Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mancang Gu is active.

Publication


Featured researches published by Mancang Gu.


Oncotarget | 2016

Tumor LINE-1 methylation level and colorectal cancer location in relation to patient survival

Kosuke Mima; Jonathan A. Nowak; Zhi Rong Qian; Yin Cao; Mingyang Song; Yohei Masugi; Yan Shi; Annacarolina da Silva; Mancang Gu; Wanwan Li; Tsuyoshi Hamada; Xuehong Zhang; Kana Wu; Jeffrey A. Meyerhardt; Hideo Baba; Edward Giovannucci; Andrew T. Chan; Charles S. Fuchs; Shuji Ogino; Reiko Nishihara

Colorectal tumors arise with genomic and epigenomic alterations through interactions between neoplastic cells, immune cells, and microbiota that vary along the proximal to distal axis of colorectum. Long interspersed nucleotide element-1 (LINE-1) hypomethylation in colorectal cancer has been associated with worse clinical outcome. Utilizing 1,317 colon and rectal carcinoma cases in two U.S.-nationwide prospective cohort studies, we examined patient survival according to LINE-1 methylation level stratified by tumor location. Cox proportional hazards model was used to assess a statistical interaction between LINE-1 methylation level and tumor location in colorectal cancer-specific mortality analysis, controlling for potential confounders including microsatellite instability, CpG island methylator phenotype, and KRAS, BRAF, and PIK3CA mutations. A statistically significant interaction was found between LINE-1 methylation level and tumor location in colorectal cancer-specific mortality analysis (Pinteraction = 0.011). The association of LINE-1 hypomethylation with higher colorectal cancer-specific mortality was stronger in proximal colon cancers (multivariable hazard ratio [HR], 1.66; 95% confidence interval [CI], 1.21 to 2.28) than in distal colon cancers (multivariable HR, 1.18; 95% CI, 0.81 to 1.72) or rectal cancers (multivariable HR, 0.87; 95% CI, 0.57 to 1.34). Our data suggest the interactive effect of LINE-1 methylation level and colorectal cancer location on clinical outcome.


Cancer immunology research | 2016

MicroRNA let-7, T Cells, and Patient Survival in Colorectal Cancer

Ruoxu Dou; Reiko Nishihara; Yin Cao; Tsuyoshi Hamada; Kosuke Mima; Atsuhiro Masuda; Yohei Masugi; Yan Shi; Mancang Gu; Wanwan Li; Annacarolina da Silva; Katsuhiko Nosho; Xuehong Zhang; Jeffrey A. Meyerhardt; Edward Giovannucci; Andrew T. Chan; Charles S. Fuchs; Zhi Rong Qian; Shuji Ogino

The population-based data presented in this study support a possible role for microRNA let-7a in the suppression of antitumor immunity in colorectal cancer patients. This may have implications for expanding the benefit of immunotherapy through targeting microRNAs. Experimental evidence suggests that the let-7 family of noncoding RNAs suppresses adaptive immune responses, contributing to immune evasion by the tumor. We hypothesized that the amount of let-7a and let-7b expression in colorectal carcinoma might be associated with limited T-lymphocyte infiltrates in the tumor microenvironment and worse clinical outcome. Utilizing the molecular pathological epidemiology resources of 795 rectal and colon cancers in two U.S.-nationwide prospective cohort studies, we measured tumor-associated let-7a and let-7b expression levels by quantitative reverse-transcription PCR, and CD3+, CD8+, CD45RO (PTPRC)+, and FOXP3+ cell densities by tumor tissue microarray immunohistochemistry and computer-assisted image analysis. Logistic regression analysis and Cox proportional hazards regression were used to assess associations of let-7a (and let-7b) expression (quartile predictor variables) with T-cell densities (binary outcome variables) and mortality, respectively, controlling for tumor molecular features, including microsatellite instability, CpG island methylator phenotype, LINE-1 methylation, and KRAS, BRAF, and PIK3CA mutations. Compared with cases in the lowest quartile of let-7a expression, those in the highest quartile were associated with lower densities of CD3+ [multivariate odds ratio (OR), 0.40; 95% confidence interval (CI), 0.23–0.67; Ptrend = 0.003] and CD45RO+ cells (multivariate OR, 0.31; 95% CI, 0.17–0.58; Ptrend = 0.0004), and higher colorectal cancer-specific mortality (multivariate hazard ratio, 1.82; 95% CI, 1.42–3.13; Ptrend = 0.001). In contrast, let-7b expression was not significantly associated with T-cell density or colorectal cancer prognosis. Our data support the role of let-7a in suppressing antitumor immunity in colorectal cancer and suggest let-7a as a potential target of immunotherapy. Cancer Immunol Res; 4(11); 927–35. ©2016 AACR.


