction effect of pCR with the AUC value of 0.961. CONCLUSIONS In this study, we established a chemotherapy predictive model with a non-invasive tool that is built based on genomic features, ctDNA status, as well as clinical characteristics for predicting pCR to recognize the responders and non-responders to NAC, and also predicting prognosis for DFS in breast cancer. Adding pretreatment ctDNA levels to a model containing gene profile mutation and clinical characteristics significantly improves stratification over the clinical variables alone.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
- Shandong University Cancer Center, Jinan, 250117, Shandong, China
| | - Bo Yu
- Berry Oncology Institutes, Beijing, China
| | - Mu Su
- Berry Oncology Institutes, Beijing, China
| | - Chenxi Yuan
- Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| | - Cuicui Liu
- Thyroid & Breast Surgery Department, LiaoCheng Peoples's Hospital, Liaocheng, 252000, China
| | - Xinzhao Wang
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Xiang Song
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Chao Li
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Fukai Wang
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Jianli Ma
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Meng Wu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
- Shandong University Cancer Center, Jinan, 250117, Shandong, China
| | - Dawei Chen
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
- Shandong University Cancer Center, Jinan, 250117, Shandong, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
- Shandong University Cancer Center, Jinan, 250117, Shandong, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, 250117, China.
| | - Zhiyong Yu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| |
Collapse
|
14
|
Pacheco JHL, Elizondo G. Interplay between Estrogen, Kynurenine, and AHR Pathways: An immunosuppressive axis with therapeutic potential for breast cancer treatment. Biochem Pharmacol 2023; 217:115804. [PMID: 37716620 DOI: 10.1016/j.bcp.2023.115804] [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: 06/09/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Breast cancer is one of the most common malignancies among women worldwide. Estrogen exposure via endogenous and exogenous sources during a lifetime, together with environmental exposure to estrogenic compounds, represent the most significant risk factor for breast cancer development. As breast tumors establish, multiple pathways are deregulated. Among them is the aryl hydrocarbon receptor (AHR) signaling pathway. AHR, a ligand-activated transcription factor associated with the metabolism of polycyclic aromatic hydrocarbons and estrogens, is overexpressed in breast cancer. Furthermore, AHR and estrogen receptor (ER) cross-talk pathways have been observed. Additionally, the Tryptophan (Trp) catabolizing enzymes indolamine-2,3-dioxygenase (IDO) and tryptophan-2,3-dioxygenase (TDO) are overexpressed in breast cancer. IDO/TDO catalyzes the formation of Kynurenine (KYN) and other tryptophan-derived metabolites, which are ligands of AHR. Once KYN activates AHR, it stimulates the expression of the IDO enzyme, increases the level of KYN, and activates non-canonical pathways to control inflammation and immunosuppression in breast tumors. The interplay between E2, AHR, and IDO/TDO/KYN pathways and their impact on the immune system represents an immunosuppressive axis on breast cancer. The potential modulation of the immunosuppressive E2-AHR-IDO/TDO/KYN axis has aroused great expectations in oncotherapy. The present article will review the mechanisms implicated in generating the immunosuppressive axis E2-AHR-IDO/TDO/KYN in breast cancer and the current state of knowledge as a potential therapeutic target.
Collapse
Affiliation(s)
| | - Guillermo Elizondo
- Departamento de Biología Celular, CINVESTAV-IPN, Av. IPN 2508, C.P. 07360 Ciudad de México, México.
| |
Collapse
|
15
|
Gong Z, Zhu J, Chen J, Feng F, Zhang H, Zhang Z, Song C, Liang K, Yang S, Fan S, Fang X, Shen S. CircRREB1 mediates lipid metabolism related senescent phenotypes in chondrocytes through FASN post-translational modifications. Nat Commun 2023; 14:5242. [PMID: 37640697 PMCID: PMC10462713 DOI: 10.1038/s41467-023-40975-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
Osteoarthritis is a prevalent age-related disease characterized by dysregulation of extracellular matrix metabolism, lipid metabolism, and upregulation of senescence-associated secretory phenotypes. Herein, we clarify that CircRREB1 is highly expressed in secondary generation chondrocytes and its deficiency can alleviate FASN related senescent phenotypes and osteoarthritis progression. CircRREB1 impedes proteasome-mediated degradation of FASN by inhibiting acetylation-mediated ubiquitination. Meanwhile, CircRREB1 induces RanBP2-mediated SUMOylation of FASN and enhances its protein stability. CircRREB1-FASN axis inhibits FGF18 and FGFR3 mediated PI3K-AKT signal transduction, then increased p21 expression. Intra-articular injection of adenovirus-CircRreb1 reverses the protective effects in CircRreb1 deficiency mice. Further therapeutic interventions could have beneficial effects in identifying CircRREB1 as a potential prognostic and therapeutic target for age-related OA.
