1
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
2
|
Yuan W, Lu G, Zhao Y, He X, Liao S, Wang Z, Lei X, Xie Z, Yang X, Tang S, Tang G, Deng X. Intranuclear TCA and mitochondrial overload: The nascent sprout of tumors metabolism. Cancer Lett 2025; 613:217527. [PMID: 39909232 DOI: 10.1016/j.canlet.2025.217527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/19/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Abnormal glucose metabolism in tumors is a well-known form of metabolic reprogramming in tumor cells, the most representative of which, the Warburg effect, has been widely studied and discussed since its discovery. However, contradictions in a large number of studies and suboptimal efficacy of drugs targeting glycolysis have prompted us to further deepen our understanding of glucose metabolism in tumors. Here, we review recent studies on mitochondrial overload, nuclear localization of metabolizing enzymes, and intranuclear TCA (nTCA) in the context of the anomalies produced by inhibition of the Warburg effect. We provide plausible explanations for many of the contradictory points in the existing studies, including the causes of the Warburg effect. Furthermore, we provide a detailed prospective discussion of these studies in the context of these new findings, providing new ideas for the use of nTCA and mitochondrial overload in tumor therapy.
Collapse
Affiliation(s)
- Weixi Yuan
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guozhong Lu
- 922nd Hospital of Hengyang, 421001, Hunan, China
| | - Yin Zhao
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiang He
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Senyi Liao
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhe Wang
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyong Lei
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Department of Pharmacy, Xiangnan University, Chenzhou, 423000, China
| | - Zhizhong Xie
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaoyan Yang
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Department of Pharmacy, Xiangnan University, Chenzhou, 423000, China
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery Systems (2018TP1044), Hunan, 410007, China.
| | - Guotao Tang
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Xiangping Deng
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
3
|
Niu Z, He J, Wang S, Xue B, Zhang H, Hou R, Xu Z, Sun J, He F, Pei X. Targeting Glycolysis for Treatment of Breast Cancer Resistance: Current Progress and Future Prospects. Int J Biol Sci 2025; 21:2589-2605. [PMID: 40303296 PMCID: PMC12035887 DOI: 10.7150/ijbs.109803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/07/2025] [Indexed: 05/02/2025] Open
Abstract
Breast cancer stands as one of the most prevalent malignant tumors threatening women's health and is a leading cause of cancer-related mortality. Its treatment faces significant challenges, including drug tolerance and disease recurrence. Glycolysis serves not only as a critical metabolic pathway for energy acquisition in breast cancer cells but also essentially promotes tumor proliferation, invasion, metastasis, and the development of resistance to therapy. Recent studies have revealed a close association between glycolytic reprogramming and drug resistance in breast cancer, with high-level glycolysis emerging as a hallmark of malignancy, deeply involved in the initiation and progression of tumors. This review summarizes recent advances in research on key enzymes and signaling pathways regulating glycolysis within the bodies of breast cancer patients. It explores in depth these molecular mechanisms and their complex interaction networks, offering a fresh perspective on overcoming drug resistance in breast cancer. Moreover, it underscores the importance of developing specific inhibitors targeting key enzymes and regulators of glycolysis and suggests that combining such inhibitors with existing anticancer drugs could substantially enhance therapeutic outcomes for breast cancer patients and reduce the occurrence of drug resistance.
Collapse
Affiliation(s)
- Zixu Niu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jing He
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Siyuan Wang
- Pharmaceutical College, Guangxi Medical University, Nanning, 530021, China
| | - Bingjian Xue
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ruohan Hou
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zimeng Xu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jing Sun
- The First Clinical Medical College of Zhengzhou University, Zhengzhou, 450052, China
| | - Fucheng He
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xinhong Pei
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
4
|
Li D, Jiang M, Song Y, Liang X, Lv J, Zhang L, Li Z, Fan L, Du H. Preventive vs. therapeutic effects of Shoutai Wan: Maintaining an acidic microenvironment at the maternal-fetal interface to promote angiogenesis and minimize pregnancy loss in RSA mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119345. [PMID: 39824268 DOI: 10.1016/j.jep.2025.119345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The classic TCM prescription, Shoutai Wan (STW), is extensively used in clinical settings to manage threatened miscarriage and Recurrent spontaneous abortion (RSA). The complexity of pregnancy physiology, coupled with diverse etiologies, and the specificity of energy metabolism for normal embryo attachment and development, pose challenges to clinical diagnosis and treatment. The specific molecular mechanisms of how STW regulates these biological processes and contributes to the treatment of RSA remain to be elucidated. AIM OF THE STUDY This study aims to investigate the causes of early pregnancy loss in RSA mice and explore how STW mitigates this loss. MATERIALS AND METHODS An RSA mouse model will be established and treated with STW and Dydrogesterone (DYD). Embryo loss will be quantified on the 14th day of pregnancy, and embryos will be collected on the 6th and 10th days to observe the embryonic condition and assess pathological changes. The study will analyze aerobic glycolysis and angiogenesis at the maternal-fetal interface (MFI). Additionally, STW on a knockdown LDHA mouse model and Human Endometrial Microvascular Endothelial Cells (HEMECs) in vitro will also be examined to verify the mechanism. RESULTS Compared with the control group, the RSA group exhibited significant embryo loss, and reduced levels of aerobic glycolysis at the MFI, the precarious acidic microenvironment (AME), and the PI3K/AKT/mTOR signaling axis downregulated, leading to impaired angiogenesis, which ameliorated following STW treatment. STW treatment enhanced key aerobic glycolysis enzymes-HK2, PKM2, LDHA-and lactate levels, thereby maintaining the AME and upregulating the PI3K/AKT/mTOR axis. This, in turn, promoted the expression of angiogenesis-related factors (VEGFA and VEGFR2) at the MFI, thereby improving angiogenesis, and the same was seen in sh-LDHA mice. In vitro studies confirmed that STW could counteract the glycolysis decline caused by increased oxygen levels, a recovery that was impaired after LDHA knockdown or PI3K inhibition. CONCLUSIONS In RSA mice, disturbances in aerobic glycolysis at the MFI prevent the maintenance of a stable AME, thus impairing angiogenesis and leading to embryo loss, and STW effectively improve early pregnancy outcomes, and laying the foundation for uterine spiral artery remodeling.
Collapse
Affiliation(s)
- Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China
| | - Min Jiang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China
| | - Yajing Song
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China
| | - Xiao Liang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China
| | - Jingfang Lv
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China
| | - Li Zhang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; First College of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Lijie Fan
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China; Hebei Key Laboratory of Integrative Medicine on Liver-kidney Patterns, Shijiazhuang, 050091, China
| | - Huilan Du
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China; Hebei Key Laboratory of Integrative Medicine on Liver-kidney Patterns, Shijiazhuang, 050091, China.
| |
Collapse
|
5
|
Maiti A, Mondal S, Choudhury S, Bandopadhyay A, Mukherjee S, Sikdar N. Oncometabolites in pancreatic cancer: Strategies and its implications. World J Exp Med 2024; 14:96005. [PMID: 39713078 PMCID: PMC11551704 DOI: 10.5493/wjem.v14.i4.96005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/24/2024] [Accepted: 09/14/2024] [Indexed: 10/31/2024] Open
Abstract
Pancreatic cancer (PanCa) is a catastrophic disease, being third lethal in both the genders around the globe. The possible reasons are extreme disease invasiveness, highly fibrotic and desmoplastic stroma, dearth of confirmatory diagnostic approaches and resistance to chemotherapeutics. This inimitable tumor microenvironment (TME) or desmoplasia with excessive extracellular matrix accumulation, create an extremely hypovascular, hypoxic and nutrient-deficient zone inside the tumor. To survive, grow and proliferate in such tough TME, pancreatic tumor and stromal cells transform their metabolism. Transformed glucose, glutamine, fat, nucleotide metabolism and inter-metabolite communication between tumor and TME in synergism, impart therapy resistance, and immunosuppression in PanCa. Thus, a finer knowledge of altered metabolism would uncover its metabolic susceptibilities. These unique metabolic targets may help to device novel diagnostic/prognostic markers and therapeutic strategies for better management of PanCa. In this review, we sum up reshaped metabolic pathways in PanCa to formulate detection and remedial strategies of this devastating disease.
Collapse
Affiliation(s)
- Arunima Maiti
- Suraksha Diagnostics Pvt Ltd, Newtown, Rajarhat, Kolkata 700156, West Bengal, India
| | - Susmita Mondal
- Department of Zoology, Diamond Harbour Women’s University, Diamond Harbour 743368, West Bengal, India
| | - Sounetra Choudhury
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, West Bengal, India
| | | | - Sanghamitra Mukherjee
- Department of Pathology, RG Kar Medical College and Hospital, Kolkata 700004, West Bengal, India
| | - Nilabja Sikdar
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, West Bengal, India
- Scientist G, Estuarine and Coastal Studies Foundation, Howrah 711101, West Bengal, India
| |
Collapse
|
6
|
Su C, Yang J, Ding J, Ding H. Differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma based on serum lactate dehydrogenase combined with CA-125 and CA19-9: A retrospective cohort study. Medicine (Baltimore) 2024; 103:e40776. [PMID: 39612391 PMCID: PMC11608665 DOI: 10.1097/md.0000000000040776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
This study aims to construct and validate a nomogram for the differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma. We retrospectively studied the clinical characteristics of patients with ovarian endometriosis cysts and ovarian cystadenomas from January 1, 2021, to June 1, 2022. Independent risk factors for differential diagnosis were investigated using univariate and multivariate logistic regression analyses. Based on these factors, a differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma was established. The performance of the nomogram model was assessed by internal validation using bootstrapping resampling. Decision curve analysis (DCA) was performed to evaluate the net clinical benefit of the model. Immunohistochemistry showed that lactate dehydrogenase (LDH) A was overexpressed in ectopic endometrial tissues compared to that in normal endometrial tissues. In multivariate analysis, LDH, CA-125, and CA19-9 were identified as independent risk factors for the differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma. LDH levels >135.50 U/L combined with CA-125 levels >25.20 U/mL and CA19-9 levels >13.59 U/mL as single covariates had a high value in the differential diagnosis of ovarian endometriosis cysts versus ovarian cystadenoma. The area under the receiver operating characteristic curve (ROC) of the nomogram constructed using LDH, CA-125, and CA19-9 expression data was 0.873 (95% CI, 0.827-0.920), and the bootstrap-validated concordance index (C-index) was 0.871. Decision curve analysis confirmed that the nomogram model had excellent clinical utility. Based on serum lactate dehydrogenase combined with CA-125 and CA19-9, we constructed and validated a nomogram for the differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma to help physicians formulate the optimal treatment strategy.
Collapse
Affiliation(s)
- Chang Su
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Anhui, Wuhu, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Anhui, Wuhu, China
| | - Jian Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Anhui, Wuhu, China
| | - Jin Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Anhui, Wuhu, China
| | - Huafeng Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Anhui, Wuhu, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Anhui, Wuhu, China
| |
Collapse
|
7
|
Cenigaonandia‐Campillo A, Garcia‐Bautista A, Rio‐Vilariño A, Cebrian A, del Puerto L, Pellicer JA, Gabaldón JA, Pérez‐Sánchez H, Carmena‐Bargueño M, Meroño C, Traba J, Fernandez‐Aceñero MJ, Baños‐Herraiz N, Mozas‐Vivar L, Núñez‐Delicado E, Garcia‐Foncillas J, Aguilera Ó. Vitamin-C-dependent downregulation of the citrate metabolism pathway potentiates pancreatic ductal adenocarcinoma growth arrest. Mol Oncol 2024; 18:2212-2233. [PMID: 38425123 PMCID: PMC11467799 DOI: 10.1002/1878-0261.13616] [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: 07/31/2023] [Revised: 01/17/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
In pancreatic ductal adenocarcinoma (PDAC), metabolic rewiring and resistance to standard therapy are closely associated. PDAC cells show enormous requirements for glucose-derived citrate, the first rate-limiting metabolite in the synthesis of new lipids. Both the expression and activity of citrate synthase (CS) are extraordinarily upregulated in PDAC. However, no previous relationship between gemcitabine response and citrate metabolism has been documented in pancreatic cancer. Here, we report for the first time that pharmacological doses of vitamin C are capable of exerting an inhibitory action on the activity of CS, reducing glucose-derived citrate levels. Moreover, ascorbate targets citrate metabolism towards the de novo lipogenesis pathway, impairing fatty acid synthase (FASN) and ATP citrate lyase (ACLY) expression. Lowered citrate availability was found to be directly associated with diminished proliferation and, remarkably, enhanced gemcitabine response. Moreover, the deregulated citrate-derived lipogenic pathway correlated with a remarkable decrease in extracellular pH through inhibition of lactate dehydrogenase (LDH) and overall reduced glycolytic metabolism. Modulation of citric acid metabolism in highly chemoresistant pancreatic adenocarcinoma, through molecules such as vitamin C, could be considered as a future clinical option to improve patient response to standard chemotherapy regimens.