Cancer immunology research | 2017

Tumor PDCD1LG2 (PD-L2) Expression and the Lymphocytic Reaction to Colorectal Cancer

Yohei Masugi; Reiko Nishihara; Tsuyoshi Hamada; Mingyang Song; Annacarolina da Silva; Keisuke Kosumi; Mancang Gu; Yan Shi; Wanwan Li; Li Liu; Daniel Nevo; Kentaro Inamura; Yin Cao; Xiaoyun Liao; Katsuhiko Nosho; Andrew T. Chan; Marios Giannakis; Adam J. Bass; F. Stephen Hodi; Gordon J. Freeman; Scott J. Rodig; Charles S. Fuchs; Zhi Rong Qian; Jonathan A. Nowak; Shuji Ogino

Colorectal tumor PDCD1LG2 (PD-L2) expression was inversely associated with Crohns-like lymphoid reactions, which are a key adaptive antitumor response in colorectal carcinogenesis. This population data can inform the development of immunotherapy strategies targeting immune checkpoint mechanisms. Expression of the immune checkpoint ligand CD274 (programmed cell death 1 ligand 1, PD-L1, from gene CD274) contributes to suppression of antitumor T cell–mediated immune response in various tumor types. However, the role of PDCD1LG2 (PD-L2, CD273, from gene PDCD1LG2) in the tumor microenvironment remains unclear. We hypothesized that tumor PDCD1LG2 expression might be inversely associated with lymphocytic reactions to colorectal cancer. We examined tumor PDCD1LG2 expression by IHC in 823 colon and rectal carcinoma cases within two U.S.-nationwide cohort studies and categorized tumors into quartiles according to the percentage of PDCD1LG2–expressing carcinoma cells. We conducted multivariable ordinal logistic regression analysis to assess the associations of tumor PDCD1LG2 expression with Crohn-like lymphoid reaction, peritumoral lymphocytic reaction, intratumoral periglandular reaction, or tumor-infiltrating lymphocytes, controlling for potential confounders, including microsatellite instability, CpG island methylator phenotype, long-interspersed nucleotide element-1 methylation, and KRAS, BRAF, and PIK3CA mutations. Tumor PDCD1LG2 expression was inversely associated with Crohn-like lymphoid reaction (Ptrend = 0.0003). For a unit increase in the three-tiered ordinal categories of Crohn-like lymphoid reaction, a multivariable OR in the highest (vs. lowest) quartile of the percentage of PDCD1LG2–expressing tumor cells was 0.38 (95% confidence interval, 0.22–0.67). Tumor PDCD1LG2 expression was not associated with peritumoral lymphocytic reaction, intratumoral periglandular reaction, tumor-infiltrating lymphocytes, or patient survival (Ptrend > 0.13). Thus, tumor PDCD1LG2 expression is inversely associated with Crohn-like lymphoid reaction to colorectal cancer, suggesting a possible role of PDCD1LG2-expressing tumor cells in inhibiting the development of tertiary lymphoid tissues during colorectal carcinogenesis. Cancer Immunol Res; 5(11); 1046–55. ©2017 AACR.