Collapse
Affiliation(s)
- Zhe Gong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Jinjin Zhu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Junxin Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Fan Feng
- Obstetrics and Gynecology Hospital, Kunpeng Road, Hangzhou, 310016, Zhejiang Province, China
| | - Haitao Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Zheyuan Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Chenxin Song
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Kaiyu Liang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Shuhui Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
| | - Xiangqian Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
| | - Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
| |
Collapse
|
16
|
Chaturvedi S, Biswas M, Sadhukhan S, Sonawane A. Role of EGFR and FASN in breast cancer progression. J Cell Commun Signal 2023:10.1007/s12079-023-00771-w. [PMID: 37490191 DOI: 10.1007/s12079-023-00771-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/22/2023] [Indexed: 07/26/2023] Open
Abstract
Breast cancer (BC) emerged as one of the life-threatening diseases among females. Despite notable improvements made in cancer detection and treatment worldwide, according to GLOBACAN 2020, BC is the fifth leading cancer, with an estimated 1 in 6 cancer deaths, in a majority of countries. However, the exact cause that leads to BC progression still needs to be determined. Here, we reviewed the role of two novel biomarkers responsible for 50-70% of BC progression. The first one is epidermal growth factor receptor (EGFR) which belongs to the ErbB tyrosine kinases family, signalling pathways associated with it play a significant role in regulating cell proliferation and division. Another one is fatty acid synthase (FASN), a key enzyme responsible for the de novo lipid synthesis required for cancer cell development. This review presents a rationale for the EGFR-mediated pathways, their interaction with FASN, communion of these two biomarkers with BC, and improvements to overcome drug resistance caused by them.
Collapse
Affiliation(s)
- Suchi Chaturvedi
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Madhya Pradesh, 453552, India
| | - Mainak Biswas
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, 751024, India
| | - Sushabhan Sadhukhan
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678623, India.
- Physical & Chemical Biology Laboratory and Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678623, India.
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Madhya Pradesh, 453552, India.
| |
Collapse
|
17
|
Huang X, Tan W, Liu Z, Fu X, Li Z, Lai S, Li Q, Zhong X, Qu F, Zhang H, Li H. EIF4A3-induced circZFAND6 promotes breast cancer proliferation and metastasis through the miR-647/FASN axis. Life Sci 2023; 324:121745. [PMID: 37127184 DOI: 10.1016/j.lfs.2023.121745] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
AIMS Circular RNAs (circRNAs) are important regulators in breast cancer progression. However, the underlying mechanism of circRNAs functions in breast cancer remain largely unclear. MAIN METHODS To investigate the circRNAs expression pattern in breast cancer, high-throughput circRNA microarray assay was used. The top up-regulated circRNA, circZFAND6, was submitted to further experiments, including cell counting kit-8 (CCK-8) assay, colony formation assay, transwell assay and mouse xenograft assay. To investigate the underlying mechanism of circZFAND6 function in breast cancer progression, luciferase reporter assay and RNA immunoprecipitation (RIP) assay were conducted. KEY FINDINGS We found a novel circRNA, circZFAND6, was up-regulated in breast cancer tissues and cell lines. Inhibition of circZFAND6 reduced proliferation and metastasis of breast cancer. Mechanically, circZFAND6 acted as a competing endogenous RNA (ceRNA) to sponge miR-647 and increase fatty acid synthase (FASN) expression. And eukaryotic translation initiation factor 4A3 (EIF4A3) was found to bind to circZFAND6 pre-mRNA transcript upstream region, leading to the high expression of circZFAND6 in breast cancer. Inhibition of EIF4A3 also suppressed proliferation and metastasis of breast cancer. SIGNIFICANCE EIF4A3-induced circZFAND6 up-regulation promoted proliferation and metastasis of breast cancer through the miR-647/FASN axis. Our results uncovered a possible mechanism underlying breast cancer progression and might provide a breast cancer treatment target.