Collapse
Affiliation(s)
| | - Ana Garcia‐Bautista
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Anxo Rio‐Vilariño
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Arancha Cebrian
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Laura del Puerto
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - José Antonio Pellicer
- Molecular Recognition and Encapsulation Research Group (REM), Health Sciences DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - José Antonio Gabaldón
- Molecular Recognition and Encapsulation Research Group (REM), Health Sciences DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Horacio Pérez‐Sánchez
- Bioinformatics and High‐Performance Computing Research Group (BIO‐HPC), Computer Engineering DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Miguel Carmena‐Bargueño
- Bioinformatics and High‐Performance Computing Research Group (BIO‐HPC), Computer Engineering DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Carolina Meroño
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones CientíficasUniversidad Autónoma de Madrid (CSIC‐UAM)Spain
- Instituto Universitario de Biología Molecular‐UAM (IUBM‐UAM), Departamento de Biología MolecularUniversidad Autónoma de MadridSpain
| | - Javier Traba
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones CientíficasUniversidad Autónoma de Madrid (CSIC‐UAM)Spain
- Instituto Universitario de Biología Molecular‐UAM (IUBM‐UAM), Departamento de Biología MolecularUniversidad Autónoma de MadridSpain
| | | | | | - Lorena Mozas‐Vivar
- Preclinical programe START Madrid‐FJD Hospital fundación Jiménez DíazSpain
| | - Estrella Núñez‐Delicado
- Molecular Recognition and Encapsulation Research Group (REM), Health Sciences DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Jesús Garcia‐Foncillas
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Óscar Aguilera
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
- Universidad Católica de Murcia (UCAM)Spain
| |
Collapse
|
8
|
Lim SA. Metabolic reprogramming of the tumor microenvironment to enhance immunotherapy. BMB Rep 2024; 57:388-399. [PMID: 38919017 PMCID: PMC11444991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/27/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024] Open
Abstract
Immunotherapy represents a promising treatment strategy for targeting various tumor types. However, the overall response rate is low due to the tumor microenvironment (TME). In the TME, numerous distinct factors actively induce immunosuppression, restricting the efficacy of anticancer immune reactions. Recently, metabolic reprogramming of tumors has been recognized for its role in modulating the tumor microenvironment to enhance immune cell responses in the TME. Furthermore, recent elucidations underscore the critical role of metabolic limitations imposed by the tumor microenvironment on the effectiveness of antitumor immune cells, guiding the development of novel immunotherapeutic approaches. Hence, achieving a comprehensive understanding of the metabolic requirements of both cancer and immune cells within the TME is pivotal. This insight not only aids in acknowledging the current limitations of clinical practices but also significantly shapes the trajectory of future research endeavors in the domain of cancer immunotherapy. In addition, therapeutic interventions targeting metabolic limitations have exhibited promising potential as combinatory treatments across diverse cancer types. In this review, we first discuss the metabolic barriers in the TME. Second, we explore how the immune response is regulated by metabolites. Finally, we will review the current strategy for targeting metabolism to not simply inhibit tumor growth but also enhance antitumor immune responses. Thus, we could suggest potent combination therapy for improving immunotherapy with metabolic inhibitors. [BMB Reports 2024; 57(9): 388-399].
Collapse
Affiliation(s)
- Seon Ah Lim
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
- Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
9
|
Vats P, Singh J, Srivastava SK, Kumar A, Nema R. LncRNA TMPO-AS1 Promotes Triple-Negative Breast Cancer by Sponging miR-383-5p to Trigger the LDHA Axis. Asian Pac J Cancer Prev 2024; 25:2929-2944. [PMID: 39205592 PMCID: PMC11495453 DOI: 10.31557/apjcp.2024.25.8.2929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Understanding the heterogeneous nature of breast cancer, including the role of LDHA expression regulation via non-coding RNAs in prognosis, is still unknown, highlighting the need for more research into its molecular roles and diagnostic approaches. METHODS The study utilized various computer tools to analyze the differences between LDHA in tissues and cancer cells. It used data from TIMER 2.0, UALCAN, and TISIDB to study gene expression and survival outcomes in breast cancer patients. The study also used the Breast Cancer Gene Expression Miner to examine the relationship between LDHA gene expression and breast cancer type. Other tools included TCGAPortal, TNMplot, ctcRbase, GSCA, Enrichr, TISIDB, Oncomx, and TANRIC. The study then explored the relationship between tumor-infiltrating immune cells and LDHA formation using the GSCA and TISIDB repositories. We used Auto Dock Tools 1.5.6 to perform ligand binding analysis for LDHA, withanolides, and the known inhibitor LDH-IN-1. LigPlot+ and Pymol were used for visualization of protein-ligand complexes. RESULTS LDHA overexpression in breast cancer cells, metastatic tissue, and circulating tumor cells leads to shortened recurrence-free survival, overall survival, and distant metastasis-free survival. In invasive breast cell carcinoma, we observed that LDHA/HIF-1α /TMPO-AS1 are overexpressed while miR-383-5p is downregulated. This overexpression is associated with poor prognosis and may lead to Act_DC infiltration into the tumor microenvironment. Withanolides, viz., Withaferine A and Withanolide D, have shown high binding affinity with LDHA, with binding energies of -9.3kcal/mol and -10kcal/mol respectively. These could be attractive choices for small-molecule inhibitor design against LDHA.
Collapse
Affiliation(s)
- Prerna Vats
- Department of Bioscience, Manipal University Jaipur, University Jaipur, Dehmi Kalan, Jaipur-Ajmer Expressway, Jaipur, Rajasthan, 303007, India.
| | - Jai Singh
- Department of Bioscience, Manipal University Jaipur, University Jaipur, Dehmi Kalan, Jaipur-Ajmer Expressway, Jaipur, Rajasthan, 303007, India.
| | - Sandeep K. Srivastava
- Department of Bioscience, Manipal University Jaipur, University Jaipur, Dehmi Kalan, Jaipur-Ajmer Expressway, Jaipur, Rajasthan, 303007, India.
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Bhopal, Saket Nagar, Bhopal 462 020, Madhya Pradesh, India.
| | - Rajeev Nema
- Department of Bioscience, Manipal University Jaipur, University Jaipur, Dehmi Kalan, Jaipur-Ajmer Expressway, Jaipur, Rajasthan, 303007, India.
| |
Collapse
|
10
|
Kolokotronis T, Majchrzak-Stiller B, Buchholz M, Mense V, Strotmann J, Peters I, Skrzypczyk L, Liffers ST, Menkene LM, Wagner M, Glanemann M, Betsou F, Ammerlaan W, Schmidt R, Schröder C, Uhl W, Braumann C, Höhn P. Differential miRNA and Protein Expression Reveals miR-1285, Its Targets TGM2 and CDH-1, as Well as CD166 and S100A13 as Potential New Biomarkers in Patients with Diabetes Mellitus and Pancreatic Adenocarcinoma. Cancers (Basel) 2024; 16:2726. [PMID: 39123454 PMCID: PMC11311671 DOI: 10.3390/cancers16152726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/16/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Early detection of PDAC remains challenging due to the lack of early symptoms and the absence of reliable biomarkers. The aim of the present project was to identify miRNA and proteomics signatures discriminating PDAC patients with DM from nondiabetic PDAC patients. Proteomics analysis and miRNA array were used for protein and miRNA screening. We used Western blotting and Real-Time Quantitative Reverse Transcription polymerase chain reaction (qRT-PCR) for protein and miRNA validation. Comparisons between experimental groups with normal distributions were performed using one-way ANOVA followed by Tukey's post hoc test, and pairwise tests were performed using t-tests. p ≤ 0.05 was considered statistically significant. Protein clusters of differentiation 166 (CD166), glycoprotein CD63 (CD63), S100 calcium-binding protein A13 (S100A13), and tumor necrosis factor-β (TNF-β) were detected in the proteomics screening. The miRNA assay revealed a differential miRNA 1285 regulation. Previously described target proteins of miR-1285 cadherin-1 (CDH-1), cellular Jun (c-Jun), p53, mothers against decapentaplegic homolog 4 (Smad4), human transglutaminase 2 (TGM2) and yes-associated protein (YAP), were validated via Western blotting. miR-1285-3p was successfully validated as differentially regulated in PDAC + DM via qRT-PCR. Overall, our data suggest miRNA1285-3p, TGM2, CDH-1, CD166, and S100A13 as potential meaningful biomarker candidates to characterize patients with PDAC + DM. Data are available via ProteomeXchange with the identifier PXD053169.
Collapse
Affiliation(s)
- Theodoros Kolokotronis
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
- Institute of Pathology and Surgical Clinic, University Hospital of Saarland, Kirrberger Str. 100, 66424 Homburg, Germany; (L.M.M.); (M.W.)
| | - Britta Majchrzak-Stiller
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
| | - Marie Buchholz
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
| | - Vanessa Mense
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
| | - Johanna Strotmann
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
| | - Ilka Peters
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
| | - Lea Skrzypczyk
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
| | - Sven-Thorsten Liffers
- University Hospital Essen, Bridging Institute for Experimental Tumor Therapy, West German Tumor Center Essen, Hufelandstr. 55, 45147 Essen, Germany;
| | - Louise Massia Menkene
- Institute of Pathology and Surgical Clinic, University Hospital of Saarland, Kirrberger Str. 100, 66424 Homburg, Germany; (L.M.M.); (M.W.)
| | - Mathias Wagner
- Institute of Pathology and Surgical Clinic, University Hospital of Saarland, Kirrberger Str. 100, 66424 Homburg, Germany; (L.M.M.); (M.W.)
| | - Matthias Glanemann
- Institute of Pathology and Surgical Clinic, University Hospital of Saarland, Kirrberger Str. 100, 66424 Homburg, Germany; (L.M.M.); (M.W.)
| | - Fay Betsou
- CRBIP, Institut Pasteur, Université Paris Cite, 25 rue du Dr Roux, 75015 Paris, France;
| | - Wim Ammerlaan
- IBBL (Integrated BioBank of Luxembourg), 1, Rue Louis Rech, L-3555 Dudelange, Luxembourg;
| | - Ronny Schmidt
- Sciomics GmbH, Karl-Landsteiner Str. 6, 69151 Heidelberg, Germany; (R.S.); (C.S.)
| | - Christoph Schröder
- Sciomics GmbH, Karl-Landsteiner Str. 6, 69151 Heidelberg, Germany; (R.S.); (C.S.)
| | - Waldemar Uhl
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
| | - Chris Braumann
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
- Department of General, Visceral and Vascular Surgery, EvK Gelsenkirchen, University Duisburg-Essen, Munckelstr. 27, 45879 Gelsenkirchen, Germany
| | - Philipp Höhn
- St. Josef Hospital Bochum, Surgical Clinic, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany; (B.M.-S.); (M.B.); (V.M.); (J.S.); (I.P.); (L.S.); (W.U.); (C.B.); (P.H.)
| |
Collapse
|
11
|
Zhang S, Ta N, Zhang S, Li S, Zhu X, Kong L, Gong X, Guo M, Liu Y. Unraveling pancreatic ductal adenocarcinoma immune prognostic signature through a naive B cell gene set. Cancer Lett 2024; 594:216981. [PMID: 38795761 DOI: 10.1016/j.canlet.2024.216981] [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: 02/07/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC), a leading cause of cancer mortality, has a complex pathogenesis involving various immune cells, including B cells and their subpopulations. Despite emerging research on the role of these cells within the tumor microenvironment (TME), the detailed molecular interactions with tumor-infiltrating immune cells (TIICs) are not fully understood. METHODS We applied CIBERSORT to quantify TIICs and naive B cells, which are prognostic for PDAC. Marker genes from scRNA-seq and modular genes from weighted gene co-expression network analysis (WGCNA) were integrated to identify naive B cell-related genes. A prognostic signature was constructed utilizing ten machine-learning algorithms, with validation in external cohorts. We further assessed the immune cell diversity, ESTIMATE scores, and immune checkpoint genes (ICGs) between patient groups stratified by risk to clarify the immune landscape in PDAC. RESULTS Our analysis identified 994 naive B cell-related genes across single-cell and bulk transcriptomes, with 247 linked to overall survival. We developed a 12-gene prognostic signature using Lasso and plsRcox algorithms, which was confirmed by 10-fold cross-validation and showed robust predictive power in training and real-world cohorts. Notably, we observed substantial differences in immune infiltration between patients with high and low risk. CONCLUSION Our study presents a robust prognostic signature that effectively maps the complex immune interactions in PDAC, emphasizing the critical function of naive B cells and suggesting new avenues for immunotherapeutic interventions. This signature has potential clinical applications in personalizing PDAC treatment, enhancing the understanding of immune dynamics, and guiding immunotherapy strategies.