JAMA Oncology | 2017

Association of Alterations in Main Driver Genes With Outcomes of Patients With Resected Pancreatic Ductal Adenocarcinoma

Zhi Rong Qian; Douglas A. Rubinson; Jonathan A. Nowak; Vicente Morales-Oyarvide; Richard F. Dunne; Margaret M. Kozak; Marisa W. Welch; Lauren K. Brais; Annacarolina da Silva; Tingting Li; Wanwan Li; Atsuhiro Masuda; Juhong Yang; Yan Shi; Mancang Gu; Yohei Masugi; Justin L. Bui; Caitlin Zellers; Chen Yuan; Ana Babic; Natalia Khalaf; Andrew J. Aguirre; Kimmie Ng; Rebecca A. Miksad; Andrea J. Bullock; Daniel T. Chang; Jennifer F. Tseng; Thomas E. Clancy; David C. Linehan; Jennifer J. Findeis-Hosey

Importance Although patients with resected pancreatic adenocarcinoma are at high risk for disease recurrence, few biomarkers are available to inform patient outcomes. Objective To evaluate the alterations of the 4 main driver genes in pancreatic adenocarcinoma and patient outcomes after cancer resection. Design, Setting, and Participants This study analyzed protein expression and DNA alterations for the KRAS, CDKN2A, SMAD4, and TP53 genes by immunohistochemistry and next-generation sequencing in formalin-fixed, paraffin-embedded tumors in 356 patients with resected pancreatic adenocarcinoma who were treated at the Dana-Farber/Brigham and Women’s Cancer Center (October 26, 2002, to May 21, 2012), University of Rochester Medical Center (March 1, 2006, to November 1, 2013), or Stanford Cancer Institute (September 26, 1995, to May 22, 2013). Associations of driver gene alterations with disease-free survival (DFS) and overall survival (OS) were evaluated using Cox proportional hazards regression with estimation of hazard ratios (HRs) and 95% CIs and adjustment for age, sex, tumor characteristics, institution, and perioperative treatment. Data were collected September 9, 2012, to June 28, 2016, and analyzed December 17, 2016, to March 14, 2017. Main Outcomes and Measures The DFS and OS among patients with resected pancreatic adenocarcinoma. Results Of the 356 patients studied, 191 (53.7%) were men and 165 (46.3%) were women, with a median (interquartile range [IQR]) age of 67 (59.0-73.5) years. Patients with KRAS mutant tumors had worse DFS (median [IQR], 12.3 [6.7 -27.2] months) and OS (20.3 [11.3-38.3] months) compared with patients with KRAS wild-type tumors (DFS, 16.2 [8.9-30.5] months; OS, 38.6 [16.6-63.1] months) and had 5-year OS of 13.0% vs 30.2%. Particularly poor outcomes were identified in patients with KRAS G12D-mutant tumors, who had a median (IQR) OS of 15.3 (9.8-32.7) months. Patients whose tumors lacked CDKN2A expression had worse DFS (median, 11.5 [IQR, 6.2-24.5] months) and OS (19.7 [10.9-37.1] months) compared with patients who had intact CDKN2A (DFS, 14.8 [8.2-30.5] months; OS, 24.6 [14.1-44.6] months). The molecular status of SMAD4 was not associated with DFS or OS, whereas TP53 status was associated only with shorter DFS (HR, 1.33; 95% CI, 1.02-1.75; P = .04). Patients had worse DFS and OS if they had a greater number of altered driver genes. Compared with patients with 0 to 2 altered genes, those with 4 altered genes had worse DFS (HR, 1.79 [95% CI, 1.24-2.59; P = .002]) and OS (HR, 1.38 [95% CI, 0.98-1.94; P = .06]). Five-year OS was 18.4% for patients with 0 to 2 gene alterations, 14.1% for those with 3 alterations, and 8.2% for those with 4 alterations. Conclusions and Relevance Patient outcomes are associated with alterations of the 4 main driver genes in resected pancreatic adenocarcinoma.


