Network


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

Hotspot


Dive into the research topics where Marios Giannakis is active.

Publication


Featured researches published by Marios Giannakis.


Journal of Biological Chemistry | 2006

Molecular properties of adult mouse gastric and intestinal epithelial progenitors in their niches

Marios Giannakis; Thaddeus S. Stappenbeck; Jason C. Mills; Douglas G. Leip; Michael Lovett; Sandra W. Clifton; Joseph E. Ippolito; Jarret I. Glasscock; Manimozhiyan Arumugam; Michael R. Brent; Jeffrey I. Gordon

We have sequenced 36,641 expressed sequence tags from laser capture microdissected adult mouse gastric and small intestinal epithelial progenitors, obtaining 4031 and 3324 unique transcripts, respectively. Using Gene Ontology (GO) terms, each data set was compared with cDNA libraries from intact adult stomach and small intestine. Genes in GO categories enriched in progenitors were filtered against genes in GO categories represented in hematopoietic, neural, and embryonic stem cell transcriptomes and mapped onto transcription factor networks, plus canonical signal transduction and metabolic pathways. Wnt/β-catenin, phosphoinositide-3/Akt kinase, insulin-like growth factor-1, vascular endothelial growth factor, integrin, and γ-aminobutyric acid receptor signaling cascades, plus glycerolipid, fatty acid, and amino acid metabolic pathways are among those prominently represented in adult gut progenitors. The results reveal shared as well as distinctive features of adult gut stem cells when compared with other stem cell populations.


Nature Genetics | 2014

RNF43 is frequently mutated in colorectal and endometrial cancers

Marios Giannakis; Eran Hodis; Xinmeng Jasmine Mu; Mai Yamauchi; Joseph Rosenbluh; Kristian Cibulskis; Gordon Saksena; Michael S. Lawrence; Zhi Rong Qian; Reiko Nishihara; Eliezer M. Van Allen; William C. Hahn; Stacey Gabriel; Eric S. Lander; Gad Getz; Shuji Ogino; Charles S. Fuchs; Levi A. Garraway

We report somatic mutations of RNF43 in over 18% of colorectal adenocarcinomas and endometrial carcinomas. RNF43 encodes an E3 ubiquitin ligase that negatively regulates Wnt signaling. Truncating mutations of RNF43 are more prevalent in microsatellite-unstable tumors and show mutual exclusivity with inactivating APC mutations in colorectal adenocarcinomas. These results indicate that RNF43 is one of the most commonly mutated genes in colorectal and endometrial cancers.


Cell Reports | 2016

Genomic Correlates of Immune-Cell Infiltrates in Colorectal Carcinoma

Marios Giannakis; Xinmeng Jasmine Mu; Sachet A. Shukla; Zhi Rong Qian; Ofir Cohen; Reiko Nishihara; Samira Bahl; Yin Cao; Ali Amin-Mansour; Mai Yamauchi; Yasutaka Sukawa; Chip Stewart; Mara Rosenberg; Kosuke Mima; Kentaro Inamura; Katsuhiko Nosho; Jonathan A. Nowak; Michael S. Lawrence; Edward Giovannucci; Andrew T. Chan; Kimmie Ng; Jeffrey A. Meyerhardt; Eliezer M. Van Allen; Gad Getz; Stacey Gabriel; Eric S. Lander; Catherine J. Wu; Charles S. Fuchs; Shuji Ogino; Levi A. Garraway

Summary Large-scale genomic characterization of tumors from prospective cohort studies may yield new insights into cancer pathogenesis. We performed whole-exome sequencing of 619 incident colorectal cancers (CRCs) and integrated the results with tumor immunity, pathology, and survival data. We identified recurrently mutated genes in CRC, such as BCL9L, RBM10, CTCF, and KLF5, that were not previously appreciated in this disease. Furthermore, we investigated the genomic correlates of immune-cell infiltration and found that higher neoantigen load was positively associated with overall lymphocytic infiltration, tumor-infiltrating lymphocytes (TILs), memory T cells, and CRC-specific survival. The association with TILs was evident even within microsatellite-stable tumors. We also found positive selection of mutations in HLA genes and other components of the antigen-processing machinery in TIL-rich tumors. These results may inform immunotherapeutic approaches in CRC. More generally, this study demonstrates a framework for future integrative molecular epidemiology research in colorectal and other malignancies.