Collapse
Affiliation(s)
- Shichen Zhang
- Software Engineering Institute, East China Normal University, Shanghai 200062, China
| | - Na Ta
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai 200433, China
| | - Shihao Zhang
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai 200433, China
| | - Senhao Li
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai 200433, China
| | - Xinyu Zhu
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai 200433, China
| | - Lingyun Kong
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai 200433, China
| | - Xueqing Gong
- Software Engineering Institute, East China Normal University, Shanghai 200062, China.
| | - Meng Guo
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai 200433, China.
| | - Yanfang Liu
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai 200433, China; National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai 200433, China.
| |
Collapse
|
12
|
Frańczak MA, Borea F, Smoleński RT, Granchi C, Minutolo F, Giovannetti E, Peters GJ. Evaluation of the combination of gemcitabine, carboplatin, and lactate dehydrogenase A inhibitor on malignant mesothelioma. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 43:862-869. [PMID: 38898808 DOI: 10.1080/15257770.2024.2356201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/10/2024] [Accepted: 05/10/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVES Lactate dehydrogenase A (LDH-A) catalyzes the last step of glycolysis: supplying cells rapidly but inefficiently with ATP. Many tumors, including malignant mesothelioma (MM), have a high expression of LDH-A, which is associated with cancer aggressiveness. We aimed to determine whether the efficacy of the gemcitabine/carboplatin (Gem + Carbo) combination, widely used to treat this disease, could be increased by inhibition of LDH-A (by NHI-2). To this aim, we analyzed the growth inhibition of pleural and peritoneal MM by multiple combinations. METHODS The 72 h sulforhodamine B assay (SRB) was used to test the cytotoxicity of the combination of gemcitabine (in the range 0.1 - 400 nM) and carboplatin (0.01 - 40 µM) with a fixed concentration of NHI-2 (at IC25). We used pleural (H2452) and primary peritoneal (STO, MESO-II) MM cell lines, cultured at normoxic conditions. RESULTS NHI-2 did not increase the cytotoxicity of the combination of 100 nM gemcitabine and 10 µM carboplatin in peritoneal MM cell lines. The cell growth inhibition was 10% smaller after the triple combination than the Gem + Carbo treatment. CONCLUSIONS Inhibition of LDH-A did not increase the efficacy of gemcitabine and carboplatin in MM under normoxic conditions.
Collapse
Affiliation(s)
- Marika A Frańczak
- Department of Biochemistry, Medical University of Gdansk, Poland
- Department of Medical Oncology, Amsterdam University Medical Centers, The Netherlands
| | - Federica Borea
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Rozzano, Milan, Italy
| | | | | | | | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Centers, The Netherlands
- Fondazione Pisana per la Scienza, Pisa, Italy
| | - Godefridus J Peters
- Department of Biochemistry, Medical University of Gdansk, Poland
- Department of Medical Oncology, Amsterdam University Medical Centers, The Netherlands
| |
Collapse
|
13
|
Yadav D, Yadav A, Bhattacharya S, Dagar A, Kumar V, Rani R. GLUT and HK: Two primary and essential key players in tumor glycolysis. Semin Cancer Biol 2024; 100:17-27. [PMID: 38494080 DOI: 10.1016/j.semcancer.2024.03.001] [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: 02/04/2024] [Revised: 03/02/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024]
Abstract
Cancer cells reprogram their metabolism to become "glycolysis-dominant," which enables them to meet their energy and macromolecule needs and enhancing their rate of survival. This glycolytic-dominancy is known as the "Warburg effect", a significant factor in the growth and invasion of malignant tumors. Many studies confirmed that members of the GLUT family, specifically HK-II from the HK family play a pivotal role in the Warburg effect, and are closely associated with glucose transportation followed by glucose metabolism in cancer cells. Overexpression of GLUTs and HK-II correlates with aggressive tumor behaviour and tumor microenvironment making them attractive therapeutic targets. Several studies have proven that the regulation of GLUTs and HK-II expression improves the treatment outcome for various tumors. Therefore, small molecule inhibitors targeting GLUT and HK-II show promise in sensitizing cancer cells to treatment, either alone or in combination with existing therapies including chemotherapy, radiotherapy, immunotherapy, and photodynamic therapy. Despite existing therapies, viable methods to target the glycolysis of cancer cells are currently lacking to increase the effectiveness of cancer treatment. This review explores the current understanding of GLUT and HK-II in cancer metabolism, recent inhibitor developments, and strategies for future drug development, offering insights into improving cancer treatment efficacy.
Collapse
Affiliation(s)
- Dhiraj Yadav
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India; Drug Discovery, Jubilant Biosys, Greater Noida, Noida, Uttar Pradesh, India
| | - Anubha Yadav
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India
| | - Sujata Bhattacharya
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India
| | - Akansha Dagar
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-Ku, Yokohama 236-0027, Japan
| | - Vinit Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India.
| | - Reshma Rani
- Drug Discovery, Jubilant Biosys, Greater Noida, Noida, Uttar Pradesh, India.
| |
Collapse
|
14
|
Littleflower AB, Parambil ST, Antony GR, Subhadradevi L. The determinants of metabolic discrepancies in aerobic glycolysis: Providing potential targets for breast cancer treatment. Biochimie 2024; 220:107-121. [PMID: 38184121 DOI: 10.1016/j.biochi.2024.01.003] [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: 08/10/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 01/08/2024]
Abstract
Altered aerobic glycolysis is the robust mechanism to support cancer cell survival and proliferation beyond the maintenance of cellular energy metabolism. Several investigators portrayed the important role of deregulated glycolysis in different cancers, including breast cancer. Breast cancer is the most ubiquitous form of cancer and the primary cause of cancer death in women worldwide. Breast cancer with increased glycolytic flux is hampered to eradicate with current therapies and can result in tumor recurrence. In spite of the low order efficiency of ATP production, cancer cells are highly addicted to glycolysis. The glycolytic dependency of cancer cells provides potential therapeutic strategies to preferentially kill cancer cells by inhibiting glycolysis using antiglycolytic agents. The present review emphasizes the most recent research on the implication of glycolytic enzymes, including glucose transporters (GLUTs), hexokinase (HK), phosphofructokinase (PFK), pyruvate kinase (PK), lactate dehydrogenase-A (LDHA), associated signalling pathways and transcription factors, as well as the antiglycolytic agents that target key glycolytic enzymes in breast cancer. The potential activity of glycolytic inhibitors impinges cancer prevalence and cellular resistance to conventional drugs even under worse physiological conditions such as hypoxia. As a single agent or in combination with other chemotherapeutic drugs, it provides the feasibility of new therapeutic modalities against a wide spectrum of human cancers.
Collapse
Affiliation(s)
- Ajeesh Babu Littleflower
- Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, Kerala, 695011, India
| | - Sulfath Thottungal Parambil
- Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, Kerala, 695011, India
| | - Gisha Rose Antony
- Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, Kerala, 695011, India
| | - Lakshmi Subhadradevi
- Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, Kerala, 695011, India.
| |
Collapse
|
15
|
Bani N, Rahmani F, Shakour N, Amerizadeh F, Khalili-Tanha G, Khazaei M, Hassanian SM, Kerachian MA, Abbaszadegan MR, Mojarad M, Hadizadeh F, Ferns GA, Avan A. Wortmannin Inhibits Cell Growth and Induces Apoptosis in Colorectal Cancer Cells by Suppressing the PI3K/AKT Pathway. Anticancer Agents Med Chem 2024; 24:916-927. [PMID: 38584531 DOI: 10.2174/0118715206296355240325113920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/02/2024] [Accepted: 03/13/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) remains a significant contributor to mortality, often exacerbated by metastasis and chemoresistance. Novel therapeutic strategies are imperative to enhance current treatments. The dysregulation of the PI3K/Akt signaling pathway is implicated in CRC progression. This study investigates the therapeutic potential of Wortmannin, combined with 5-fluorouracil (5-FU), to target the PI3K/Akt pathway in CRC. METHODS Anti-migratory and antiproliferative effects were assessed through wound healing and MTT assays. Apoptosis and cell cycle alterations were evaluated using Annexin V/Propidium Iodide Apoptosis Assay. Wortmannin's impact on the oxidant/antioxidant equilibrium was examined via ROS, SOD, CAT, MDA, and T-SH levels. Downstream target genes of the PI3K/AKT pathway were analyzed at mRNA and protein levels using RTPCR and western blot, respectively. RESULTS Wortmannin demonstrated a significant inhibitory effect on cell proliferation, modulating survivin, cyclinD1, PI3K, and p-Akt. The PI3K inhibitor attenuated migratory activity, inducing E-cadherin expression. Combined Wortmannin with 5-FU induced apoptosis, increasing cells in sub-G1 via elevated ROS levels. CONCLUSION This study underscores Wortmannin's potential in inhibiting CRC cell growth and migration through PI3K/Akt pathway modulation. It also highlights its candidacy for further investigation as a promising therapeutic option in colorectal cancer treatment.
Collapse
Affiliation(s)
- Nastaran Bani
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Rahmani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Forouzan Amerizadeh
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Department of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Rathor R, Srivastava S, Suryakumar G. A Comparative Biochemical Study Between L-Carnosine and β-Alanine in Amelioration of Hypobaric Hypoxia-Induced Skeletal Muscle Protein Loss. High Alt Med Biol 2023; 24:302-311. [PMID: 37643283 DOI: 10.1089/ham.2023.0014] [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] [Indexed: 08/31/2023] Open
Abstract
Rathor, Richa, Sukanya Srivastava, and Geetha Suryakumar. A comparative biochemical study between L-carnosine and β-alanine in amelioration of hypobaric hypoxia-induced skeletal muscle protein loss. High Alt Med Biol. 24:302-311, 2023. Background: Carnosine (CAR; β-alanyl-L-histidine), a biologically active dipeptide is known for its unique pH-buffering capacity, metal chelating activity, and antioxidant and antiglycation property. β-Alanine (ALA) is a nonessential amino acid and used to enhance performance and cognitive functions. Hypobaric hypoxia (HH)-induced muscle protein loss is regulated by multifaceted signaling pathways. The present study investigated the beneficial effects of CAR and ALA against HH-associated muscle loss. Methodology: Simulated HH exposure was performed in an animal decompression chamber. Gastric oral administration of CAR (50 mg·kg-1) and ALA (450 mg·kg-1) were given daily for 3 days and at the end of the treatment, hindlimb skeletal muscle tissue was excised for western blot and biochemical assays. Results: Cosupplementation of CAR and ALA alone was able to ameliorate the hypoxia-induced inflammation, oxidative stress (FOXO), ER stress (GRP-78), and atrophic signaling (MuRF-1) in the skeletal muscles. Creatinine phospho kinase activity and apoptosis were also decreased in CAR- and ALA-supplemented rats. However, CAR showed enhanced protection in HH-induced muscle loss as CAR supplementation was able to enhance protein concentration, body weight, and decreased the protein oxidation and ALA administration was not able to restore the same. Conclusions: Hence, the present comprehensive study supports the fact that CAR (50 mg·kg-1) is more beneficial as compared with ALA (450 mg·kg-1) in ameliorating the hypoxia-induced skeletal muscle loss.
Collapse
Affiliation(s)
- Richa Rathor
- Pathophysiology and Disruptive Technologies, Defense Institute of Physiology and Allied Sciences (DIPAS), New Delhi, India
| | - Sukanya Srivastava
- Pathophysiology and Disruptive Technologies, Defense Institute of Physiology and Allied Sciences (DIPAS), New Delhi, India
| | - Geetha Suryakumar
- Pathophysiology and Disruptive Technologies, Defense Institute of Physiology and Allied Sciences (DIPAS), New Delhi, India
| |
Collapse
|
17
|
Zhou Y, Guo Y, Ran M, Shan W, Granchi C, Giovannetti E, Minutolo F, Peters GJ, Tam KY. Combined inhibition of pyruvate dehydrogenase kinase 1 and lactate dehydrogenase a induces metabolic and signaling reprogramming and enhances lung adenocarcinoma cell killing. Cancer Lett 2023; 577:216425. [PMID: 37805163 DOI: 10.1016/j.canlet.2023.216425] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/27/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023]
Abstract
Lung adenocarcinoma (LUAD) is one of the most prevalent and aggressive types of lung cancer. Metabolic reprogramming plays a critical role in the development and progression of LUAD. Pyruvate dehydrogenase kinase 1 (PDK1) and lactate dehydrogenase A (LDHA) are two key enzymes involved in glucose metabolism, whilst their aberrant expressions are often associated with tumorigenesis. Herein, we investigated the anticancer effects of combined inhibition of PDK1 and LDHA in LUAD in vitro and in vivo and its underlying mechanisms of action. The combination of a PDK1 inhibitor, 64, and a LDHA inhibitor, NHI-Glc-2, led to a synergistic growth inhibition in 3 different LUAD cell lines and more than additively suppressed tumor growth in the LUAD xenograft H1975 model. This combination also inhibited cellular migration and colony formation, while it induced a metabolic shift from glycolysis to oxidative phosphorylation (OXPHOS) resulting in mitochondrial depolarization and apoptosis in LUAD cells. These effects were related to modulation of multiple cell signaling pathways, including AMPK, RAS/ERK, and AKT/mTOR. Our findings demonstrate that simultaneous inhibition of multiple glycolytic enzymes (PDK1 and LDHA) is a promising novel therapeutic approach for LUAD.