Collapse
Affiliation(s)
- Xiaojia Huang
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Department of Breast Oncology Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, China
| | - Weige Tan
- The First Affiliated Hospital of Guangzhou Medical University, The First Clinical School of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Ziteng Liu
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Xiaoyan Fu
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Zongyan Li
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Shengqing Lai
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Qian Li
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Xiaofang Zhong
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Fanli Qu
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Huayao Zhang
- Department of Breast and Thyroid Surgery, SSL Central Hospital of Dongguan City, Dongguan, Guangdong 523325, China
| | - Haiyan Li
- Department of Breast Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, China.
| |
Collapse
|
18
|
Wedam R, Greer YE, Wisniewski DJ, Weltz S, Kundu M, Voeller D, Lipkowitz S. Targeting Mitochondria with ClpP Agonists as a Novel Therapeutic Opportunity in Breast Cancer. Cancers (Basel) 2023; 15:cancers15071936. [PMID: 37046596 PMCID: PMC10093243 DOI: 10.3390/cancers15071936] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Breast cancer is the most frequently diagnosed malignancy worldwide and the leading cause of cancer mortality in women. Despite the recent development of new therapeutics including targeted therapies and immunotherapy, triple-negative breast cancer remains an aggressive form of breast cancer, and thus improved treatments are needed. In recent decades, it has become increasingly clear that breast cancers harbor metabolic plasticity that is controlled by mitochondria. A myriad of studies provide evidence that mitochondria are essential to breast cancer progression. Mitochondria in breast cancers are widely reprogrammed to enhance energy production and biosynthesis of macromolecules required for tumor growth. In this review, we will discuss the current understanding of mitochondrial roles in breast cancers and elucidate why mitochondria are a rational therapeutic target. We will then outline the status of the use of mitochondria-targeting drugs in breast cancers, and highlight ClpP agonists as emerging mitochondria-targeting drugs with a unique mechanism of action. We also illustrate possible drug combination strategies and challenges in the future breast cancer clinic.
Collapse
Affiliation(s)
- Rohan Wedam
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yoshimi Endo Greer
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David J Wisniewski
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sarah Weltz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Manjari Kundu
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Donna Voeller
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
19
|
Scheinberg T, Mak B, Butler L, Selth L, Horvath LG. Targeting lipid metabolism in metastatic prostate cancer. Ther Adv Med Oncol 2023; 15:17588359231152839. [PMID: 36743527 PMCID: PMC9893394 DOI: 10.1177/17588359231152839] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023] Open
Abstract
Despite key advances in the treatment of prostate cancer (PCa), a proportion of men have de novo resistance, and all will develop resistance to current therapeutics over time. Aberrant lipid metabolism has long been associated with prostate carcinogenesis and progression, but more recently there has been an explosion of preclinical and clinical data which is informing new clinical trials. This review explores the epidemiological links between obesity and metabolic syndrome and PCa, the evidence for altered circulating lipids in PCa and their potential role as biomarkers, as well as novel therapeutic strategies for targeting lipids in men with PCa, including therapies widely used in cardiovascular disease such as statins, metformin and lifestyle modification, as well as novel targeted agents such as sphingosine kinase inhibitors, DES1 inhibitors and agents targeting FASN and beta oxidation.