Gut | 2016

Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis

Kosuke Mima; Reiko Nishihara; Zhi Rong Qian; Yin Cao; Yasutaka Sukawa; Jonathan A. Nowak; Juhong Yang; Ruoxu Dou; Yohei Masugi; Mingyang Song; Aleksandar D. Kostic; Marios Giannakis; Susan Bullman; Danny A. Milner; Hideo Baba; Edward Giovannucci; Levi A. Garraway; Gordon J. Freeman; Glenn Dranoff; Wendy S. Garrett; Curtis Huttenhower; Matthew Meyerson; Jeffrey A. Meyerhardt; Andrew T. Chan; Charles S. Fuchs; Shuji Ogino

Objective Accumulating evidence links the intestinal microbiota and colorectal carcinogenesis. Fusobacterium nucleatum may promote colorectal tumour growth and inhibit T cell-mediated immune responses against colorectal tumours. Thus, we hypothesised that the amount of F. nucleatum in colorectal carcinoma might be associated with worse clinical outcome. Design We used molecular pathological epidemiology database of 1069 rectal and colon cancer cases in the Nurses’ Health Study and the Health Professionals Follow-up Study, and measured F. nucleatum DNA in carcinoma tissue. Cox proportional hazards model was used to compute hazard ratio (HR), controlling for potential confounders, including microsatellite instability (MSI, mismatch repair deficiency), CpG island methylator phenotype (CIMP), KRAS, BRAF, and PIK3CA mutations, and LINE-1 hypomethylation (low-level methylation). Results Compared with F. nucleatum-negative cases, multivariable HRs (95% CI) for colorectal cancer-specific mortality in F. nucleatum-low cases and F. nucleatum-high cases were 1.25 (0.82 to 1.92) and 1.58 (1.04 to 2.39), respectively, (p for trend=0.020). The amount of F. nucleatum was associated with MSI-high (multivariable odd ratio (OR), 5.22; 95% CI 2.86 to 9.55) independent of CIMP and BRAF mutation status, whereas CIMP and BRAF mutation were associated with F. nucleatum only in univariate analyses (p<0.001) but not in multivariate analysis that adjusted for MSI status. Conclusions The amount of F. nucleatum DNA in colorectal cancer tissue is associated with shorter survival, and may potentially serve as a prognostic biomarker. Our data may have implications in developing cancer prevention and treatment strategies through targeting GI microflora by diet, probiotics and antibiotics.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Helicobacter pylori evolution during progression from chronic atrophic gastritis to gastric cancer and its impact on gastric stem cells

Marios Giannakis; Swaine L. Chen; Sherif M. Karam; Lars Engstrand; Jeffrey I. Gordon

We have characterized the adaptations of Helicobacter pylori to a rarely captured event in the evolution of its impact on host biology—the transition from chronic atrophic gastritis (ChAG) to gastric adenocarcinoma—and defined the impact of these adaptations on an intriguing but poorly characterized interaction between this bacterium and gastric epithelial stem cells. Bacterial isolates were obtained from a single human host colonized with a single dominant strain before and after his progression from ChAG to gastric adenocarcinoma during a 4-year interval. Draft genome assemblies were generated from two isolates, one ChAG-associated, the other cancer-associated. The cancer-associated strain was less fit in a gnotobiotic transgenic mouse model of human ChAG and better able to establish itself within a mouse gastric epithelial progenitor-derived cell line (mGEP) that supports bacterial attachment. GeneChip-based comparisons of the transcriptomes of mGEPs and a control mouse gastric epithelial cell line revealed that, upon infection, the cancer-associated strain regulates expression of GEP-associated signaling and metabolic pathways, and tumor suppressor genes associated with development of gastric cancer in humans, in a manner distinct from the ChAG-associated isolate. The effects on GEP metabolic pathways, some of which were confirmed in gnotobiotic mice, together with observed changes in the bacterial transcriptome are predicted to support aspects of an endosymbiosis between this microbe and gastric stem cells. These results provide insights about how H. pylori may adapt to and influence stem cell biology and how its intracellular residency could contribute to gastric tumorigenesis.


Nature Genetics | 2016

The genomic landscape and evolution of endometrial carcinoma progression and abdominopelvic metastasis

William J. Gibson; Erling A. Hoivik; Mari K. Halle; Amaro Taylor-Weiner; Andrew D. Cherniack; Anna Berg; Frederik Holst; Travis I. Zack; Henrica Maria Johanna Werner; Kjersti Mangseth Staby; Mara Rosenberg; Ingunn Stefansson; Kanthida Kusonmano; Aaron Chevalier; Karen Klepsland Mauland; Jone Trovik; Camilla Krakstad; Marios Giannakis; Eran Hodis; Kathrine Woie; Line Bjørge; Olav Karsten Vintermyr; Jeremiah Wala; Michael S. Lawrence; Gad Getz; Scott L. Carter; Rameen Beroukhim; Helga B. Salvesen