Collapse
Affiliation(s)
- Yan Zhou
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Yizhen Guo
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Maoxin Ran
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Wenying Shan
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Carlotta Granchi
- Dipartimento di Farmacia, Università di Pisa, 56126, Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Centers, Location VUmc, Cancer Center Amsterdam, 1081, HV Amsterdam, the Netherlands; Fondazione Pisana per La Scienza, Pisa, Italy
| | - Filippo Minutolo
- Dipartimento di Farmacia, Università di Pisa, 56126, Pisa, Italy
| | - Godefridus J Peters
- Department of Biochemistry, Medical University of Gdansk, 80-210, Gdańsk, Poland; Department of Medical Oncology, Amsterdam University Medical Centers, Location VUmc, Cancer Center Amsterdam, 1081, HV Amsterdam, the Netherlands
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau.
| |
Collapse
|
18
|
Zhang M, Zhao Y, Liu X, Ruan X, Wang P, Liu L, Wang D, Dong W, Yang C, Xue Y. Pseudogene MAPK6P4-encoded functional peptide promotes glioblastoma vasculogenic mimicry development. Commun Biol 2023; 6:1059. [PMID: 37853052 PMCID: PMC10584926 DOI: 10.1038/s42003-023-05438-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 10/10/2023] [Indexed: 10/20/2023] Open
Abstract
Glioma is the most common primary malignancy of the central nervous system. Glioblastoma (GBM) has the highest degree of malignancy among the gliomas and the strongest resistance to chemotherapy and radiotherapy. Vasculogenic mimicry (VM) provides tumor cells with a blood supply independent of endothelial cells and greatly restricts the therapeutic effect of anti-angiogenic tumor therapy for glioma patients. Vascular endothelial growth factor receptor 2 (VEGFR2) and vascular endothelial cadherin (VE-cadherin) are currently recognized molecular markers of VM in tumors. In the present study, we show that pseudogene MAPK6P4 deficiency represses VEGFR2 and VE-cadherin protein expression levels, as well as inhibits the proliferation, migration, invasion, and VM development of GBM cells. The MAPK6P4-encoded functional peptide P4-135aa phosphorylates KLF15 at the S238 site, promoting KLF15 protein stability and nuclear entry to promote GBM VM formation. KLF15 was further confirmed as a transcriptional activator of LDHA, where LDHA binds and promotes VEGFR2 and VE-cadherin lactylation, thereby increasing their protein expression. Finally, we used orthotopic and subcutaneous xenografted nude mouse models of GBM to verify the inhibitory effect of the above factors on GBM VM development. In summary, this study may represent new targets for the comprehensive treatment of glioma.
Collapse
Affiliation(s)
- Mengyang Zhang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, PR China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, PR China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, PR China
| | - Yubo Zhao
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
- Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, 110004, PR China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, PR China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
- Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, 110004, PR China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, PR China
| | - Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, PR China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, PR China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, PR China
| | - Ping Wang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, PR China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, PR China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, PR China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, PR China
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, PR China
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, PR China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
- Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, 110004, PR China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, PR China
| | - Weiwei Dong
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
- Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, 110004, PR China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, PR China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
- Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, 110004, PR China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, PR China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, PR China.
- Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, 110122, PR China.
- Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, PR China.
| |
Collapse
|
19
|
El-Sayed NNE, Al-Otaibi TM, Barakat A, Almarhoon ZM, Hassan MZ, Al-Zaben MI, Krayem N, Masand VH, Ben Bacha A. Synthesis and Biological Evaluation of Some New 3-Aryl-2-thioxo-2,3-dihydroquinazolin-4(1 H)-ones and 3-Aryl-2-(benzylthio)quinazolin-4(3 H)-ones as Antioxidants; COX-2, LDHA, α-Glucosidase and α-Amylase Inhibitors; and Anti-Colon Carcinoma and Apoptosis-Inducing Agents. Pharmaceuticals (Basel) 2023; 16:1392. [PMID: 37895863 PMCID: PMC10610505 DOI: 10.3390/ph16101392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Oxidative stress, COX-2, LDHA and hyperglycemia are interlinked contributing pathways in the etiology, progression and metastasis of colon cancer. Additionally, dysregulated apoptosis in cells with genetic alternations leads to their progression in malignant transformation. Therefore, quinazolinones 3a-3h and 5a-5h were synthesized and evaluated as antioxidants, enzymes inhibitors and cytotoxic agents against LoVo and HCT-116 cells. Moreover, the most active cytotoxic derivatives were evaluated as apoptosis inducers. The results indicated that 3a, 3g and 5a were efficiently scavenged DPPH radicals with lowered IC50 values (mM) ranging from 0.165 ± 0.0057 to 0.191 ± 0.0099, as compared to 0.245 ± 0.0257 by BHT. Derivatives 3h, 5a and 5h were recognized as more potent dual inhibitors than quercetin against α-amylase and α-glucosidase, in addition to 3a, 3c, 3f and 5b-5f against α-amylase. Although none of the compounds demonstrated a higher efficiency than the reference inhibitors against COX-2 and LDHA, 3a and 3g were identified as the most active derivatives. Molecular docking studies were used to elucidate the binding affinities and binding interactions between the inhibitors and their target proteins. Compounds 3a and 3f showed cytotoxic activities, with IC50 values (µM) of 294.32 ± 8.41 and 383.5 ± 8.99 (LoVo), as well as 298.05 ± 13.26 and 323.59 ± 3.00 (HCT-116). The cytotoxicity mechanism of 3a and 3f could be attributed to the modulation of apoptosis regulators (Bax and Bcl-2), the activation of intrinsic and extrinsic apoptosis pathways via the upregulation of initiator caspases-8 and -9 as well as executioner caspase-3, and the arrest of LoVo and HCT-116 cell cycles in the G2/M and G1 phases, respectively. Lastly, the physicochemical, medicinal chemistry and ADMET properties of all compounds were predicted.
Collapse
Affiliation(s)
| | - Taghreed M. Al-Otaibi
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (T.M.A.-O.); (A.B.); (M.I.A.-Z.)
| | - Assem Barakat
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (T.M.A.-O.); (A.B.); (M.I.A.-Z.)
| | - Zainab M. Almarhoon
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (T.M.A.-O.); (A.B.); (M.I.A.-Z.)
| | - Mohd. Zaheen Hassan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Maha I. Al-Zaben
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (T.M.A.-O.); (A.B.); (M.I.A.-Z.)
| | - Najeh Krayem
- Laboratoire de Biochimie et de Génie Enzymatique des Lipases, ENIS, Université de Sfax, Route de Soukra 3038, Sfax BP 1173, Tunisia;
| | - Vijay H. Masand
- Department of Chemistry, Vidya Bharati College, Camp, Amravati, Maharashtra 444602, India;
| | - Abir Ben Bacha
- Biochemistry Department, College of Sciences, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia;
| |
Collapse
|
20
|
Zhang B, Sun J, Guan H, Guo H, Huang B, Chen X, Chen F, Yuan Q. Integrated single-cell and bulk RNA sequencing revealed the molecular characteristics and prognostic roles of neutrophils in pancreatic cancer. Aging (Albany NY) 2023; 15:9718-9742. [PMID: 37728418 PMCID: PMC10564426 DOI: 10.18632/aging.205044] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023]
Abstract
Pancreatic cancer, one of the most prevalent tumors of the digestive system, has a dismal prognosis. Cancer of the pancreas is distinguished by an inflammatory tumor microenvironment rich in fibroblasts and different immune cells. Neutrophils are important immune cells that infiltrate the microenvironment of pancreatic cancer tumors. The purpose of this work was to examine the complex mechanism by which neutrophils influence the carcinogenesis and development of pancreatic cancer and to construct a survival prediction model based on neutrophil marker genes. We incorporated the GSE111672 dataset, comprising RNA expression data from 27,000 cells obtained from 3 patients with PC, and conducted single-cell data analysis. Thorough investigation of pancreatic cancer single-cell RNA sequencing data found 350 neutrophil marker genes. Using The Cancer Genome Atlas (TCGA), GSE28735, GSE62452, GSE57495, and GSE85916 datasets to gather pancreatic cancer tissue transcriptome data, and consistent clustering was used to identify two categories for analyzing the influence of neutrophils on pancreatic cancer. Using the Random Forest algorithm and Cox regression analysis, a survival prediction model for pancreatic cancer was developed, the model showed independent performance for survival prognosis, clinic pathological features, immune infiltration, and drug sensitivity. Multivariate Cox analysis findings revealed that the risk scores derived from predictive models is independent prognostic markers for pancreatic patients. In conclusion, based on neutrophil marker genes, this research created a molecular typing and prognostic grading system for pancreatic cancer, this system was very accurate in predicting the prognosis, tumor immune microenvironment status, and pharmacological treatment responsiveness of pancreatic cancer patients.
Collapse
Affiliation(s)
- Biao Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jiaao Sun
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hewen Guan
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hui Guo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bingqian Huang
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Xu Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Feng Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qihang Yuan
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
21
|
Sharma D, Singh M, Joshi J, Garg M, Chaudhary V, Blankenberg D, Chandna S, Kumar V, Rani R. Design and Synthesis of Thiazole Scaffold-Based Small Molecules as Anticancer Agents Targeting the Human Lactate Dehydrogenase A Enzyme. ACS OMEGA 2023; 8:17552-17562. [PMID: 37251149 PMCID: PMC10210175 DOI: 10.1021/acsomega.2c07569] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 04/24/2023] [Indexed: 05/31/2023]
Abstract
A new series of thiazole central scaffold-based small molecules of hLDHA inhibitors were designed using an in silico approach. Molecular docking analysis of designed molecules with hLDHA (PDB ID: 1I10) demonstrates that Ala 29, Val 30, Arg 98, Gln 99, Gly 96, and Thr 94 possessed strong interaction with the compounds. Compounds 8a, 8b, and 8d showed good binding affinity (-8.1 to -8.8 kcal/mol), whereas an additional interaction of NO2 at the ortho position in compounds 8c with Gln 99 through hydrogen bonding enhanced the affinity to -9.8 kcal/mol. Selected high-scored compounds were synthesized and screened for hLDHA inhibitory activities and in vitro anticancer activity in six cancer cell lines. Biochemical enzyme inhibition assays showed the highest hLDHA inhibitory activity observed with compounds 8b, 8c, and 8l. Compounds 8b, 8c, 8j, 8l, and 8m depicted significant anticancer activities, exhibiting IC50 values in the range of 1.65-8.60 μM in HeLa and SiHa cervical cancer cell lines. Compounds 8j and 8m exhibited notable anticancer activity with IC50 values of 7.90 and 5.15 μM, respectively, in liver cancer cells (HepG2). Interestingly, compounds 8j and 8m did not induce noticeable toxicity in the human embryonic kidney cells (HEK293). Insilico absorption, distribution, metabolism, and excretion profiling demonstrates that the compounds possess drug-likeness, and results may pave the way for the development of novel thiazole-based biologically active small molecules for therapeutics.
Collapse
Affiliation(s)
- Dolly Sharma
- Amity
Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
- Amity
Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida 201303, Uttar
Pradesh, India
| | - Mamta Singh
- Amity
Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida 201303, Uttar
Pradesh, India
| | - Jayadev Joshi
- Genomic
Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Manoj Garg
- Amity
Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida 201303, Uttar
Pradesh, India
| | | | - Daniel Blankenberg
- Genomic
Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, United States
| | - Sudhir Chandna
- Institute
of Nuclear Medicine & Allied Science, Defense Research Development Organization, Delhi 110054, India
| | - Vinit Kumar
- Amity
Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida 201303, Uttar
Pradesh, India
| | - Reshma Rani
- Drug Discovery,
Jubilant Biosys, Knowledge
Park-2, Greater Noida 201306, India
| |
Collapse
|
22
|
Goenka A, Khan F, Verma B, Sinha P, Dmello CC, Jogalekar MP, Gangadaran P, Ahn B. Tumor microenvironment signaling and therapeutics in cancer progression. Cancer Commun (Lond) 2023; 43:525-561. [PMID: 37005490 PMCID: PMC10174093 DOI: 10.1002/cac2.12416] [Citation(s) in RCA: 109] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/22/2023] [Accepted: 03/20/2023] [Indexed: 04/04/2023] Open
Abstract
Tumor development and metastasis are facilitated by the complex interactions between cancer cells and their microenvironment, which comprises stromal cells and extracellular matrix (ECM) components, among other factors. Stromal cells can adopt new phenotypes to promote tumor cell invasion. A deep understanding of the signaling pathways involved in cell-to-cell and cell-to-ECM interactions is needed to design effective intervention strategies that might interrupt these interactions. In this review, we describe the tumor microenvironment (TME) components and associated therapeutics. We discuss the clinical advances in the prevalent and newly discovered signaling pathways in the TME, the immune checkpoints and immunosuppressive chemokines, and currently used inhibitors targeting these pathways. These include both intrinsic and non-autonomous tumor cell signaling pathways in the TME: protein kinase C (PKC) signaling, Notch, and transforming growth factor (TGF-β) signaling, Endoplasmic Reticulum (ER) stress response, lactate signaling, Metabolic reprogramming, cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) and Siglec signaling pathways. We also discuss the recent advances in Programmed Cell Death Protein 1 (PD-1), Cytotoxic T-Lymphocyte Associated Protein 4 (CTLA4), T-cell immunoglobulin mucin-3 (TIM-3) and Lymphocyte Activating Gene 3 (LAG3) immune checkpoint inhibitors along with the C-C chemokine receptor 4 (CCR4)- C-C class chemokines 22 (CCL22)/ and 17 (CCL17), C-C chemokine receptor type 2 (CCR2)- chemokine (C-C motif) ligand 2 (CCL2), C-C chemokine receptor type 5 (CCR5)- chemokine (C-C motif) ligand 3 (CCL3) chemokine signaling axis in the TME. In addition, this review provides a holistic understanding of the TME as we discuss the three-dimensional and microfluidic models of the TME, which are believed to recapitulate the original characteristics of the patient tumor and hence may be used as a platform to study new mechanisms and screen for various anti-cancer therapies. We further discuss the systemic influences of gut microbiota in TME reprogramming and treatment response. Overall, this review provides a comprehensive analysis of the diverse and most critical signaling pathways in the TME, highlighting the associated newest and critical preclinical and clinical studies along with their underlying biology. We highlight the importance of the most recent technologies of microfluidics and lab-on-chip models for TME research and also present an overview of extrinsic factors, such as the inhabitant human microbiome, which have the potential to modulate TME biology and drug responses.