Collapse
Affiliation(s)
- Tahlia Scheinberg
- Medical Oncology, Chris O’Brien Lifehouse, Camperdown NSW, Australia,Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia,University of Sydney, Camperdown, NSW, Australia
| | - Blossom Mak
- Medical Oncology, Chris O’Brien Lifehouse, Camperdown NSW, Australia,Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia,University of Sydney, Camperdown, NSW, Australia
| | - Lisa Butler
- Prostate Cancer Research Group, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia,South Australian Immunogenomics Cancer Institute and Freemason’s Centre for Male Health and Wellbeing, University of Adelaide, South Australia, Australia
| | - Luke Selth
- South Australian Immunogenomics Cancer Institute and Freemason’s Centre for Male Health and Wellbeing, University of Adelaide, South Australia, Australia,Dame Roma Mitchell Cancer Research Labs, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia,Flinders Health and Medical Research Institute, Flinders University, College of Medicine and Public Health, Bedford Park, Australia
| | | |
Collapse
|
20
|
De Martino M, Daviaud C, Hajjar E, Vanpouille-Box C. Fatty acid metabolism and radiation-induced anti-tumor immunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 376:121-141. [PMID: 36997267 DOI: 10.1016/bs.ircmb.2023.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fatty acid metabolic reprogramming has emerged as a major regulator of anti-tumor immune responses with large body of evidence that demonstrate its ability to impact the differentiation and function of immune cells. Therefore, depending on the metabolic cues that stem in the tumor microenvironment, the tumor fatty acid metabolism can tilt the balance of inflammatory signals to either promote or impair anti-tumor immune responses. Oxidative stressors such as reactive oxygen species generated from radiation therapy can rewire the tumor energy supply, suggesting that radiation therapy can further perturb the energy metabolism of a tumor by promoting fatty acid production. In this review, we critically discuss the network of fatty acid metabolism and how it regulates immune response especially in the context of radiation therapy.
Collapse
|
21
|
Beebe J, Josephraj S, Wang CJ, Danielson J, Cui Q, Huang C, Barlow L, Zhang RH, Zhang T, Nakshatri H, Dong Z, Li X, Liu JY, Zhang JT. Therapeutic Activity of the Lansoprazole Metabolite 5-Hydroxy Lansoprazole Sulfide in Triple-Negative Breast Cancer by Inhibiting the Enoyl Reductase of Fatty Acid Synthase. J Med Chem 2022; 65:13681-13691. [PMID: 36257066 DOI: 10.1021/acs.jmedchem.2c00642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fatty acid synthase (FASN), a sole cytosolic enzyme responsible for de-novo lipid synthesis, is overexpressed in cancer but not in normal non-lipogenic tissues. FASN has been targeted, albeit no such inhibitor has been approved. Proton pump inhibitors (PPIs), approved for digestive disorders, were found to inhibit FASN with anticancer activities in attempting to repurpose Food and Drug Administration-approved drugs. Indeed, PPI usage benefited breast cancer patients and increased their response rate. Due to structural similarity, we thought that their metabolites might extend anticancer effects of PPIs by inhibiting FASN. Here, we tested this hypothesis and found that 5-hydroxy lansoprazole sulfide (5HLS), the end lansoprazole metabolite, was more active than lansoprazole in inhibiting FASN function and regulation of NHEJ repair of oxidative DNA damage via PARP1. Surprisingly, 5HLS inhibits the enoyl reductase, whereas lansoprazole inhibits the thioesterase of FASN. Thus, PPI metabolites may contribute to the lasting anticancer effects of PPIs by inhibiting FASN.
Collapse
Affiliation(s)
- Jenny Beebe
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Sophia Josephraj
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Chao J Wang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Jacob Danielson
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Qingbin Cui
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Caoqinglong Huang
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Lincoln Barlow
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Ryan H Zhang
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Taolan Zhang
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Zizheng Dong
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Xiaohong Li
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jing-Yuan Liu
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jian-Ting Zhang
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| |
Collapse
|
22
|
Ballinger TJ, Djuric Z, Sardesai S, Hovey KM, Andrews CA, Brasky TM, Zhang JT, Rohan TE, Saquib N, Shadyab AH, Simon M, Wactawski-Wende J, Wallace R, Kato I. Proton Pump Inhibitor Use and Obesity-Associated Cancer in the Women's Health Initiative. Nutr Cancer 2022; 75:265-275. [PMID: 35968582 PMCID: PMC9772040 DOI: 10.1080/01635581.2022.2108467] [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: 05/06/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 12/24/2022]
Abstract
Proton pump inhibitors (PPIs) have off-target activity on fatty acid synthase (FASN), a critical enzyme in energy balance and cancer growth. We evaluated risk of common obesity-related cancers: breast, colorectal (CRC), and endometrial, with use of PPI and histamine-2 receptor antagonists (H2RA) in 124,931 postmenopausal women enrolled in the Women's Health Initiative. Incident cancer cases were physician-adjudicated. Cox proportional hazards models were used to estimate multivariable hazard ratios (HR) and 95% confidence intervals (CI) for cancer incidence after year 3. There were 7956 PPI ever users and 9398 H2RA only users. Ever use of either PPI or H2RA was not associated with risk of breast cancer (n = 9186) nor risk of endometrial cancer (n = 1231). The risk of CRC (n = 2280) was significantly lower in PPI users (HR = 0.75, 95% CI = 0.61-0.92), but not in H2RA users (HR = 1.13, 95% CI = 0.97-1.31). The association of PPI use with CRC was apparent regardless of BMI or NSAID use, and was stronger with longer PPI duration (p = 0.006) and potency (p = 0.005). The findings that PPI use, but not H2RA use, demonstrate an inverse dose-response relationship with risk of CRC is consistent with preclinical data showing FASN inhibition prevents colon cancer progression and supports a role of PPI in CRC prevention.
Collapse
Affiliation(s)
- Tarah J. Ballinger
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Zora Djuric
- Departments of Family Medicine and Nutritional Sciences, University of Michigan, Ann Arbor, MI
| | - Sagar Sardesai
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH
| | - Kathleen M. Hovey
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY
| | - Chris A. Andrews
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY
| | - Theodore M. Brasky
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH
| | - Jian Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Nazmus Saquib
- College of Medicine, Sulaiman AlRajhi University, Saudi Arabia
| | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA
| | - Michael Simon
- Department of Oncology, Karmanos Cancer Institute at Wayne State University School of Medicine, Detroit, MI
| | - Jean Wactawski-Wende
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Robert Wallace
- Department of Epidemiology, Iowa College of Public Health, Iowa City, IA
| | - Ikuko Kato
- Department of Oncology, Karmanos Cancer Institute at Wayne State University School of Medicine, Detroit, MI
- Department of Pathology, Wayne State University School of Medicine, Detroit
| |
Collapse
|
23
|
Yang R, Yi M, Xiang B. Novel Insights on Lipid Metabolism Alterations in Drug Resistance in Cancer. Front Cell Dev Biol 2022; 10:875318. [PMID: 35646898 PMCID: PMC9136290 DOI: 10.3389/fcell.2022.875318] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/13/2022] [Indexed: 12/26/2022] Open
Abstract
Chemotherapy is one of the primary treatments for most human cancers. Despite great progress in cancer therapeutics, chemotherapy continues to be important for improving the survival of cancer patients, especially for those who has unresectable metastatic tumors or fail to respond to immunotherapy. However, intrinsic or acquired chemoresistance results in tumor recurrence, which remains a major obstacle in anti-cancer treatment. The high prevalence of chemoresistant cancer makes it urgent to deepen our understanding on chemoresistance mechanisms and to develop novel therapeutic strategies. Multiple mechanisms, including drug efflux, enhanced DNA damage reparability, increased detoxifying enzymes levels, presence of cancer stem cells (CSCs), epithelial mesenchymal transition (EMT), autophagy, ferroptosis and resistance to apoptosis, underlie the development of chemoresistance. Recently, accumulating evidence suggests that lipid metabolism alteration is closely related to drug resistance in tumor. Targeting lipid metabolism in combination with traditional chemotherapeutic drugs is a promising strategy to overcome drug resistance. Therefore, this review compiles the current knowledge about aberrant lipid metabolism in chemoresistant cancer, mainly focusing on aberrant fatty acid metabolism, and presents novel therapeutic strategies targeting altered lipid metabolism to overcome chemoresistance in cancer.
Collapse
Affiliation(s)
- Ruixue Yang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hypertension Center, FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Mei Yi
- Department of Dermatology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Bo Xiang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
24
|
Xiong Q, Feng D, Wang Z, Ying Y, Xu C, Wei Q, Zeng S, Yang L. Fatty Acid Synthase Is the Key Regulator of Fatty Acid Metabolism and Is Related to Immunotherapy in Bladder Cancer. Front Immunol 2022; 13:836939. [PMID: 35392075 PMCID: PMC8982515 DOI: 10.3389/fimmu.2022.836939] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/16/2022] [Indexed: 02/05/2023] Open
Abstract
Fatty acid metabolism (FAM) genes are potentially useful for predicting prognosis and immunotherapy response in bladder cancer (BC). To examine this, we constructed a prognostic model and identified key FAM genes in BC. Using transcriptional expression profiles and clinical data of BC patients from public datasets and Changhai (CH) hospital, we built and validated a risk-score model based on 13 prognostic FAM genes. Differential gene expression identified fatty acid synthase (FASN) as central to fatty acid metabolism in BC. FASN was differentially expressed between normal and tumor tissue, and was related to survival. In the CH dataset, FASN independently predicted muscle-invasive BC. FASN differential expression was significantly related to immune-cell infiltration and patients with low FASN expression responded better to immune checkpoint inhibitor (ICI) treatment. SREBF1 was predicted as the most significant transcription factor for FASN. Competing endogenous RNA network analysis suggested that lncRNA AC107027.3 may upregulate FASN by competitively binding miR-27A-3p, thereby regulating the immunotherapy response in BC. Dasatinib and temsirolimus are potential FASN-targeting drugs. Our model efficiently predicted prognosis in BC. FASN is central to fatty acid metabolism, and a potential indicator and regulator of ICI treatment.