Recent studies have detailed the genomic landscape of primary endometrial cancers, but the evolution of these cancers into metastases has not been characterized. We performed whole-exome sequencing of 98 tumor biopsies including complex atypical hyperplasias, primary tumors and paired abdominopelvic metastases to survey the evolutionary landscape of endometrial cancer. We expanded and reanalyzed The Cancer Genome Atlas (TCGA) data, identifying new recurrent alterations in primary tumors, including mutations in the estrogen receptor cofactor gene NRIP1 in 12% of patients. We found that likely driver events were present in both primary and metastatic tissue samples, with notable exceptions such as ARID1A mutations. Phylogenetic analyses indicated that the sampled metastases typically arose from a common ancestral subclone that was not detected in the primary tumor biopsy. These data demonstrate extensive genetic heterogeneity in endometrial cancers and relative homogeneity across metastatic sites.


Cell Reports | 2016

Erratum: Genomic Correlates of Immune-Cell Infiltrates in Colorectal Carcinoma (Cell Reports (2016) 15(4) (857–865) (S2211124716303643) (10.1016/j.celrep.2016.03.075))

Marios Giannakis; Xinmeng Jasmine Mu; Sachet A. Shukla; Zhi Rong Qian; Ofir Cohen; Reiko Nishihara; Samira Bahl; Yin Cao; Ali Amin-Mansour; Mai Yamauchi; Yasutaka Sukawa; Chip Stewart; Mara Rosenberg; Kosuke Mima; Kentaro Inamura; Katsuhiko Nosho; Jonathan A. Nowak; Michael S. Lawrence; Edward Giovannucci; Andrew T. Chan; Kimmie Ng; Jeffrey A. Meyerhardt; Eliezer M. Van Allen; Gad Getz; Stacey Gabriel; Eric S. Lander; Catherine J. Wu; Charles S. Fuchs; Shuji Ogino; Levi A. Garraway

Marios Giannakis, Xinmeng Jasmine Mu, Sachet A. Shukla, Zhi Rong Qian, Ofir Cohen, Reiko Nishihara, Samira Bahl, Yin Cao, Ali Amin-Mansour, Mai Yamauchi, Yasutaka Sukawa, Chip Stewart, Mara Rosenberg, Kosuke Mima, Kentaro Inamura, Katsuhiko Nosho, Jonathan A. Nowak, Michael S. Lawrence, Edward L. Giovannucci, Andrew T. Chan, Kimmie Ng, Jeffrey A. Meyerhardt, Eliezer M. Van Allen, Gad Getz, Stacey B. Gabriel, Eric S. Lander, Catherine J. Wu, Charles S. Fuchs, Shuji Ogino,* and Levi A. Garraway* *Correspondence: [email protected] (S.O.), [email protected] (L.A.G.) http://dx.doi.org/10.1016/j.celrep.2016.10.009


Cancer immunology research | 2015

Long-term Benefit of PD-L1 Blockade in Lung Cancer Associated with JAK3 Activation.

Eliezer M. Van Allen; Hadrien G Golay; Yan Liu; Shohei Koyama; Karrie Wong; Amaro Taylor-Weiner; Marios Giannakis; Maegan Harden; Vanesa Rojas-Rudilla; Aaron Chevalier; Tran C. Thai; Christine A. Lydon; Stacy L. Mach; Ada G. Avila; Joshua A. Wong; Alexandra R. Rabin; Joshua Helmkamp; Lynette M. Sholl; Scott L. Carter; Geoffrey R. Oxnard; Pasi A. Jänne; Gad Getz; Neal I. Lindeman; Peter S. Hammerman; Levi A. Garraway; F. Stephen Hodi; Scott J. Rodig; Glenn Dranoff; Kwok-Kin Wong; David A. Barbie

Van Allen, Golay, Liu, and colleagues genomically profiled tumor and germline samples from a patient with activating JAK3 variants who achieved long-term clinical benefit from anti–PD-L1 therapy, suggesting that alterations in JAK signaling may be immunogenomic modulators of response to PD-L1 immunotherapy. PD-1 immune checkpoint blockade occasionally results in durable clinical responses in advanced metastatic cancers. However, mechanism-based predictors of response to this immunotherapy remain incompletely characterized. We performed comprehensive genomic profiling on a tumor and germline sample from a patient with refractory lung adenocarcinoma who achieved marked long-term clinical benefit from anti–PD-L1 therapy. We discovered activating somatic and germline amino acid variants in JAK3 that promoted PD-L1 induction in lung cancer cells and in the tumor immune microenvironment. These findings suggest that genomic alterations that deregulate cytokine receptor signal transduction could contribute to PD-L1 activation and engagement of the PD-1 immune checkpoint in lung cancer. Cancer Immunol Res; 3(8); 855–63. ©2015 AACR.