Collapse
Affiliation(s)
- Anshika Goenka
- The Ken & Ruth Davee Department of NeurologyThe Robert H. Lurie Comprehensive Cancer CenterNorthwestern University Feinberg School of MedicineChicago, 60611ILUSA
| | - Fatima Khan
- Department of Neurological SurgeryFeinberg School of MedicineNorthwestern UniversityChicago, 60611ILUSA
| | - Bhupender Verma
- Department of OphthalmologySchepens Eye Research InstituteMassachusetts Eye and Ear InfirmaryHarvard Medical SchoolBoston, 02114MAUSA
| | - Priyanka Sinha
- Department of NeurologyMassGeneral Institute for Neurodegenerative DiseaseMassachusetts General Hospital, Harvard Medical SchoolCharlestown, 02129MAUSA
| | - Crismita C. Dmello
- Department of Neurological SurgeryFeinberg School of MedicineNorthwestern UniversityChicago, 60611ILUSA
| | - Manasi P. Jogalekar
- Helen Diller Family Comprehensive Cancer CenterUniversity of California San FranciscoSan Francisco, 94143CAUSA
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future TalentsDepartment of Biomedical Science, School of MedicineKyungpook National UniversityDaegu, 41944South Korea
- Department of Nuclear MedicineSchool of Medicine, Kyungpook National University, Kyungpook National University HospitalDaegu, 41944South Korea
| | - Byeong‐Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future TalentsDepartment of Biomedical Science, School of MedicineKyungpook National UniversityDaegu, 41944South Korea
- Department of Nuclear MedicineSchool of Medicine, Kyungpook National University, Kyungpook National University HospitalDaegu, 41944South Korea
| |
Collapse
|
23
|
Bhoopathi P, Mannangatti P, Das SK, Fisher PB, Emdad L. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy. Adv Cancer Res 2023; 159:285-341. [PMID: 37268399 DOI: 10.1016/bs.acr.2023.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a prominent cause of cancer deaths worldwide, is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. More than 90% of patients with PDAC die within a year of being diagnosed. PDAC is increasing at a rate of 0.5-1.0% per year, and it is expected to be the second leading cause of cancer-related mortality by 2030. The resistance of tumor cells to chemotherapeutic drugs, which can be innate or acquired, is the primary factor contributing to the ineffectiveness of cancer treatments. Although many PDAC patients initially responds to standard of care (SOC) drugs they soon develop resistance caused partly by the substantial cellular heterogeneity seen in PDAC tissue and the tumor microenvironment (TME), which are considered key factors contributing to resistance to therapy. A deeper understanding of molecular mechanisms involved in PDAC progression and metastasis development, and the interplay of the TME in all these processes is essential to better comprehend the etiology and pathobiology of chemoresistance observed in PDAC. Recent research has recognized new therapeutic targets ushering in the development of innovative combinatorial therapies as well as enhancing our comprehension of several different cell death pathways. These approaches facilitate the lowering of the therapeutic threshold; however, the possibility of subsequent resistance development still remains a key issue and concern. Discoveries, that can target PDAC resistance, either alone or in combination, have the potential to serve as the foundation for future treatments that are effective without posing undue health risks. In this chapter, we discuss potential causes of PDAC chemoresistance and approaches for combating chemoresistance by targeting different pathways and different cellular functions associated with and mediating resistance.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
24
|
Syamprasad NP, Jain S, Rajdev B, Prasad N, Kallipalli R, Naidu VGM. Aldose reductase and cancer metabolism: The master regulator in the limelight. Biochem Pharmacol 2023; 211:115528. [PMID: 37011733 DOI: 10.1016/j.bcp.2023.115528] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
It is strongly established that metabolic reprogramming mediates the initiation, progression, and metastasis of a variety of cancers. However, there is no common biomarker identified to link the dysregulated metabolism and cancer progression. Recent studies strongly advise the involvement of aldose reductase (AR) in cancer metabolism. AR-mediated glucose metabolism creates a Warburg-like effect and an acidic tumour microenvironment in cancer cells. Moreover, AR overexpression is associated with the impairment of mitochondria and the accumulation of free fatty acids in cancer cells. Further, AR-mediated reduction of lipid aldehydes and chemotherapeutics are involved in the activation of factors promoting proliferation and chemo-resistance. In this review, we have delineated the possible mechanisms by which AR modulates cellular metabolism for cancer proliferation and survival. An in-depth understanding of cancer metabolism and the role of AR might lead to the use of AR inhibitors as metabolic modulating agents for the therapy of cancer.
Collapse
Affiliation(s)
- N P Syamprasad
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Siddhi Jain
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Bishal Rajdev
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Neethu Prasad
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Ravindra Kallipalli
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - V G M Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India.
| |
Collapse
|
25
|
The Role of Reprogrammed Glucose Metabolism in Cancer. Metabolites 2023; 13:metabo13030345. [PMID: 36984785 PMCID: PMC10051753 DOI: 10.3390/metabo13030345] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Cancer cells reprogram their metabolism to meet biosynthetic needs and to adapt to various microenvironments. Accelerated glycolysis offers proliferative benefits for malignant cells by generating glycolytic products that move into branched pathways to synthesize proteins, fatty acids, nucleotides, and lipids. Notably, reprogrammed glucose metabolism and its associated events support the hallmark features of cancer such as sustained cell proliferation, hijacked apoptosis, invasion, metastasis, and angiogenesis. Overproduced enzymes involved in the committed steps of glycolysis (hexokinase, phosphofructokinase-1, and pyruvate kinase) are promising pharmacological targets for cancer therapeutics. In this review, we summarize the role of reprogrammed glucose metabolism in cancer cells and how it can be manipulated for anti-cancer strategies.
Collapse
|
26
|
Phukhum P, Phetcharaburanin J, Chaleekarn K, Kittirat Y, Kulthawatsiri T, Namwat N, Loilome W, Khuntikeo N, Titapun A, Wangwiwatsin A, Khampitak T, Suksawat M, Klanrit P. The impact of hypoxia and oxidative stress on proteo-metabolomic alterations of 3D cholangiocarcinoma models. Sci Rep 2023; 13:3072. [PMID: 36810897 PMCID: PMC9944917 DOI: 10.1038/s41598-023-30204-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
The three-dimensional multicellular spheroid (3D MCS) model has been employed in cholangiocarcinoma research as it generates 3D architecture and includes more physiological relevance with the multicellular arrangement. However, it is also essential to explain the molecular signature in this microenvironment and its structural complexity. The results indicated that poorly differentiated CCA cell lines were unable to form 3D MCS due to the lack of cell adhesion molecules with more mesenchymal marker expression. The well-differentiated CCA and cholangiocyte cell lines were able to develop 3D MCSs with round shapes, smooth perimeter, and cell adhesion molecules that led to the hypoxic and oxidative microenvironment detected. For MMNK-1, KKU-213C, and KKU-213A MCSs, the proteo-metabolomic analysis showed proteins and metabolic products altered compared to 2D cultures, including cell-cell adhesion molecules, energy metabolism-related enzymes and metabolites, and oxidative-related metabolites. Therefore, the 3D MCSs provide different physiological states with different phenotypic signatures compared to 2D cultures. Considering the 3D model mimics more physiological relevance, it might lead to an alternate biochemical pathway, targeting to improve drug sensitivity for CCA treatment.
Collapse
Affiliation(s)
- Pimpawadee Phukhum
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Jutarop Phetcharaburanin
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Kwuanjira Chaleekarn
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Yingpinyapat Kittirat
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Thanaporn Kulthawatsiri
- grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Nisana Namwat
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Watcharin Loilome
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Narong Khuntikeo
- grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Attapol Titapun
- grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Arporn Wangwiwatsin
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Tueanjit Khampitak
- grid.9786.00000 0004 0470 0856Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Manida Suksawat
- grid.9786.00000 0004 0470 0856Khon Kaen University Phenome Centre, Khon Kaen, 40002 Thailand ,grid.9786.00000 0004 0470 0856Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002 Thailand
| | - Poramate Klanrit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand. .,Khon Kaen University Phenome Centre, Khon Kaen, 40002, Thailand. .,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
27
|
Sharma H, Sharma P, Urquiza U, Chastain LR, Ihnat MA. Exploration of a Large Virtual Chemical Space: Identification of Potent Inhibitors of Lactate Dehydrogenase-A against Pancreatic Cancer. J Chem Inf Model 2023; 63:1028-1043. [PMID: 36646658 PMCID: PMC9930117 DOI: 10.1021/acs.jcim.2c01544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
It is imperative to explore the gigantic available chemical space to identify new scaffolds for drug lead discovery. Identifying potent hits from virtual screening of large chemical databases is challenging and computationally demanding. Rather than the traditional two-dimensional (2D)/three-dimensional (3D) approaches on smaller chemical libraries of a few hundred thousand compounds, we screened a ZINC library of 15 million compounds using multiple computational methods. Here, we present the successful application of a virtual screening methodology that identifies several chemotypes as starting hits against lactate dehydrogenase-A (LDHA). From 29 compounds identified from virtual screening, 17 (58%) showed IC50 values < 63 μM, two showed single-digit micromolar inhibition, and the most potent hit compound had IC50 down to 117 nM. We enriched the database and employed an ensemble approach by combining 2D fingerprint similarity searches, pharmacophore modeling, molecular docking, and molecular dynamics. WaterMap calculations were carried out to explore the thermodynamics of surface water molecules and gain insights into the LDHA binding pocket. The present work has led to the discovery of two new chemical classes, including compounds with a succinic acid monoamide moiety or a hydroxy pyrimidinone ring system. Selected hits block lactate production in cells and inhibit pancreatic cancer cell lines with cytotoxicity IC50 down to 12.26 μM against MIAPaCa-2 cells and 14.64 μM against PANC-1, which, under normoxic conditions, is already comparable or more potent than most currently available known LDHA inhibitors.
Collapse
Affiliation(s)
- Horrick Sharma
- Department
of Pharmaceutical Sciences, College of Pharmacy, Southwestern Oklahoma State University, Weatherford, Oklahoma73096, United States,, . Phone: (+1)580-774-3064. Fax: (+1)(580)-774-7020
| | - Pragya Sharma
- Department
of Biological Sciences, Southwestern Oklahoma
State University, Weatherford, Oklahoma73096, United States
| | - Uzziah Urquiza
- Department
of Biological Sciences, Southwestern Oklahoma
State University, Weatherford, Oklahoma73096, United States
| | - Lerin R. Chastain
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma73117, United States
| | - Michael A. Ihnat
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma73117, United States
| |
Collapse
|
28
|
Liu C, Li C, Liu Y. The role of metabolic reprogramming in pancreatic cancer chemoresistance. Front Pharmacol 2023; 13:1108776. [PMID: 36699061 PMCID: PMC9868425 DOI: 10.3389/fphar.2022.1108776] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/28/2022] [Indexed: 01/10/2023] Open
Abstract
Pancreatic cancer is characterized by hidden onset, high malignancy, and early metastasis. Although a few cases meet the surgical indications, chemotherapy remains the primary treatment, and the resulting chemoresistance has become an urgent clinical problem that needs to be solved. In recent years, the importance of metabolic reprogramming as one of the hallmarks of cancers in tumorigenesis has been validated. Metabolic reprogramming involves glucose, lipid, and amino acid metabolism and interacts with oncogenes to affect the expression of key enzymes and signaling pathways, modifying the tumor microenvironment and contributing to the occurrence of drug tolerance. Meanwhile, the mitochondria are hubs of the three major nutrients and energy metabolisms, which are also involved in the development of drug resistance. In this review, we summarized the characteristic changes in metabolism during the progression of pancreatic cancer and their impact on chemoresistance, outlined the role of the mitochondria, and summarized current studies on metabolic inhibitors.
Collapse
|
29
|
Li Z, Wang Q, Huang X, Yang M, Zhou S, Li Z, Fang Z, Tang Y, Chen Q, Hou H, Li L, Fei F, Wang Q, Wu Y, Gong A. Lactate in the tumor microenvironment: A rising star for targeted tumor therapy. Front Nutr 2023; 10:1113739. [PMID: 36875841 PMCID: PMC9978120 DOI: 10.3389/fnut.2023.1113739] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Metabolic reprogramming is one of fourteen hallmarks of tumor cells, among which aerobic glycolysis, often known as the "Warburg effect," is essential to the fast proliferation and aggressive metastasis of tumor cells. Lactate, on the other hand, as a ubiquitous molecule in the tumor microenvironment (TME), is generated primarily by tumor cells undergoing glycolysis. To prevent intracellular acidification, malignant cells often remove lactate along with H+, yet the acidification of TME is inevitable. Not only does the highly concentrated lactate within the TME serve as a substrate to supply energy to the malignant cells, but it also works as a signal to activate multiple pathways that enhance tumor metastasis and invasion, intratumoral angiogenesis, as well as immune escape. In this review, we aim to discuss the latest findings on lactate metabolism in tumor cells, particularly the capacity of extracellular lactate to influence cells in the tumor microenvironment. In addition, we examine current treatment techniques employing existing medications that target and interfere with lactate generation and transport in cancer therapy. New research shows that targeting lactate metabolism, lactate-regulated cells, and lactate action pathways are viable cancer therapy strategies.