Genetics in Medicine | 2018

Germline cancer susceptibility gene variants, somatic second hits, and survival outcomes in patients with resected pancreatic cancer

Matthew B. Yurgelun; Anu Chittenden; Vicente Morales-Oyarvide; Douglas A. Rubinson; Richard F. Dunne; Margaret M. Kozak; Zhi Rong Qian; Marisa W. Welch; Lauren K. Brais; Annacarolina da Silva; Justin L. Bui; Chen Yuan; Tingting Li; Wanwan Li; Atsuhiro Masuda; Mancang Gu; Andrea J. Bullock; Daniel T. Chang; Thomas E. Clancy; David C. Linehan; Jennifer J. Findeis-Hosey; Leona A. Doyle; Aaron R. Thorner; Matthew Ducar; Bruce M. Wollison; Natalia Khalaf; Kimberly Perez; Sapna Syngal; Andrew J. Aguirre; William C. Hahn

PurposeGermline variants in double-strand DNA damage repair (dsDDR) genes (e.g., BRCA1/2) predispose to pancreatic adenocarcinoma (PDAC) and may predict sensitivity to platinum-based chemotherapy and poly(ADP) ribose polymerase (PARP) inhibitors. We sought to determine the prevalence and significance of germline cancer susceptibility gene variants in PDAC with paired somatic and survival analyses.MethodsUsing a customized next-generation sequencing panel, germline/somatic DNA was analyzed from 289 patients with resected PDAC ascertained without preselection for high-risk features (e.g., young age, personal/family history). All identified variants were assessed for pathogenicity. Outcomes were analyzed using multivariable-adjusted Cox proportional hazards regression.ResultsWe found that 28/289 (9.7%; 95% confidence interval [CI] 6.5–13.7%) patients carried pathogenic/likely pathogenic germline variants, including 21 (7.3%) dsDDR gene variants (3 BRCA1, 4 BRCA2, 14 other dsDDR genes [ATM, BRIP1, CHEK2, NBN, PALB2, RAD50, RAD51C]), 3 Lynch syndrome, and 4 other genes (APC p.I1307K, CDKN2A, TP53). Somatic sequencing and immunohistochemistry identified second hits in the tumor in 12/27 (44.4%) patients with germline variants (1 failed sequencing). Compared with noncarriers, patients with germline dsDDR gene variants had superior overall survival (hazard ratio [HR] 0.54; 95% CI 0.30–0.99; P = 0.05).ConclusionNearly 10% of PDAC patients harbor germline variants, although the majority lack somatic second hits, the therapeutic significance of which warrants further study.


Clinical Gastroenterology and Hepatology | 2018

Diets That Promote Colon Inflammation Associate With Risk of Colorectal Carcinomas That Contain Fusobacterium nucleatum

Li Liu; Fred K. Tabung; Xuehong Zhang; Jonathan A. Nowak; Zhi Rong Qian; Tsuyoshi Hamada; Daniel Nevo; Susan Bullman; Kosuke Mima; Keisuke Kosumi; Annacarolina da Silva; Mingyang Song; Yin Cao; Tyler S. Twombly; Yan Shi; Hongli Liu; Mancang Gu; Hideo Koh; Wanwan Li; Chunxia Du; Yang Chen; Chenxi Li; Wenbin Li; Raaj S. Mehta; Kana Wu; Molin Wang; Aleksander D. Kostic; Marios Giannakis; Wendy S. Garrett; Curtis Hutthenhower