Collapse
Affiliation(s)
- Qiao Xiong
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China.,Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Ziwei Wang
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yidie Ying
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chuanliang Xu
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Shuxiong Zeng
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Tang Y, Tian W, Xie J, Zou Y, Wang Z, Li N, Zeng Y, Wu L, Zhang Y, Wu S, Xie X, Yang L. Prognosis and Dissection of Immunosuppressive Microenvironment in Breast Cancer Based on Fatty Acid Metabolism-Related Signature. Front Immunol 2022; 13:843515. [PMID: 35432381 PMCID: PMC9009264 DOI: 10.3389/fimmu.2022.843515] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 12/18/2022] Open
Abstract
Fatty acid metabolism has been deciphered to augment tumorigenesis and disease progression in addition to therapy resistance via strengthened lipid synthesis, storage, and catabolism. Breast cancer is strongly associated with the biological function of fatty acid metabolism owing to the abundant presence of adipocytes in breast tissue. It has been unraveled that tumor cells exhibit considerable plasticity based on fatty acid metabolism, responding to extra-tumoral and a range of metabolic signals, in which tumor microenvironment plays a pivotal role. However, the prognostic significance of fatty acid metabolism in breast cancer remains to be further investigated. Alongside these insights, we retrieved 269 reliable fatty acid metabolism-related genes (FMGs) and identified the landscape of copy number variations and expression level among those genes. Additionally, 11 overall survival-related FMGs were clarified by univariate Cox hazards regression analysis in The Cancer Genome Atlas (TCGA) and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) databases. Subsequently, a prognostic signature based on 6 overall survival (OS)-related FMGs was generated using Lasso Cox hazards regression analysis in TCGA dataset and was validated in two external cohorts. The correlation between the signature and several essential clinical parameters, including T, N, and PAM50 subtypes, was unveiled by comparing the accumulating signature value in various degrees. Furthermore, an optimal nomogram incorporating the signature, age, and American Joint Committee on Cancer (AJCC) stage was constructed, and the discrimination was verified by C-index, the calibration curve, and the decision curve analysis. The underlying implications for immune checkpoints inhibitors, the landscape of tumor immune microenvironment, and the predictive significance in therapy resistance to diverse strategies were depicted ultimately. In conclusion, our findings indicate the potential prognostic connotation of fatty acid metabolism in breast cancer, supporting novel insights into breast cancer patients' prognosis and administrating effective immunotherapy.