Collapse
Affiliation(s)
- Zhangzuo Li
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.,Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Mengting Yang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shujing Zhou
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zhengrui Li
- School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhengzou Fang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yidan Tang
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Qian Chen
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hanjin Hou
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Li Li
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Fei Fei
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiaowei Wang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuqing Wu
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Aihua Gong
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.,Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
30
|
Abstract
C-Myc overexpression is a common finding in pancreatic cancer and predicts the aggressive behavior of cancer cells. It binds to the promoter of different genes, thereby regulating their transcription. C-Myc is downstream of KRAS and interacts with several oncogenic and proliferative pathways in pancreatic cancer. C-Myc enhances aerobic glycolysis in cancer cells and regulates glutamate biosynthesis from glutamine. It provides enough energy for cancer cells' metabolism and sufficient substrate for the synthesis of organic molecules. C-Myc overexpression is associated with chemoresistance, intra-tumor angiogenesis, epithelial-mesenchymal transition (EMT), and metastasis in pancreatic cancer. Despite its title, c-Myc is not "undruggable" and recent studies unveiled that it can be targeted, directly or indirectly. Small molecules that accelerate c-Myc ubiquitination and degradation have been effective in preclinical studies. Small molecules that hinder c-Myc-MAX heterodimerization or c-Myc/MAX/DNA complex formation can functionally inhibit c-Myc. In addition, c-Myc can be targeted through transcriptional, post-transcriptional, and translational modifications.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
31
|
Priego-Hernández VD, Arizmendi-Izazaga A, Soto-Flores DG, Santiago-Ramón N, Feria-Valadez MD, Navarro-Tito N, Jiménez-Wences H, Martínez-Carrillo DN, Salmerón-Bárcenas EG, Leyva-Vázquez MA, Illades-Aguiar B, Alarcón-Romero LDC, Ortiz-Ortiz J. Expression of HIF-1α and Genes Involved in Glucose Metabolism Is Increased in Cervical Cancer and HPV-16-Positive Cell Lines. Pathogens 2022; 12:pathogens12010033. [PMID: 36678382 PMCID: PMC9865746 DOI: 10.3390/pathogens12010033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Cervical cancer (CC) is the most common cancer in women in the lower genital tract. The main risk factor for developing CC is persistent infection with HPV 16. The E6 and E7 oncoproteins of HPV 16 have been related to metabolic reprogramming in cancer through the regulation of the expression and stability of HIF-1α and consequently of the expression of its target genes, such as HIF1A (HIF-1α), SLC2A1 (GLUT1), LDHA, CA9 (CAIX), SLC16A3 (MCT4), and BSG (Basigin or CD147), which are involved in glucose metabolism. This work aimed to evaluate the expression of HIF-1α, GLUT1, LDHA, CAIX, MCT4, and Basigin in patient samples and CC cell lines. To evaluate the expression level of HIF1A, SLC2A1, LDHA, CA9, SLC16A3, and BSG genes in tissue from patients with CC and normal tissue, the TCGA dataset was used. To evaluate the expression level of these genes by RT-qPCR in CC cell lines, HPV-negative (C-33A) and HPV-16-positive (SiHa and Ca Ski) cell lines were used. Increased expression of HIF1A, SLC2A1, LDHA, SLC16A3, and BSG was found in Ca Ski and CA9 in SiHa compared to C-33A. Similar results were observed in CC tissues compared to normal tissue obtained by bioinformatics analysis. In conclusion, the expression of HIF-1α, GLUT1, LDHA, CAIX, MCT4, and BSG genes is increased in CC and HPV-16-positive cell lines.
Collapse
Affiliation(s)
- Víctor D. Priego-Hernández
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Adán Arizmendi-Izazaga
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Diana G. Soto-Flores
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Norma Santiago-Ramón
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Milagros D. Feria-Valadez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Hilda Jiménez-Wences
- Laboratorio de Investigación Clínica, Facultad de Ciencias, Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Dinorah N. Martínez-Carrillo
- Laboratorio de Investigación Clínica, Facultad de Ciencias, Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Eric G. Salmerón-Bárcenas
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico
| | - Marco A. Leyva-Vázquez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Luz del C. Alarcón-Romero
- Laboratorio de Investigación en Citopatología e Histoquímica de la Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Julio Ortiz-Ortiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
- Correspondence: ; Tel.: +52-747-471-0901
| |
Collapse
|
32
|
Dong M, Cao L, Cui R, Xie Y. The connection between innervation and metabolic rearrangements in pancreatic cancer through serine. Front Oncol 2022; 12:992927. [PMID: 36582785 PMCID: PMC9793709 DOI: 10.3389/fonc.2022.992927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/31/2022] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a kind of aggressive tumor famous for its lethality and intractability, and pancreatic ductal adenocarcinoma is the most common type. Patients with pancreatic cancer often suffer a rapid loss of weight and abdominal neuropathic pain in their early stages and then go through cachexia in the advanced stage. These features of patients are considered to be related to metabolic reprogramming of pancreatic cancer and abundant nerve innervation responsible for the pain. With increasing literature certifying the relationship between nerves and pancreatic ductal adenocarcinoma (PDAC), more evidence point out that innervation's role is not limited to neuropathic pain but explore its anti/pro-tumor functions in PDAC, especially the neural-metabolic crosstalks. This review aims to unite pancreatic cancer's innervation and metabolic rearrangements with terminated published articles. Hopefully, this article could explore the pathogenesis of PDAC and further promote promising detecting or therapeutic measurements for PDAC according to the lavish innervation in PDAC.
Collapse
Affiliation(s)
- Mengmeng Dong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Lidong Cao
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China,Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, Second Hospital of Jilin University, Changchun, China,Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial Peoples Hospital, Hangzhou, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China,*Correspondence: Ranji Cui, ; Yingjun Xie,
| | - Yingjun Xie
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China,Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, Second Hospital of Jilin University, Changchun, China,*Correspondence: Ranji Cui, ; Yingjun Xie,
| |
Collapse
|
33
|
Wang X, He X, Liu C, Zhao W, Yuan X, Li R. Progress and perspectives of platinum nanozyme in cancer therapy. Front Chem 2022; 10:1092747. [DOI: 10.3389/fchem.2022.1092747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
Malignant tumors, one of the worst-case scenarios within human health problems, are now posing an increasing threat to the well-being of the global population. At present, the treatment of malignant tumors mainly includes surgery, radiotherapy, chemotherapy, immunotherapy, etc. Radiotherapy and chemotherapy are often applied to inoperable tumors, and some other tumors after surgery as important adjuvant therapies. Nonetheless, both radiotherapy and chemotherapy have a series of side effects, such as radiation-induced lung injury, and chemotherapy-induced bone marrow suppression. In addition, the positioning accuracy of radiotherapy and chemotherapy is not assured and satisfactory, and the possibility of tumor cells not being sensitive to radiation and chemotherapy drugs is also problematic. Nanozymes are nanomaterials that display natural enzyme activities, and their applications to tumor therapy have made great progress recently. The most studied one, platinum nanozyme, has been shown to possess a significant correlation with radiotherapy sensitization of tumors as well as photodynamic therapy. However, there are still several issues that limited the usage of platinum-based nanozymes in vivo. In this review, we briefly summarize the representative studies regarding platinum nanozymes, and especially emphasize on the current challenges and the directions of future development for platinum nanozymes therapy.
Collapse
|
34
|
Targeting hypoxia-related metabolism molecules: How to improve tumour immune and clinical treatment? Biomed Pharmacother 2022; 156:113917. [DOI: 10.1016/j.biopha.2022.113917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/20/2022] Open
|
35
|
Sharma D, Singh M, Rani R. Role of LDH in tumor glycolysis: Regulation of LDHA by small molecules for cancer therapeutics. Semin Cancer Biol 2022; 87:184-195. [PMID: 36371026 DOI: 10.1016/j.semcancer.2022.11.007] [Citation(s) in RCA: 151] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/11/2022] [Accepted: 11/08/2022] [Indexed: 11/10/2022]
Abstract
Lactate dehydrogenase (LDH) is one of the crucial enzymes in aerobic glycolysis, catalyzing the last step of glycolysis, i.e. the conversion of pyruvate to lactate. Most cancer cells are characterized by an enhanced rate of tumor glycolysis to ensure the energy demand of fast-growing cancer cells leading to increased lactate production. Excess lactate creates extracellular acidosis which facilitates invasion, angiogenesis, and metastasis and affects the immune response. Lactate shuttle and lactate symbiosis is established in cancer cells, which may further increase the poor prognosis. Several genetic and phenotypic studies established the potential role of lactate dehydrogenase A (LDHA) or LDH5, the one homo-tetramer of subunit A, in cancer development and metastasis. The LDHA is considered a viable target for drug design and discovery. Several small molecules have been discovered to date exhibiting significant LDHA inhibitory activities and anticancer activities, therefore the starvation of cancer cells by targeting tumor glycolysis through LDHA inhibition with improved selectivity can generate alternative anticancer therapeutics. This review provides an overview of the role of LDHA in metabolic reprogramming and its association with proto-oncogenes and oncogenes. This review also aims to deliver an update on significant LDHA inhibitors with anticancer properties and future direction in this area.
Collapse
Affiliation(s)
- Dolly Sharma
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Mamta Singh
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Reshma Rani
- Jubilant Biosys, Drug Discovery chemistry, Greater Noida, 201310 Uttar Pradesh, India.
| |
Collapse
|
36
|
Comandatore A, Franczak M, Smolenski RT, Morelli L, Peters GJ, Giovannetti E. Lactate Dehydrogenase and its clinical significance in pancreatic and thoracic cancers. Semin Cancer Biol 2022; 86:93-100. [PMID: 36096316 DOI: 10.1016/j.semcancer.2022.09.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 10/31/2022]
Abstract
The energy metabolism of tumor cells is considered one of the hallmarks of cancer because it is different from normal cells and mainly consists of aerobic glycolysis, fatty acid oxidation, and glutaminolysis. It is about one hundred years ago since Warburg observed that cancer cells prefer aerobic glycolysis even in normoxic conditions, favoring their high proliferation rate. A pivotal enzyme driving this phenomenon is lactate dehydrogenase (LDH), and this review describes prognostic and therapeutic opportunities associated with this enzyme, focussing on tumors with limited therapeutic strategies and life expectancy (i.e., pancreatic and thoracic cancers). Expression levels of LDH-A in pancreatic cancer tissues correlate with clinicopathological features: LDH-A is overexpressed during pancreatic carcinogenesis and showed significantly higher expression in more aggressive tumors. Similarly, LDH levels are a marker of negative prognosis in patients with both adenocarcinoma or squamous cell lung carcinoma, as well as in malignant pleural mesothelioma. Additionally, serum LDH levels may play a key role in the clinical management of these diseases because they are associated with tissue damage induced by tumor burden. Lastly, we discuss the promising results of strategies targeting LDH as a treatment strategy, reporting recent preclinical and translational studies supporting the use of LDH-inhibitors in combinations with current/novel chemotherapeutics that can synergistically target the oxygenated cells present in the tumor.
Collapse
Affiliation(s)
- Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy; Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (Amsterdam UMC), Vrije Universiteit Amsterdam, the Netherlands
| | - Marika Franczak
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (Amsterdam UMC), Vrije Universiteit Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | | | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (Amsterdam UMC), Vrije Universiteit Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers (Amsterdam UMC), Vrije Universiteit Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, Pisa, Italy.
| |
Collapse
|
37
|
Lin J, Liu G, Chen L, Kwok HF, Lin Y. Targeting lactate-related cell cycle activities for cancer therapy. Semin Cancer Biol 2022; 86:1231-1243. [PMID: 36328311 DOI: 10.1016/j.semcancer.2022.10.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Lactate has long been considered as a metabolic by-product of aerobic glycolysis for cancer. However, more and more studies have shown that lactate can regulate cancer progression via multiple mechanisms such as cell cycle regulation, immune suppression, energy metabolism and so on. A recent discovery of lactylation attracted a lot of attention and is already a hot topic in the cancer field. In this review, we summarized the latest functions of lactate and its underlying mechanisms in cancer. We also included our analysis of protein lactylation in different rat organs and compared them with other published lactylation data. The unresolved challenges in this field were discussed, and the potential application of these new discoveries of lactate-related cell cycle activities for cancer target therapy was speculated.
Collapse
Affiliation(s)
- Jia Lin
- Central Laboratory at the Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Collaborative Innovation Center for Rehabilitation Technology, the Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, China; Collaborative Innovation Center for Rehabilitation Technology, the Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, China
| | - Geng Liu
- Laboratory of Cancer Biology, Department of Oncology, University of Oxford, UK
| | - Lidian Chen
- Collaborative Innovation Center for Rehabilitation Technology, the Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, China.
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR; MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
| | - Yao Lin
- Central Laboratory at the Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Collaborative Innovation Center for Rehabilitation Technology, the Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, China; Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, China.
| |
Collapse
|
38
|
Abstract
High serum lactate dehydrogenase (LDH) levels are typically associated with a poor prognosis in many cancer types. Even the most effective drugs, which have radically improved outcomes in patients with melanoma over the past decade, provide only marginal benefit to those with high serum LDH levels. When viewed separately from the oncological, biochemical, biological and immunological perspectives, serum LDH is often interpreted in very different ways. Oncologists usually see high serum LDH only as a robust biomarker of a poor prognosis, and biochemists are aware of the complexity of the various LDH isoforms and of their key roles in cancer metabolism, whereas LDH is typically considered to be oncogenic and/or immunosuppressive by cancer biologists and immunologists. Integrating these various viewpoints shows that the regulation of the five LDH isoforms, and their enzymatic and non-enzymatic functions is closely related to key oncological processes. In this Review, we highlight that serum LDH is far more than a simple indicator of tumour burden; it is a complex biomarker associated with the activation of several oncogenic signalling pathways as well as with the metabolic activity, invasiveness and immunogenicity of many tumours, and constitutes an extremely attractive target for cancer therapy.
Collapse
|
39
|
Targeting Glucose Metabolism Enzymes in Cancer Treatment: Current and Emerging Strategies. Cancers (Basel) 2022; 14:cancers14194568. [PMID: 36230492 PMCID: PMC9559313 DOI: 10.3390/cancers14194568] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Reprogramming of glucose metabolism is a hallmark of cancer and can be targeted by therapeutic agents. Some metabolism regulators, such as ivosidenib and enasidenib, have been approved for cancer treatment. Currently, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Furthermore, some natural products have shown efficacy in killing tumor cells by regulating glucose metabolism, offering novel therapeutic opportunities in cancer. However, most of them have failed to be translated into clinical applications due to low selectivity, high toxicity, and side effects. Recent studies suggest that combining glucose metabolism modulators with chemotherapeutic drugs, immunotherapeutic drugs, and other conventional anticancer drugs may be a future direction for cancer treatment. Abstract Reprogramming of glucose metabolism provides sufficient energy and raw materials for the proliferation, metastasis, and immune escape of cancer cells, which is enabled by glucose metabolism-related enzymes that are abundantly expressed in a broad range of cancers. Therefore, targeting glucose metabolism enzymes has emerged as a promising strategy for anticancer drug development. Although several glucose metabolism modulators have been approved for cancer treatment in recent years, some limitations exist, such as a short half-life, poor solubility, and numerous adverse effects. With the rapid development of medicinal chemicals, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Additionally, several studies have found that some natural products can suppress cancer progression by regulating glucose metabolism enzymes. In this review, we summarize the mechanisms underlying the reprogramming of glucose metabolism and present enzymes that could serve as therapeutic targets. In addition, we systematically review the existing drugs targeting glucose metabolism enzymes, including small-molecule modulators and natural products. Finally, the opportunities and challenges for glucose metabolism enzyme-targeted anticancer drugs are also discussed. In conclusion, combining glucose metabolism modulators with conventional anticancer drugs may be a promising cancer treatment strategy.
Collapse
|
40
|
Wang C, Xu R, Song J, Chen Y, Yin X, Ruze R, Xu Q. Prognostic value of glycolysis markers in pancreatic cancer: A systematic review and meta-analysis. Front Oncol 2022; 12:1004850. [PMID: 36172154 PMCID: PMC9510923 DOI: 10.3389/fonc.2022.1004850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Previous studies have investigated the prognostic significance of glycolysis markers in pancreatic cancer; however, conclusions from these studies are still controversial. Methods PubMed, Embase, and Web of Science were systematically searched to investigate the prognostic role of glycolysis markers in pancreatic cancer up to May 2022. Pooled hazard ratios (HRs) with 95% confidence intervals (CIs) related to overall survival (OS), disease free survival (DFS), recurrence-free survival (RFS), and distant metastasis-free survival (DMFS) were calculated using the STATA 12.0 software. Results A total of 28 studies comprising 2010 patients were included in this meta-analysis. High expression of the five glycolysis markers was correlated with a poorer OS (HR = 1.72, 95% CI: 1.34-2.22), DFS (HR = 3.09, 95% CI: 1.91-5.01), RFS (HR = 1.73, 95% CI: 1.21-2.48) and DMFS (HR = 2.60, 95% CI: 1.09-6.20) in patients with pancreatic cancer. In subgroup analysis, it was shown that higher expression levels of the five glycolysis markers were related to a poorer OS in Asians (HR = 1.85, 95% CI: 1.46-2.35, P < 0.001) and Caucasians (HR = 1.97, 95% CI: 1.40-2.77, P < 0.001). Besides, analysis based on the expression levels of specific glycolysis markers demonstrated that higher expression levels of GLUT1 (HR = 2.11, 95% CI: 1.58-2.82, P < 0.001), MCT4 (HR = 2.26, 95% CI: 1.36-3.76, P = 0.002), and ENO1 (HR = 2.16, 95% CI: 1.28-3.66, P =0.004) were correlated with a poorer OS in patients with pancreatic cancer. Conclusions High expression of the five glycolysis markers are associated with poorer OS, DFS, RFS and DMFS in patients with pancreatic cancer, indicating that the glycolysis markers could be potential prognostic predictors and therapeutic targets in pancreatic cancer.
Collapse
|
41
|
Hypoxia-driven metabolic heterogeneity and immune evasive behaviour of gastrointestinal cancers: Elements of a recipe for disaster. Cytokine 2022; 156:155917. [PMID: 35660715 DOI: 10.1016/j.cyto.2022.155917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/28/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022]
Abstract
Gastrointestinal (GI) cancers refer to a group of malignancies associated with the GI tract (GIT). Like other solid tumors, hypoxic regions consistently feature inside the GI tumor microenvironment (TME) and contribute towards metabolic reprogramming of tumor-resident cells by modulating hypoxia-induced factors. We highlight here how the metabolic crosstalk between cancer cells and immune cells generate immunosuppressive environment inside hypoxic tumors. Given the fluctuating nature of tumor hypoxia, the metabolic fluxes between immune cells and cancer cells change dynamically. These changes alter cellular phenotypes and functions, resulting in the acceleration of cancer progression. These evolved properties of hypoxic tumors make metabolism-targeting monotherapy approaches or immunotherapy-measures unsuccessful. The current review highlights the advantages of combined immunometabolic treatment strategies to target hypoxic GI cancers and also identifies research areas to develop better combinational therapeutics for future.
Collapse
|
42
|
Kubik J, Humeniuk E, Adamczuk G, Madej-Czerwonka B, Korga-Plewko A. Targeting Energy Metabolism in Cancer Treatment. Int J Mol Sci 2022; 23:ijms23105572. [PMID: 35628385 PMCID: PMC9146201 DOI: 10.3390/ijms23105572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer is the second most common cause of death worldwide after cardiovascular diseases. The development of molecular and biochemical techniques has expanded the knowledge of changes occurring in specific metabolic pathways of cancer cells. Increased aerobic glycolysis, the promotion of anaplerotic responses, and especially the dependence of cells on glutamine and fatty acid metabolism have become subjects of study. Despite many cancer treatment strategies, many patients with neoplastic diseases cannot be completely cured due to the development of resistance in cancer cells to currently used therapeutic approaches. It is now becoming a priority to develop new treatment strategies that are highly effective and have few side effects. In this review, we present the current knowledge of the enzymes involved in the different steps of glycolysis, the Krebs cycle, and the pentose phosphate pathway, and possible targeted therapies. The review also focuses on presenting the differences between cancer cells and normal cells in terms of metabolic phenotype. Knowledge of cancer cell metabolism is constantly evolving, and further research is needed to develop new strategies for anti-cancer therapies.
Collapse
Affiliation(s)
- Joanna Kubik
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
| | - Ewelina Humeniuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
- Correspondence: ; Tel.: +48-81-448-65-20
| | - Grzegorz Adamczuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
| | - Barbara Madej-Czerwonka
- Human Anatomy Department, Faculty of Medicine, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Agnieszka Korga-Plewko
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
| |
Collapse
|
43
|
Zhang W, Wang C, Hu X, Lian Y, Ding C, Ming L. Inhibition of LDHA suppresses cell proliferation and increases mitochondrial apoptosis via the JNK signaling pathway in cervical cancer cells. Oncol Rep 2022; 47:77. [PMID: 35191522 PMCID: PMC8892607 DOI: 10.3892/or.2022.8288] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/28/2022] [Indexed: 11/08/2022] Open
Abstract
The Warburg effect or aerobic glycolysis is a hallmark of cancer. Lactate dehydrogenase (LDH), which catalyzes conversion of pyruvate into lactate, serves a critical role during Warburg effect. LDH A chain (LDHA), a member of the LDH family, is upregulated in multiple types of cancer and serves a vital role in tumor growth and progression. However, its expression and function in cervical cancer has not been characterized. The present study evaluated LDHA expression in The Cancer Genome Atlas database and found that LDHA was upregulated in cervical cancer compared with normal tissue. To clarify the role of LDHA in cervical cancer HeLa and SiHa cells, lentiviral shRNA was used to stably knockdown LDHA and oxamate, a small-molecule inhibitor of LDHA, was used to inhibit the activity of LDHA. Glucose uptake assay, lactate production measurement and ATP detection assay demonstrated LDHA inhibition notably decreased glucose consumption, lactate production and ATP levels in both HeLa and SiHa cells. Furthermore, the effect of LDHA inhibition on cell proliferation, cell cycle and apoptosis was investigated by MTT, BrdU incorporation, colony formation assay, flow cytometry and western blotting; LDHA knockdown or oxamate treatment led to decreased cell proliferation and increased apoptosis. Inhibition of LDHA induced G2/M cell cycle arrest and activated the mitochondrial apoptosis pathway. Mechanistically, the JNK signaling pathway was key for LDHA inhibition-mediated cell cycle arrest and apoptosis. Collectively, these results indicated that LDHA was involved in cervical cancer pathogenesis and may be a promising therapeutic target for treatment.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R China
| | - Cui Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R China
| | - Xiaomei Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R China
| | - Yanzhen Lian
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R China
| | - Caili Ding
- Zhengzhou Hang Gang Ding Shi Medical Laboratory Co., Ltd., Zhengzhou, Henan 450000, P.R. China
| | - Liang Ming
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R Chin
| |
Collapse
|
44
|
Chang X, Liu X, Wang H, Yang X, Gu Y. Glycolysis in the progression of pancreatic cancer. Am J Cancer Res 2022; 12:861-872. [PMID: 35261808 PMCID: PMC8900001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/14/2022] [Indexed: 06/14/2023] Open
Abstract
Metabolic reprogramming, as a key hallmark of cancers, leads to the malignant behavior of pancreatic cancer, which is closely related to tumor development and progression, as well as the supportive tumor microenvironments. Although cells produce adenosine triphosphate (ATP) from glucose by glycolysis when lacking oxygen, pancreatic cancer cells elicit metabolic conversion from oxide phosphorylation to glycolysis, which is well-known as "Warburg effect". Glycolysis is critical for cancer cells to maintain their robust biosynthesis and energy requirement, and it could promote tumor initiation, invasion, angiogenesis, and metastasis to distant organs. Multiple pathways are involved in the alternation of glycolysis for pancreatic cancer cells, including UHRF1/SIRT4 axis, PRMT5/FBW7/cMyc axis, JWA/AMPK/FOXO3a/FAK axis, KRAS/TP53/TIGAR axis, etc. These signaling pathways play an important role in glycolysis and are potential targets for the treatment of pancreatic cancer. Mutations in glycolytic enzymes (such as LDH, PKM2, and PGK1) also contribute to the early diagnosis and monitoring of pancreatic cancer. In this review, we summarized the recent advances on the mechanisms for glycolysis in pancreatic cancer and the function of glycolysis in the progression of pancreatic cancer, which suggested new targets for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Xinyao Chang
- Department of Immunology, College of Basic Medicine, Naval Medical UniversityShanghai 200433, China
| | - Xingchen Liu
- Department of Pathology, Changhai Hospital, Naval Medical UniversityShanghai 200433, China
| | - Haoze Wang
- Department of Immunology, College of Basic Medicine, Naval Medical UniversityShanghai 200433, China
| | - Xuan Yang
- Department of Immunology, College of Basic Medicine, Naval Medical UniversityShanghai 200433, China
| | - Yan Gu
- Department of Immunology, College of Basic Medicine, Naval Medical UniversityShanghai 200433, China
| |
Collapse
|
45
|
Franczak M, Kutryb-Zajac B, El Hassouni B, Giovannetti E, Granchi C, Minutolo F, Smolenski RT, Peters GJ. The effect of lactate dehydrogenase-A inhibition on intracellular nucleotides and mitochondrial respiration in pancreatic cancer cells. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2022; 41:1375-1385. [PMID: 35130822 DOI: 10.1080/15257770.2022.2031215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 01/14/2023]
Abstract
Pancreatic cancer (PC) is one of the most lethal malignancies. PC is characterized by a high expression of the glucose transporter GLUT-1 and of lactate dehydrogenase A (LDH-A). The novel LDH-A inhibitor NHI-Glc-2 was designed for a better uptake via GLUT-1 and was shown to be cytotoxic against the PC cell line PANC-1. Using RP-HPLC we investigated its effect on adenine nucleotides and NADH/NAD+, while the Seahorse analyzer was used to determine its effect on glycolysis and mitochondrial function. A 24 hour exposure to 10 µM NHI-Glc-2 (around the IC50) decreased the ATP concentration by about 10%, but at 25 µM this decrease was 38%, while NAD+ decreased by 26%, associated with a 35% decrease in the NADH/NAD+ ratio. A 10 µM NHI-Glc-2 decreased extracellular acidification and oxygen consumption (about 75%), as well as the mitochondrial respiration parameters by 50%. In conclusion, LDH-A inhibition markedly affected the energy supply of PANC-1 cells. The respiration data indicated a dependency of the cells on glycolysis and fatty acid oxidation.Supplemental data for this article is available online at https://doi.org/10.1080/15257770.2022.2031215 .