Background & Aims Specific nutritional components are likely to induce intestinal inflammation, which is characterized by increased levels of interleukin 6 (IL6), C‐reactive protein (CRP), and tumor necrosis factor–receptor superfamily member 1B (TNFRSF1B) in the circulation and promotes colorectal carcinogenesis. The inflammatory effects of a diet can be estimated based on an empiric dietary inflammatory pattern (EDIP) score, calculated based on intake of 18 foods associated with plasma levels of IL6, CRP, and TNFRSF1B. An inflammatory environment in the colon (based on increased levels of IL6, CRP, and TNFRSF1B in peripheral blood) contributes to impairment of the mucosal barrier and altered immune cell responses, affecting the composition of the intestinal microbiota. Colonization by Fusobacterium nucleatum has been associated with the presence and features of colorectal adenocarcinoma. We investigated the association between diets that promote inflammation (based on EDIP score) and colorectal cancer subtypes classified by level of F nucleatum in the tumor microenvironment. Methods We calculated EDIP scores based on answers to food frequency questionnaires collected from participants in the Nurses’ Health Study (through June 1, 2012) and the Health Professionals Follow‐up Study (through January 31, 2012). Participants in both cohorts reported diagnoses of rectal or colon cancer in biennial questionnaires; deaths from unreported colorectal cancer cases were identified through the National Death Index and next of kin. Colorectal tumor tissues were collected from hospitals where the patients underwent tumor resection and F nucleatum DNA was quantified by a polymerase chain reaction assay. We used multivariable duplication‐method Cox proportional hazard regression to assess the associations of EDIP scores with risks of colorectal cancer subclassified by F nucleatum status. Results During 28 years of follow‐up evaluation of 124,433 participants, we documented 951 incident cases of colorectal carcinoma with tissue F nucleatum data. Higher EDIP scores were associated with increased risk of F nucleatum–positive colorectal tumors (Ptrend = .03); for subjects in the highest vs lowest EDIP score tertiles, the hazard ratio for F nucleatum–positive colorectal tumors was 1.63 (95% CI, 1.03–2.58). EDIP scores did not associate with F nucleatum–negative tumors (Ptrend = .44). High EDIP scores associated with proximal F nucleatum–positive colorectal tumors but not with proximal F nucleatum–negative colorectal tumors (Pheterogeneity = .003). Conclusions Diets that may promote intestinal inflammation, based on EDIP score, are associated with increased risk of F nucleatum–positive colorectal carcinomas, but not carcinomas that do not contain these bacteria. These findings indicate that diet‐induced intestinal inflammation alters the gut microbiome to contribute to colorectal carcinogenesis; nutritional interventions might be used in precision medicine and cancer prevention.


Oncotarget | 2017

Aspirin exerts high anti-cancer activity in PIK3CA -mutant colon cancer cells

Mancang Gu; Reiko Nishihara; Yang Chen; Wanwan Li; Yan Shi; Yohei Masugi; Tsuyoshi Hamada; Keisuke Kosumi; Li Liu; Annacarolina da Silva; Jonathan A. Nowak; Tyler S. Twombly; Chunxia Du; Hideo Koh; Wenbin Li; Jeffrey A. Meyerhardt; Brian M. Wolpin; Marios Giannakis; Andrew J. Aguirre; Adam J. Bass; David A. Drew; Andrew T. Chan; Charles S. Fuchs; Zhi Rong Qian; Shuji Ogino

Evidence suggests that nonsteroidal anti-inflammatory drug aspirin (acetylsalicylic acid) may improve patient survival in PIK3CA-mutant colorectal carcinoma, but not in PIK3CA-wild-type carcinoma. However, whether aspirin directly influences the viability of PIK3CA-mutant colon cancer cells is poorly understood. We conducted in vitro experiments to test our hypothesis that the anti-proliferative activity of aspirin might be stronger for PIK3CA-mutant colon cancer cells than for PIK3CA-wild-type colon cancer cells. We measured the anti-proliferative effect of aspirin at physiologic concentrations in seven PIK3CA-mutant and six PIK3CA-wild-type human colon cancer cell lines. After exposure to aspirin, the apoptotic index and cell cycle phase of colon cancer cells were assessed. In addition, the effect of aspirin was examined in parental SW48 cells and SW48 cell clones with individual knock-in PIK3CA mutations of either c.3140A>G (p.H1047R) or c.1633G>A (p.E545K). Aspirin induced greater dose-dependent loss of cell viability in PIK3CA-mutant cells than in PIK3CA-wild-type cells after treatment for 48 and 72 hours. Aspirin treatment also led to higher proportions of apoptotic cells and G0/G1 phase arrest in PIK3CA-mutant cells than in PIK3CA-wild-type cells. Aspirin treatment of isogenic SW48 cells carrying a PIK3CA mutation, either c.3140A>G (p.H1047R) or c.1633G>A (p. E545K), resulted in a more significant loss of cell viability compared to wild-type controls. Our findings indicate that aspirin causes cell cycle arrest, induces apoptosis, and leads to loss of cell viability more profoundly in PIK3CA-mutated colon cancer cells than in PIK3CA-wild-type colon cancer cells. These findings support the use of aspirin to treat patients with PIK3CA-mutant colon cancer.