Collapse
Affiliation(s)
- Yuhui Tang
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenwen Tian
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jindong Xie
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yutian Zou
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zehao Wang
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ning Li
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yan Zeng
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Linyu Wu
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yue Zhang
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Song Wu
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lu Yang
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People’s Hospital, Guangzhou, China
| |
Collapse
|
26
|
Tőkés AM, Vári-Kakas S, Kulka J, Törőcsik B. Tumor Glucose and Fatty Acid Metabolism in the Context of Anthracycline and Taxane-Based (Neo)Adjuvant Chemotherapy in Breast Carcinomas. Front Oncol 2022; 12:850401. [PMID: 35433453 PMCID: PMC9008716 DOI: 10.3389/fonc.2022.850401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is characterized by considerable metabolic diversity. A relatively high percentage of patients diagnosed with breast carcinoma do not respond to standard-of-care treatment, and alteration in metabolic pathways nowadays is considered one of the major mechanisms responsible for therapeutic resistance. Consequently, there is an emerging need to understand how metabolism shapes therapy response, therapy resistance and not ultimately to analyze the metabolic changes occurring after different treatment regimens. The most commonly applied neoadjuvant chemotherapy regimens in breast cancer contain an anthracycline (doxorubicin or epirubicin) in combination or sequentially administered with taxanes (paclitaxel or docetaxel). Despite several efforts, drug resistance is still frequent in many types of breast cancer, decreasing patients’ survival. Understanding how tumor cells rapidly rewire their signaling pathways to persist after neoadjuvant cancer treatment have to be analyzed in detail and in a more complex system to enable scientists to design novel treatment strategies that target different aspects of tumor cells and tumor resistance. Tumor heterogeneity, the rapidly changing environmental context, differences in nutrient use among different cell types, the cooperative or competitive relationships between cells pose additional challenges in profound analyzes of metabolic changes in different breast carcinoma subtypes and treatment protocols. Delineating the contribution of metabolic pathways to tumor differentiation, progression, and resistance to different drugs is also the focus of research. The present review discusses the changes in glucose and fatty acid pathways associated with the most frequently applied chemotherapeutic drugs in breast cancer, as well the underlying molecular mechanisms and corresponding novel therapeutic strategies.
Collapse
Affiliation(s)
- Anna Mária Tőkés
- 2nd Department of Pathology, Semmelweis University Budapest, Budapest, Hungary
- *Correspondence: Anna Mária Tőkés,
| | - Stefan Vári-Kakas
- Department of Computers and Information Technology, Faculty of Electrical Engineering and Information Technology, University of Oradea, Oradea, Romania
| | - Janina Kulka
- 2nd Department of Pathology, Semmelweis University Budapest, Budapest, Hungary
| | - Beáta Törőcsik
- Department of Biochemistry, Semmelweis University Budapest, Budapest, Hungary
| |
Collapse
|
27
|
Pharmacological fatty acid synthase inhibitors differently affect the malignant phenotype of oral cancer cells. Arch Oral Biol 2022; 135:105343. [DOI: 10.1016/j.archoralbio.2021.105343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 12/25/2022]
|
28
|
Hussein S, Khanna P, Yunus N, Gatza ML. Nuclear Receptor-Mediated Metabolic Reprogramming and the Impact on HR+ Breast Cancer. Cancers (Basel) 2021; 13:cancers13194808. [PMID: 34638293 PMCID: PMC8508306 DOI: 10.3390/cancers13194808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Breast cancer is the most commonly diagnosed and second leading cause of cancer-related deaths in women in the United States, with hormone receptor positive (HR+) tumors representing more than two-thirds of new cases. Recent evidence has indicated that dysregulation of multiple metabolic programs, which can be driven through nuclear receptor activity, is essential for tumor genesis, progression, therapeutic resistance and metastasis. This study will review the current advances in our understanding of the impact and implication of altered metabolic processes driven by nuclear receptors, including hormone-dependent signaling, on HR+ breast cancer. Abstract Metabolic reprogramming enables cancer cells to adapt to the changing microenvironment in order to maintain metabolic energy and to provide the necessary biological macromolecules required for cell growth and tumor progression. While changes in tumor metabolism have been long recognized as a hallmark of cancer, recent advances have begun to delineate the mechanisms that modulate metabolic pathways and the consequence of altered signaling on tumorigenesis. This is particularly evident in hormone receptor positive (HR+) breast cancers which account for approximately 70% of breast cancer cases. Emerging evidence indicates that HR+ breast tumors are dependent on multiple metabolic processes for tumor progression, metastasis, and therapeutic resistance and that changes in metabolic programs are driven, in part, by a number of key nuclear receptors including hormone-dependent signaling. In this review, we discuss the mechanisms and impact of hormone receptor mediated metabolic reprogramming on HR+ breast cancer genesis and progression as well as the therapeutic implications of these metabolic processes in this disease.
Collapse
Affiliation(s)
- Shaimaa Hussein
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; (S.H.); (P.K.)
- Department of Radiation Oncology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Pooja Khanna
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; (S.H.); (P.K.)
- Department of Radiation Oncology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- School of Arts and Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA;
| | - Neha Yunus
- School of Arts and Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA;
| | - Michael L. Gatza
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; (S.H.); (P.K.)
- Department of Radiation Oncology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- School of Arts and Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA;
- Correspondence: ; Tel.: +1-732-235-8751
| |
Collapse
|