Collapse
Affiliation(s)
- Marika Franczak
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | | | - Btissame El Hassouni
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Fondazione Pisana per la Scienza, Pisa, Italy
| | | | | | | | - Godefridus J Peters
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Shibuki T, Mizuta T, Shimokawa M, Koga F, Ueda Y, Nakazawa J, Komori A, Otsu S, Arima S, Fukahori M, Makiyama A, Taguchi H, Honda T, Mitsugi K, Nio K, Ide Y, Ureshino N, Shirakawa T, Otsuka T. Prognostic nomogram for patients with unresectable pancreatic cancer treated with gemcitabine plus nab-paclitaxel or FOLFIRINOX: A post-hoc analysis of a multicenter retrospective study in Japan (NAPOLEON study). BMC Cancer 2022; 22:19. [PMID: 34980029 PMCID: PMC8722136 DOI: 10.1186/s12885-021-09139-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 12/22/2021] [Indexed: 01/04/2023] Open
Abstract
Background No reliable nomogram has been developed until date for predicting the survival in patients with unresectable pancreatic cancer undergoing treatment with gemcitabine plus nab–paclitaxel (GnP) or FOLFIRINOX. Methods This analysis was conducted using clinical data of Japanese patients with unresectable pancreatic cancer undergoing GnP or FOLFIRINOX treatment obtained from a multicenter study (NAPOLEON study). A Cox proportional hazards model was used to identify the independent prognostic factors. A nomogram to predict 6–, 12–, and 18–month survival probabilities was generated, validated by using the concordance index (C–index), and calibrated by the bootstrapping method. And then, we attempted risk stratification for survival by classifying the patients according to the sum of the scores on the nomogram (total nomogram points). Results A total of 318 patients were enrolled. A prognostic nomogram was generated using data on the Eastern Cooperative Oncology Group performance status, liver metastasis, serum LDH, serum CRP, and serum CA19–9. The C–indexes of the nomogram were 0.77, 0.72 and 0.70 for 6–, 12–, and 18–month survival, respectively. The calibration plot showed optimal agreement at all points. Risk stratification based on tertiles of the total nomogram points yielded clear separations of the survival curves. The median survival times in the low–, moderate–, and high–risk groups were 15.8, 12.8 and 7.8 months (P<0.05), respectively. Conclusions Our nomogram might be a convenient and inexpensive tool to accurately predict survival in Japanese patients with unresectable pancreatic cancer undergoing treatment with GnP or FOLFIRINOX, and will help clinicians in selecting appropriate therapeutic strategies for individualized management. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09139-y.
Collapse
Affiliation(s)
- Taro Shibuki
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, 860 Ninose-ko, Arita-cho, Nishi-matsuura-gun, Saga, 849-4193, Japan.,Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Toshihiko Mizuta
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, 860 Ninose-ko, Arita-cho, Nishi-matsuura-gun, Saga, 849-4193, Japan.,Department of Internal Medicine, Fujikawa Hospital, 1-2-6 Matsubara, Saga-shi, Saga, 840-0831, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Hospital Organization Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka-shi, Fukuoka, 811-1395, Japan.,Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga, 840-8571, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, 2-1-1 Nagamine-minami, Higashi-ku, Kumamoto-shi, Kumamoto, 861-8520, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-cho, Kagoshima-shi, Kagoshima, 890-8760, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita, 879-5593, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita, 879-5593, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima-shi, Kagoshima, 890-8520, Japan
| | - Masaru Fukahori
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Akitaka Makiyama
- Department of Hematology/Oncology, Japan Community Healthcare Organization Kyushu Hospital, 1-8-1 Kishinoura, Yahatanishi-ku, Kitakyushu-shi, Fukuoka, 806-8501, Japan.,Cancer Center, Gifu University Hospital, 1-1 Yanagido, Gifu-shi, Gifu, 501-1194, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-machi, Satsumasendai-shi, Kagoshima, 895-0074, Japan.,Department of Gastroenterology, Izumi General Medical Center, 520 Myojincho, Izumi-shi, Kagoshima, 899-0131, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8501, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-8539, Japan.,Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-cho, Sasebo-shi, Nagasaki, 857-8575, Japan
| | - Kenta Nio
- Department of Medical Oncology, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-8539, Japan.,Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-cho, Sasebo-shi, Nagasaki, 857-8575, Japan
| | - Yasushi Ide
- Department of Internal Medicine, Karatsu Red Cross Hospital, 2430 Watada, Karatsu-shi, Saga, 847-8588, Japan
| | - Norio Ureshino
- Department of Medical Oncology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga, 840-8571, Japan.,Department of Medical Oncology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba, 292-8535, Japan
| | - Tsuyoshi Shirakawa
- Department of Medical Oncology, Fukuoka Wajiro Hospital, 2-2-75 Wajirogaoka, Higashi-ku, Fukuoka-shi, Fukuoka, 811-0213, Japan. .,Karatsu Higashi-matsuura Medical Association Center, 2566-11 Chiyoda-machi, Karatsu-shi, Saga, 847-0041, Japan.
| | - Taiga Otsuka
- Department of Medical Oncology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga, 840-8571, Japan.,Department of Internal Medicine, Minato Medical Clinic, 3-11-3 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-0072, Japan
| |
Collapse
|
47
|
Metformin Treatment or PRODH/POX-Knock out Similarly Induces Apoptosis by Reprograming of Amino Acid Metabolism, TCA, Urea Cycle and Pentose Phosphate Pathway in MCF-7 Breast Cancer Cells. Biomolecules 2021; 11:biom11121888. [PMID: 34944532 PMCID: PMC8699520 DOI: 10.3390/biom11121888] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022] Open
Abstract
It has been considered that proline dehydrogenase/proline oxidase (PRODH/POX) is involved in antineoplastic activity of metformin (MET). The aim of this study is identification of key metabolites of glycolysis, pentose phosphate pathway (PPP), tricarboxylic acids (TCA), urea cycles (UC) and some amino acids in MET-treated MCF-7 cells and PRODH/POX-knocked out MCF-7 (MCF-7crPOX) cells. MCF-7crPOX cells were generated by using CRISPR-Cas9. Targeted metabolomics was performed by LC-MS/MS/QqQ. Expression of pro-apoptotic proteins was evaluated by Western blot. In the absence of glutamine, MET treatment or PRODH/POX-knock out of MCF-7 cells contributed to similar inhibition of glycolysis (drastic increase in intracellular glucose and pyruvate) and increase in the utilization of phospho-enol-pyruvic acid, glucose-6-phosphate and some metabolites of TCA and UC, contributing to apoptosis. However, in the presence of glutamine, MET treatment or PRODH/POX-knock out of MCF-7 cells contributed to utilization of some studied metabolites (except glucose), facilitating pro-survival phenotype of MCF-7 cells in these conditions. It suggests that MET treatment or PRODH/POX-knock out induce similar metabolic effects (glucose starvation) and glycolysis is tightly linked to glutamine metabolism in MCF-7 breast cancer cells. The data provide insight into mechanism of anticancer activity of MET as an approach to further studies on experimental breast cancer therapy.
Collapse
|
48
|
Zhang Z, Zhang HJ. Glycometabolic rearrangements-aerobic glycolysis in pancreatic ductal adenocarcinoma (PDAC): roles, regulatory networks, and therapeutic potential. Expert Opin Ther Targets 2021; 25:1077-1093. [PMID: 34874212 DOI: 10.1080/14728222.2021.2015321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Glycometabolic rearrangements (aerobic glycolysis) is a hallmark of pancreatic ductal adenocarcinoma (PDAC) and contributes to tumorigenesis and progression through numerous mechanisms. The targeting of aerobic glycolysis is recognized as a potential therapeutic strategy which offers the possibility of improving treatment outcomes for PDAC patients. AREAS COVERED In this review, the role of aerobic glycolysis and its regulatory networks in PDAC are discussed. The targeting of aerobic glycolysis in PDAC is examined, and its therapeutic potential is evaluated. The relevant literature published from 2001 to 2021 was searched in databases including PubMed, Scopus, and Embase. EXPERT OPINION Regulatory networks of aerobic glycolysis in PDAC are based on key factors such as c-Myc, hypoxia-inducible factor 1α, the mammalian target of rapamycin pathway, and non-coding RNAs. Experimental evidence suggests that modulators or inhibitors of aerobic glycolysis promote therapeutic effects in preclinical tumor models. Nevertheless, successful clinical translation of drugs that target aerobic glycolysis in PDAC is an obstacle. Moreover, it is necessary to identify the potential targets for future interventions from regulatory networks to design efficacious and safer agents.
Collapse
Affiliation(s)
- Zhong Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, People's Republic of China
| | - Hai-Jun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
49
|
Carvalho TMA, Di Molfetta D, Greco MR, Koltai T, Alfarouk KO, Reshkin SJ, Cardone RA. Tumor Microenvironment Features and Chemoresistance in Pancreatic Ductal Adenocarcinoma: Insights into Targeting Physicochemical Barriers and Metabolism as Therapeutic Approaches. Cancers (Basel) 2021; 13:6135. [PMID: 34885243 PMCID: PMC8657427 DOI: 10.3390/cancers13236135] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, the median overall survival of PDAC patients rarely exceeds 1 year and has an overall 5-year survival rate of about 9%. These numbers are anticipated to worsen in the future due to the lack of understanding of the factors involved in its strong chemoresistance. Chemotherapy remains the only treatment option for most PDAC patients; however, the available therapeutic strategies are insufficient. The factors involved in chemoresistance include the development of a desmoplastic stroma which reprograms cellular metabolism, and both contribute to an impaired response to therapy. PDAC stroma is composed of immune cells, endothelial cells, and cancer-associated fibroblasts embedded in a prominent, dense extracellular matrix associated with areas of hypoxia and acidic extracellular pH. While multiple gene mutations are involved in PDAC initiation, this desmoplastic stroma plays an important role in driving progression, metastasis, and chemoresistance. Elucidating the mechanisms underlying PDAC resistance are a prerequisite for designing novel approaches to increase patient survival. In this review, we provide an overview of the stromal features and how they contribute to the chemoresistance in PDAC treatment. By highlighting new paradigms in the role of the stromal compartment in PDAC therapy, we hope to stimulate new concepts aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Tiago M. A. Carvalho
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | - Maria Raffaella Greco
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | | | - Khalid O. Alfarouk
- Al-Ghad International College for Applied Medical Sciences, Al-Madinah Al-Munwarah 42316, Saudi Arabia;
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | - Rosa A. Cardone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| |
Collapse
|
50
|
Atlı Şekeroğlu Z, Şekeroğlu V, Işık S, Aydın B. Trimetazidine alone or in combination with gemcitabine and/or abraxane decreased cell viability, migration and ATP levels and induced apoptosis of human pancreatic cells. Clin Res Hepatol Gastroenterol 2021; 45:101632. [PMID: 33662778 DOI: 10.1016/j.clinre.2021.101632] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/22/2020] [Accepted: 01/05/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Trimetazidine (TMZ) is an anti-ischemic agent that can inhibit the fatty acid oxidation. It has been stated that inhibition of fatty acid oxidation may be an acceptable approach to cancer treatment. METHODS We examined the effects of TMZ alone or together with abraxane (ABX) and/or gemcitabine (GEM) on cell viability, apoptosis, adhesion, migration and ATP levels of human pancreatic cancer cell line PANC-1. RESULTS TMZ significantly reduced the cell viability at higher concentrations. Lower cell viability values were found in cells co-treated with TMZ + GEM, TMZ + ABX and GEM + ABX. The combined treatment of TMZ with ABX and/or GEM significantly increased the apoptosis rates. The highest percentages of apoptosis were found in TMZ + ABX or TMZ + ABX + GEM treatments. TMZ alone or together with ABX and/or GEM significantly reduced the ATP levels. The lowest migration rates were also found at TMZ + ABX and TMZ + ABX + GEM treatments. CONCLUSIONS Our study is the first study to indicate that TMZ can induce cytotoxicity and apoptosis and reduce migration and ATP levels, especially in cells co-treated with ABX and/or GEM. A combination strategy based on inhibition of fatty acid oxidation and anticancer drugs may be more effective in the treatment of pancreatic cancers.
Collapse
Affiliation(s)
- Zülal Atlı Şekeroğlu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, 52200 Ordu, Turkey.
| | - Vedat Şekeroğlu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, 52200 Ordu, Turkey
| | - Sevil Işık
- Department of General Surgery, Medicana International İzmir Hospital, İzmir, Turkey
| | - Birsen Aydın
- Department of Biology, Faculty of Science and Letters, Amasya University, Amasya, Turkey
| |
Collapse
|