Pancreas | 2017

Cell Cycle Protein Expression in Neuroendocrine Tumors: Association of CDK4/CDK6, CCND1, and Phosphorylated Retinoblastoma Protein with Proliferative Index

Yan Shi; Zhi Rong Qian; Sui Zhang; Wanwan Li; Yohei Masugi; Tingting Li; Jennifer A. Chan; Juhong Yang; Annacarolina da Silva; Mancang Gu; Li Liu; Tsuyoshi Hamada; Keisuke Kosumi; Trevor Dutton; Lauren K. Brais; Reiko Nishihara; Charles S. Fuchs; Shuji Ogino; Matthew H. Kulke

Objectives Dysregulation of the cell cycle has been observed and implicated as an etiologic factor in a range of human malignancies, but remains relatively unstudied in neuroendocrine tumors (NETs). We evaluated expression of key proteins involved in cell cycle regulation in a large cohort of NETs. Methods We evaluated immunohistochemical expression of CDKN1B, CDKN1A, CDKN2A, CDK2, CDK4, CDK6, cyclin D1, cyclin E1, and phosphorylated retinoblastoma protein (phospho-RB1) in a cohort of 267 patients with NETs. We then explored associations between cell cycle protein expression, mutational status, histologic features, and overall survival. Results We found that high expression of CDK4, CDK6, CCND1, and phospho-RB1 was associated with higher proliferative index, as defined by MKI67. We additionally observed a trend toward shorter overall survival associated with low expression of CDKN1B. This association seemed strongest in SINETs (multivariate hazards ratio, 2.04; 95% confidence interval, 1.06–3.93; P = 0.03). We found no clear association between CDKN1B mutation and protein expression. Conclusions Our results suggest that dysregulation and activation of the CDK4/CDK6-CCND1-phospho-RB1 axis is associated with higher proliferative index in NETs. Investigation of the therapeutic potential of CDK4/CDK6 inhibitors in higher grade NETs is warranted.


Clinical and translational gastroenterology | 2016

Fusobacterium nucleatum in Colorectal Carcinoma Tissue According to Tumor Location

Kosuke Mima; Yin Cao; Andrew T. Chan; Zhi Rong Qian; Jonathan A. Nowak; Yohei Masugi; Yan Shi; Mingyang Song; Annacarolina da Silva; Mancang Gu; Wanwan Li; Tsuyoshi Hamada; Keisuke Kosumi; Akiko Hanyuda; Li Liu; Aleksandar D. Kostic; Marios Giannakis; Susan Bullman; Caitlin A. Brennan; Danny A. Milner; Hideo Baba; Levi A. Garraway; Jeffrey A. Meyerhardt; Wendy S. Garrett; Curtis Huttenhower; Matthew Meyerson; Edward Giovannucci; Charles S. Fuchs; Reiko Nishihara; Shuji Ogino


Journal of Clinical Oncology | 2017

Aspirin use and colorectal cancer survival according to tumor CD274 (programmed cell death 1 ligand 1) expression status

Tsuyoshi Hamada; Yin Cao; Zhi Rong Qian; Yohei Masugi; Jonathan A. Nowak; Juhong Yang; Mingyang Song; Kosuke Mima; Keisuke Kosumi; Li Liu; Yan Shi; Annacarolina da Silva; Mancang Gu; Wanwan Li; NaNa Keum; Xuehong Zhang; Kana Wu; Jeffrey A. Meyerhardt; Edward Giovannucci; Marios Giannakis; Scott J. Rodig; Gordon J. Freeman; Daniel Nevo; Molin Wang; Andrew T. Chan; Charles S. Fuchs; Reiko Nishihara; Shuji Ogino

Collaboration


Dive into the Mancang Gu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jonathan A. Nowak

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge