1
|
Li Y, Shu J, Tan P, Dong X, Zhang M, He T, Yang Z, Zhang X, Giovannucci EL, Liu Z, Zhou Z, Li Q, Xu Y, Xu X, Peng T, Lu J, Zhang Y, Zhu H, Fang A. Genetic variants in folate metabolism-related genes, serum folate and hepatocellular carcinoma survival: the Guangdong Liver Cancer Cohort study. Br J Nutr 2024; 132:1411-1422. [PMID: 39506332 DOI: 10.1017/s0007114524001776] [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: 11/08/2024]
Abstract
Folate metabolism is involved in the development and progression of various cancers. We investigated the association of single nucleotide polymorphisms (SNP) in folate-metabolising genes and their interactions with serum folate concentrations with overall survival (OS) and liver cancer-specific survival (LCSS) of newly diagnosed hepatocellular carcinoma (HCC) patients. We detected the genotypes of six SNP in three genes related to folate metabolism: methylenetetrahydrofolate reductase (MTHFR), 5-methyltetrahydrofolate-homocysteine methyltransferase reductase (MTRR) and 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR). Cox proportional hazard models were used to calculate multivariable-adjusted hazard ratios (HR) and 95 % CI. This analysis included 970 HCC patients with genotypes of six SNP, and 864 of them had serum folate measurements. During a median follow-up of 722 d, 393 deaths occurred, with 360 attributed to HCC. In the fully-adjusted models, the MTRR rs1801394 polymorphism was significantly associated with OS in additive (per G allele: HR = 0·84, 95 % CI: 0·71, 0·99), co-dominant (AG v. AA: HR = 0·77; 95 % CI: 0·62, 0·96) and dominant (AG + GG v. AA: HR = 0·78; 95 % CI: 0·63, 0·96) models. Carrying increasing numbers of protective alleles was linked to better LCSS (HR10–12 v. 2–6 = 0·70; 95 % CI: 0·49, 1·00) and OS (HR10–12 v. 2–6 = 0·67; 95 % CI: 0·47, 0·95). Furthermore, we observed significant interactions on both multiplicative and additive scales between serum folate levels and MTRR rs1801394 polymorphism. Carrying the variant G allele of the MTRR rs1801394 is associated with better HCC prognosis and may enhance the favourable association between higher serum folate levels and improved survival among HCC patients.
Collapse
Affiliation(s)
- Yunshan Li
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing Shu
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Peishan Tan
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaocong Dong
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Mingjie Zhang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Tongtong He
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhijun Yang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xuehong Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Yale University School of Nursing, Orange, CT, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zhaoyan Liu
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhongguo Zhou
- Department of Hepatobiliary Surgery, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Qijiong Li
- Department of Hepatobiliary Surgery, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yanjun Xu
- Department of Chronic Noncommunicable Disease Prevention and Control, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, People's Republic of China
| | - Xiaojun Xu
- Department of Chronic Noncommunicable Disease Prevention and Control, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, People's Republic of China
| | - Tianyou Peng
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jialin Lu
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yaojun Zhang
- Department of Hepatobiliary Surgery, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Huilian Zhu
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Aiping Fang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
2
|
Zou Y, Yuan Y, Zhou Q, Yue Z, Liu J, Fan L, Xu H, Xin L. The Role of Methionine Restriction in Gastric Cancer: A Summary of Mechanisms and a Discussion on Tumor Heterogeneity. Biomolecules 2024; 14:161. [PMID: 38397398 PMCID: PMC10887009 DOI: 10.3390/biom14020161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Gastric cancer is ranked as the fifth most prevalent cancer globally and has long been a topic of passionate discussion among numerous individuals. However, the incidence of gastric cancer in society has not decreased, but instead has shown a gradual increase in recent years. For more than a decade, the treatment effect of gastric cancer has not been significantly improved. This is attributed to the heterogeneity of cancer, which makes popular targeted therapies ineffective. Methionine is an essential amino acid, and many studies have shown that it is involved in the development of gastric cancer. Our study aimed to review the literature on methionine and gastric cancer, describing its mechanism of action to show that tumor heterogeneity in gastric cancer does not hinder the effectiveness of methionine-restricted therapies. This research also aimed to provide insight into the inhibition of gastric cancer through metabolic reprogramming with methionine-restricted therapies, thereby demonstrating their potential as adjuvant treatments for gastric cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lin Xin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang 330006, China; (Y.Z.); (Y.Y.); (Q.Z.); (Z.Y.); (J.L.); (L.F.); (H.X.)
| |
Collapse
|
3
|
Na D, Chae J, Cho SY, Kang W, Lee A, Min S, Kang J, Kim MJ, Choi J, Lee W, Shin D, Min A, Kim YJ, Lee KH, Kim TY, Suh YS, Kong SH, Lee HJ, Kim WH, Park H, Im SA, Yang HK, Lee C, Kim JI. Predictive biomarkers for 5-fluorouracil and oxaliplatin-based chemotherapy in gastric cancers via profiling of patient-derived xenografts. Nat Commun 2021; 12:4840. [PMID: 34376661 PMCID: PMC8355375 DOI: 10.1038/s41467-021-25122-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 07/16/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is commonly treated by chemotherapy using 5-fluorouracil (5-FU) derivatives and platinum combination, but predictive biomarker remains lacking. We develop patient-derived xenografts (PDXs) from 31 GC patients and treat with a combination of 5-FU and oxaliplatin, to determine biomarkers associated with responsiveness. When the PDXs are defined as either responders or non-responders according to tumor volume change after treatment, the responsiveness of PDXs is significantly consistent with the respective clinical outcomes of the patients. An integrative genomic and transcriptomic analysis of PDXs reveals that pathways associated with cell-to-cell and cell-to-extracellular matrix interactions enriched among the non-responders in both cancer cells and the tumor microenvironment (TME). We develop a 30-gene prediction model to determine the responsiveness to 5-FU and oxaliplatin-based chemotherapy and confirm the significant poor survival outcomes among cases classified as non-responder-like in three independent GC cohorts. Our study may inform clinical decision-making when designing treatment strategies.
Collapse
Affiliation(s)
- Deukchae Na
- Ewha Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, Seoul, Korea
| | - Jeesoo Chae
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Evolution Research Center, The Catholic University of Korea, Seoul, Korea
| | - Sung-Yup Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea
| | - Wonyoung Kang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Ahra Lee
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Seoyeon Min
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Jinjoo Kang
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Min Jung Kim
- Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea
| | - Jaeyong Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Woochan Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Dongjin Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Ahrum Min
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yu-Jin Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Kyung-Hun Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Tae-Yong Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Seoul National University Bundang Hospital, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk-Joon Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Woo-Ho Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Hansoo Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University, Seoul, Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| | - Han-Kwang Yang
- Cancer Research Institute, Seoul National University, Seoul, Korea.
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea.
| | - Charles Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Life Science, Ewha Womans University, Seoul, Korea.
- Precision Medicine Center, The First Affiliated Hospital of Xiu'an Jiaotong University, Shaanxi, People's Republic of China.
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
- Cancer Research Institute, Seoul National University, Seoul, Korea.
- Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea.
| |
Collapse
|
4
|
Zhao T, Li W, Chen J, Qin W. Genomic variants in Fas-mediated apoptosis pathway predict a poor response to Platinum-based Chemotherapy for Chinese Gastric Cancer Patients. J Cancer 2021; 12:849-859. [PMID: 33403042 PMCID: PMC7778532 DOI: 10.7150/jca.48120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 02/09/2020] [Indexed: 12/16/2022] Open
Abstract
Platinum-based adjuvant chemotherapy is very common for gastric cancer (GC) patients, but the chemotherapy sensitivity is very heterogeneous. The genomic variants and the gene-gene interactions involved in Fas-mediated apoptosis pathway including Fas (FAS 1377 G > A and 670 A > G), FasL (FASL 844 C > T) and caspase-8 (CASP8 -652 6N ins > del or I > D), may paly vital roles in the response to platinum-based treatment. In our investigation, 662 stage II-III postoperative GC patients were enrolled between 1998 and 2006. 261 patients accepted platinum-based regimens and the remaining 401 were not. The log rank tests, Kaplan Meier plots, Pearson chi-square tests, Student t-tests and Cox regression analyses were performed. For the chemotherapy cohort, FAS 1377 G > A or FAS 670 A > G variants alone was related with inferior survival, and a greater than additive effect was identified when patients simultaneously carrying FAS 1377 GA and FAS 670 GA genotypes. But the poor response was neutralized when patients simultaneously carrying FASL 844 C > T or CASP8 -652 6N ins > del mutations. Our study suggested that FAS 1377 G > A and FAS 670 A > G variants may serve as potential biomarkers to predict the response to platinum-based adjuvant chemotherapy, and the gene-gene interactions involved in Fas-mediated apoptosis pathway may enhance or neutralize the chemosensitivity.
Collapse
Affiliation(s)
- Tingting Zhao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 21000, China
| | - Wei Li
- Department of Gynecology, Zhenjiang Maternity and Childcare Hospital, Zhenjiang, 212000, China
| | - Jinfei Chen
- Cancer Center, Taikang Xianlin Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, 21000, China
| | - Weisong Qin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 21000, China
| |
Collapse
|
5
|
Yuan L, Liu Z, Wei G, Yang P, Hu XE, Qu F, Lu J, He X, Bao G. Genetic polymorphisms in folate-metabolizing genes associated with gastric cancer prognosis in northwest China subjects. J Cancer 2020; 11:6413-6420. [PMID: 33033524 PMCID: PMC7532508 DOI: 10.7150/jca.46978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022] Open
Abstract
Influence of folate metabolism has long been studied in cancer and copies evidences have suggested that the key genes involved were correlated with GC risk and prognosis. However, their genetically association and contribution for GC prognosis are still elusive. To evaluate the effect of folate metabolism related genes polymorphisms on the prognosis of gastric cancer (GC), the genotype of seven single nucleotide polymorphisms (SNPs) of three genes were selected and genotyped in a cohort of 664 GC patients, including genes of Methylenetetrahydrofolate reductase (MTHFR), Methionine synthase reductase (MTRR), and Methionine synthase (MTR). Kaplan-Meier Curve, long-rank tests and multivariate Cox proportional hazard model were used for prognosis analysis. The results demonstrated that TT or CT/TT genotypes of SNP rs1532268 in MTRR gene coding region are significantly associated with a poorer overall survival (OS) when compared with CC genotype (HR=2.340, 95% CI: 1.240-4.414, p=0.009; or HR=1.502, 95% CI: 1.083-2.085, p=0.015, respectively). Furthermore, comparing to that of the CC genotype, the detrimental effect of rs1532268 TT genotype was also evident in the special subgroups of GC patients, especially in patients with BMI<24 and H. pylori infection. Moreover, significant association between increased relapse and TT genotype of rs1532268 was also observed in patients who are females, BMI<24 and without chemotherapy. In addition, the joint analysis demonstrated that integration of rs1532268 genotypes and BMI, H. pylori infection status, clinical stage and tumor site may significantly improve the predictive abilities for predicting OS of GC patients. In conclusion, it suggested that the MTRR rs1532268 polymorphism is significantly associated with clinical outcomes of GC patients, especially in those with lower BMI (BMI<24) or positive H. pylori infection status, which warrants further validation. And the polymorphism of MTRR rs1532268 may be a potential prognostic factor for GC patients.
Collapse
Affiliation(s)
- Lijuan Yuan
- Department of General Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Ziyu Liu
- Department of Microbiology, The Air Force Military Medical University, Xi'an, 710032, China
| | - Gang Wei
- Department of General Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Ping Yang
- Department of General Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Xi E Hu
- Department of General Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Falin Qu
- Department of General Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Jianguo Lu
- Department of General Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Xianli He
- Department of General Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Guoqiang Bao
- Department of General Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| |
Collapse
|
6
|
Jung SY, Papp JC, Sobel EM, Zhang ZF. Mendelian Randomization Study: The Association Between Metabolic Pathways and Colorectal Cancer Risk. Front Oncol 2020; 10:1005. [PMID: 32850306 PMCID: PMC7396568 DOI: 10.3389/fonc.2020.01005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/21/2020] [Indexed: 12/24/2022] Open
Abstract
Background: The roles of obesity-related biomarkers and their molecular pathways in the development of postmenopausal colorectal cancer (CRC) have been inconclusive. We examined insulin resistance (IR) as a major hormonal pathway mediating the association between obesity and CRC risk in a Mendelian randomization (MR) framework. Methods: We performed MR analysis using individual-level data of 11,078 non-Hispanic white postmenopausal women from our earlier genome-wide association study. We identified four independent single-nucleotide polymorphisms associated with fasting glucose (FG), three with fasting insulin (FI), and six with homeostatic model assessment-IR (HOMA-IR), which were not associated with obesity. We estimated hazard ratios (HRs) for CRC by adjusting for potential confounding factors plus genetic principal components. Results: Overall, we observed no direct association between combined 13 IR genetic instruments and CRC risk (HR = 0.96, 95% confidence interval [CI]: 0.78-1.17). In phenotypic analysis, genetically raised HOMA-IR exhibited its effects on the increased risk and FG and FI on the reduced risk for CRC, but with a lack of statistical power. Subgroup analyses by physical activity level and dietary fat intake with combined phenotypes showed that genetically determined IR was associated with reduced CRC risk in both physical activity-stratified (single contributor: MTRR rs722025; HR = 0.12, 95% CI: 0.02-0.62) and high-fat diet subgroups (main contributor: G6PC2 rs560887; HR = 0.59, 95% CI: 0.37-0.94). Conclusions: Complex evidence was observed for a potential causal association between IR and CRC risk. Our findings may provide an additional value of intervention trials to lower IR and reduce CRC risk.
Collapse
Affiliation(s)
- Su Yon Jung
- Translational Sciences Section, Jonsson Comprehensive Cancer Center, School of Nursing, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jeanette C. Papp
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Eric M. Sobel
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Zuo-Feng Zhang
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
7
|
Yang J, Zheng W, Xu Z, Chen J. MAP3K1 rs889312 genotypes influence survival outcomes of Chinese gastric cancer patients who received adjuvant chemotherapy based on platinum and fluorouracil regimes. Onco Targets Ther 2019; 12:6843-6855. [PMID: 31686841 PMCID: PMC6709816 DOI: 10.2147/ott.s205438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/30/2019] [Indexed: 12/23/2022] Open
Abstract
Background For patients with gastric cancer (GC), adjuvant chemotherapy is a standard therapy. However, the responses to the treatment are quite different. Mitogen-activated protein kinase (MAPK) pathway is a core pathway that modulates the efficacy of anticancer drugs. The purpose of our study was to investigate the clinical significance of one pivotal functional gene polymorphism in the MAPK pathway – MAP3K1 rs889312 – in patients with stage II GC to stage III GC. Methods The genotypes of MAP3K1 rs889312 were analyzed in 591 GC patients enrolled in this study who had received radical gastrectomy. Among them, 204 patients accepted adjuvant chemotherapy based on platinum and fluorouracil (PF) regimens after an operation. Cox regression analysis, log-rank test and Kaplan–Meier method were used to explore the link between MAP3K1 rs889312 variant and overall survival (OS) of GC. Results Compared with the AA genotype (mean OS of 68.12 months), MAP3K1 rs889312 AC/CC significantly reduced the mean OS of 56.83 months in patients who received adjuvant chemotherapy only. In addition, AC/CC genotype had a negative impact on OS of patients who received oxaliplatin-based therapy (HR, 8.253; 95% CI: 1.119–60.853, log-rank p=0.013). Stratification analysis showed that MAP3K1 rs889312 AC/CC significantly reduced OS of patients with tumors smaller than or equal to 5 cm in size (HR, 3.706; 95% CI: 1.329–10.335, p=0.012), poorly differentiated tumors (HR, 3.002; 95% CI: 1.076–8.377, p=0.036) and intestinal tumors (HR, 4.780; 95% CI: 1.138–20.073, p=0.033). Conclusion Our findings suggested that MAP3K1 rs889312 single-nucleotide polymorphism may be considered as a biomarker for adjuvant chemotherapy reaction and can predict prognosis of GC patients who received PF-based therapy.
Collapse
Affiliation(s)
- Jian Yang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, People's Republic of China.,Department of Oncology, The Affiliated Yixing Hospital of Jiangsu University, Yixing 214200, People's Republic of China
| | - Wei Zheng
- Department of General Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing 214200, People's Republic of China
| | - Zhi Xu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, People's Republic of China.,ICR Medical Affairs, ICON Plc, Shanghai 200003, People's Republic of China
| | - Jinfei Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, People's Republic of China.,Cancer Center, TaiKang Xianlin Drum Tower Hospital, Nanjing University School of Medicine, Nanjing 210046, People's Republic of China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210006, People's Republic of China
| |
Collapse
|
8
|
Dong L, Dong Q, Chen Y, Li Y, Zhang B, Zhou F, Lyu X, Chen GG, Lai P, Kung HF, He ML. Novel HDAC5-interacting motifs of Tbx3 are essential for the suppression of E-cadherin expression and for the promotion of metastasis in hepatocellular carcinoma. Signal Transduct Target Ther 2018; 3:22. [PMID: 30151243 PMCID: PMC6107554 DOI: 10.1038/s41392-018-0025-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/22/2018] [Accepted: 07/17/2018] [Indexed: 12/15/2022] Open
Abstract
Tbx3, a transcriptional repressor, is essential in the organogenesis of vertebrates, stem cell self-renewal and differentiation, and the carcinogenesis of multiple tumor types. However, the mechanism by which Tbx3 participates in the metastasis of hepatocellular carcinoma (HCC) remains largely unknown. In this study, we show that Tbx3 was dramatically upregulated in clinical HCC samples and that elevated expression of Tbx3 promoted cancer progression. To determine the underlying mechanism, systematic glycine scan mutagenesis and deletion assays were performed. We identified two critical motifs, 585LFSYPYT591 and 604HRH606, that contribute to the repression of transcriptional activity. These motifs are also essential for Tbx3 to promote cell migration and metastasis both in vitro and in vivo via the suppression of E-cadherin expression. More importantly, Tbx3 directly interacts with HDAC5 via these motifs, and an HDAC inhibitor blocks Tbx3-mediated cell migration and the downregulation of E-cadherin in HCC. As Tbx3 is involved in the carcinogenesis of multiple types of human cancers, our findings suggest an important target for anti-cancer drug development. A regulatory protein that represses gene activity interacts with an enzyme involved in chromosome remodeling to promote the migration and metastasis of liver cancer cells. Ming-Liang He from the City University of Hong Kong and colleagues found that levels of the T-box transcription factor Tbx3 were dramatically increased in tissue biopsies of liver tumors. They injected Tbx3-expressing human liver cancer cells into mice and saw a positive correlation between Tbx3 activity and cancer progression. By mutating and deleting parts of Tbx3, the researchers identified two particular stretches of the protein that bind histone deacetylase 5, an enzyme involved in ensuring DNA coils, are wound tight to suppress gene activity. This interaction is needed for Tbx3’s tumor-promoting function and may be targetable with drugs in order to prevent metastasis in patients with aggressive liver cancer.
Collapse
Affiliation(s)
- Liang Dong
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Qi Dong
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Ying Chen
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yichen Li
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Bao Zhang
- 2School of Public Health and Tropical Medicine, Southern Medical University, 1023 Shatai Road, 510515 Guangzhou, China
| | - Fanghang Zhou
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Xiaoming Lyu
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - George G Chen
- 3Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Paul Lai
- 3Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hsiang-Fu Kung
- 4Key Laboratory of Tumor Immunopathology, Ministry of Education of China, and Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, 400038 Chongqing, China
| | - Ming-Liang He
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,Biotechnology and Health Center, CityU Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
9
|
Tang C, Yu S, Jiang H, Li W, Xu X, Cheng X, Peng K, Chen E, Cui Y, Liu T. A Meta-Analysis: Methylenetetrahydrofolate Reductase C677T Polymorphism in Gastric Cancer Patients Treated with 5-Fu Based Chemotherapy Predicts Serious Hematologic Toxicity but Not Prognosis. J Cancer 2018; 9:1057-1066. [PMID: 29581785 PMCID: PMC5868173 DOI: 10.7150/jca.23391] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/29/2018] [Indexed: 12/17/2022] Open
Abstract
5-fluorouracil (5-Fu) metabolism associated enzyme, methylenetetrahydrofolate reductase (MTHFR)'s polymorphism C677T can affect enzyme activity and a series of studies have been performed to examine the association of this MTHFR polymorphism with the clinical outcomes of gastric cancer (GC) patients treated with 5-Fu based chemotherapies. However, the results are inconsistent and inconclusive. Therefore, a more comprehensive summary like meta-analysis on this topic is needed. We performed a systematic literature search of PubMed and Embase up to May 20, 2017. Researches exploring MTHFR polymorphisms C677T's relationship with the clinical outcomes (response rate, overall survival and toxicity) of GC patients treated with 5-Fu based chemotherapy were included. The association was measured by odds ratios (ORs) or hazard ratios (HRs) combined with their 95% confidence intervals (CIs) using random/fixed effects model according to the studies' heterogeneity. Subgroup, sensitivity and publication bias analyses were conducted. Thirteen studies were finally included in this meta-analysis. No significant association was found between response rate [TT/ (CC+CT) OR=1.31, 95% CI: 0.62-2.76] or overall survival [(CT+TT)/CC HR=1.05, 95% CI: 0.86-1.26; TT/(CT+CC) HR=1.48, 95% CI: 0.53-4.15] and MTHFR polymorphism C677T. However, GC patients with CC or CT genotype tended to experience less severe hematologic toxicity than those with TT genotype [(CC+CT)/TT OR=0.66, 95% CI: 0.48-0.91]. In conclusion, MTHFR C677T polymorphism predicts severe hematologic toxicity in GC patients receiving 5-Fu based chemotherapy, but not the efficiency.
Collapse
Affiliation(s)
- Cheng Tang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huiqin Jiang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaojing Xu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xi Cheng
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ke Peng
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Erbao Chen
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Wang P, Li S, Wang M, He J, Xi S. Association of MTRR A66G polymorphism with cancer susceptibility: Evidence from 85 studies. J Cancer 2017; 8:266-277. [PMID: 28243331 PMCID: PMC5327376 DOI: 10.7150/jca.17379] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/14/2016] [Indexed: 02/07/2023] Open
Abstract
Methionine synthase reductase (MTRR) is a key regulatory enzyme involved in the folate metabolic pathway. Previous studies investigating the association of MTRR A66G polymorphism with cancer susceptibility reported inconclusive results. We performed the current meta-analysis to obtain a more precise estimation of the possible association. Published literatures were identified from PubMed, Embase and CBM databases up to October 2016. The strength of the association between the MTRR A66G polymorphism and cancer susceptibility was assessed using odds ratios (ORs) and the corresponding 95% confidence intervals (CIs). Eighty five published studies with 32,272 cases and 37,427 controls were included in this meta-analysis. Pooled results indicated that the MTRR A66G polymorphism was associated with an increased overall cancer risk (homozygous model: OR = 1.08, 95% CI = 1.02-1.15, P = 0.009; recessive model: OR = 1.06, 95% CI = 1.00-1.12, P < 0.001 and allele comparison: OR = 1.03, 95% CI = 1.00-1.06, P < 0.001). Stratification analysis further indicated significant associations in head and neck cancer, Caucasians, Africans, and high quality studies. However, to avoid the "false-positive report", the significant findings were assessed by the false-positive report probability (FPRP) test. Interestingly, the results of FPRP test revealed that the increased risk for MTRR A66G polymorphism among Africans need further validation due to the high probabilities of false-positive results. This meta-analysis suggests that the MTRR A66G polymorphism is associated with significantly increased cancer risk, a finding that needs to be confirmed in single large studies.
Collapse
Affiliation(s)
- Ping Wang
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, Henan, China
| | - Sanqiang Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang 471023, Henan, China
| | - Meilin Wang
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, Henan, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
- ✉ Corresponding authors: Shoumin Xi, The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, No. 263 Kaiyuan Avenue, Luoyang 471023, Henan, China, Tel.: (+86-379) 64830346, Fax: (+86-379) 64830345, E-mail: ; or Jing He, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou 510623, Guangdong, China, Tel./Fax: (+86-20) 38076560, E-mail:
| | - Shoumin Xi
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, Henan, China
- ✉ Corresponding authors: Shoumin Xi, The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, No. 263 Kaiyuan Avenue, Luoyang 471023, Henan, China, Tel.: (+86-379) 64830346, Fax: (+86-379) 64830345, E-mail: ; or Jing He, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou 510623, Guangdong, China, Tel./Fax: (+86-20) 38076560, E-mail:
| |
Collapse
|
11
|
Shelton J, Lu X, Hollenbaugh JA, Cho JH, Amblard F, Schinazi RF. Metabolism, Biochemical Actions, and Chemical Synthesis of Anticancer Nucleosides, Nucleotides, and Base Analogs. Chem Rev 2016; 116:14379-14455. [PMID: 27960273 DOI: 10.1021/acs.chemrev.6b00209] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nucleoside, nucleotide, and base analogs have been in the clinic for decades to treat both viral pathogens and neoplasms. More than 20% of patients on anticancer chemotherapy have been treated with one or more of these analogs. This review focuses on the chemical synthesis and biology of anticancer nucleoside, nucleotide, and base analogs that are FDA-approved and in clinical development since 2000. We highlight the cellular biology and clinical biology of analogs, drug resistance mechanisms, and compound specificity towards different cancer types. Furthermore, we explore analog syntheses as well as improved and scale-up syntheses. We conclude with a discussion on what might lie ahead for medicinal chemists, biologists, and physicians as they try to improve analog efficacy through prodrug strategies and drug combinations.
Collapse
Affiliation(s)
- Jadd Shelton
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine , 1760 Haygood Drive, NE, Atlanta, Georgia 30322, United States
| | - Xiao Lu
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine , 1760 Haygood Drive, NE, Atlanta, Georgia 30322, United States
| | - Joseph A Hollenbaugh
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine , 1760 Haygood Drive, NE, Atlanta, Georgia 30322, United States
| | - Jong Hyun Cho
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine , 1760 Haygood Drive, NE, Atlanta, Georgia 30322, United States
| | - Franck Amblard
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine , 1760 Haygood Drive, NE, Atlanta, Georgia 30322, United States
| | - Raymond F Schinazi
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine , 1760 Haygood Drive, NE, Atlanta, Georgia 30322, United States
| |
Collapse
|