Genome Medicine | 2016

The impact of tumor profiling approaches and genomic data strategies for cancer precision medicine

Andrea Garofalo; Lynette M. Sholl; Brendan Reardon; Amaro Taylor-Weiner; Ali Amin-Mansour; Diana Miao; David R. Liu; Nelly Oliver; Laura E. MacConaill; Matthew Ducar; Vanesa Rojas-Rudilla; Marios Giannakis; Arezou A. Ghazani; Stacy W. Gray; Pasi A. Jänne; Judy Garber; Steve Joffe; Neal I. Lindeman; Nikhil Wagle; Levi A. Garraway; Eliezer M. Van Allen

BackgroundThe diversity of clinical tumor profiling approaches (small panels to whole exomes with matched or unmatched germline analysis) may engender uncertainty about their benefits and liabilities, particularly in light of reported germline false positives in tumor-only profiling and use of global mutational and/or neoantigen data. The goal of this study was to determine the impact of genomic analysis strategies on error rates and data interpretation across contexts and ancestries.MethodsWe modeled common tumor profiling modalities—large (n = 300 genes), medium (n = 48 genes), and small (n = 15 genes) panels—using clinical whole exomes (WES) from 157 patients with lung or colon adenocarcinoma. We created a tumor-only analysis algorithm to assess germline false positive rates, the impact of patient ancestry on tumor-only results, and neoantigen detection.ResultsAfter optimizing a germline filtering strategy, the germline false positive rate with tumor-only large panel sequencing was 14 % (144/1012 variants). For patients whose tumor-only results underwent molecular pathologist review (n = 91), 50/54 (93 %) false positives were correctly interpreted as uncertain variants. Increased germline false positives were observed in tumor-only sequencing of non-European compared with European ancestry patients (p < 0.001; Fisher’s exact) when basic germline filtering approaches were used; however, the ExAC database (60,706 germline exomes) mitigated this disparity (p = 0.53). Matched and unmatched large panel mutational load correlated with WES mutational load (r2 = 0.99 and 0.93, respectively; p < 0.001). Neoantigen load also correlated (r2 = 0.80; p < 0.001), though WES identified a broader spectrum of neoantigens. Small panels did not predict mutational or neoantigen load.ConclusionsLarge tumor-only targeted panels are sufficient for most somatic variant identification and mutational load prediction if paired with expanded germline analysis strategies and molecular pathologist review. Paired germline sequencing reduced overall false positive mutation calls and WES provided the most neoantigens. Without patient-matched germline data, large germline databases are needed to minimize false positive mutation calling and mitigate ethnic disparities.


Gut | 2018

Integrative analysis of exogenous, endogenous, tumour and immune factors for precision medicine

Shuji Ogino; Jonathan A. Nowak; Tsuyoshi Hamada; Amanda I. Phipps; Ulrike Peters; Danny A. Milner; Edward Giovannucci; Reiko Nishihara; Marios Giannakis; Wendy S. Garrett; Mingyang Song

Immunotherapy strategies targeting immune checkpoints such as the CTLA4 and CD274 (programmed cell death 1 ligand 1, PD-L1)/PDCD1 (programmed cell death 1, PD-1) T-cell coreceptor pathways are revolutionising oncology. The approval of pembrolizumab use for solid tumours with high-level microsatellite instability or mismatch repair deficiency by the US Food and Drug Administration highlights promise of precision immuno-oncology. However, despite evidence indicating influences of exogenous and endogenous factors such as diet, nutrients, alcohol, smoking, obesity, lifestyle, environmental exposures and microbiome on tumour-immune interactions, integrative analyses of those factors and immunity lag behind. Immune cell analyses in the tumour microenvironment have not adequately been integrated into large-scale studies. Addressing this gap, the transdisciplinary field of molecular pathological epidemiology (MPE) offers research frameworks to integrate tumour immunology into population health sciences, and link the exposures and germline genetics (eg, HLA genotypes) to tumour and immune characteristics. Multilevel research using bioinformatics, in vivo pathology and omics (genomics, epigenomics, transcriptomics, proteomics and metabolomics) technologies is possible with use of tissue, peripheral blood circulating cells, cell-free plasma, stool, sputum, urine and other body fluids. This immunology-MPE model can synergise with experimental immunology, microbiology and systems biology. GI neoplasms represent exemplary diseases for the immunology-MPE model, given rich microbiota and immune tissues of intestines, and the well-established carcinogenic role of intestinal inflammation. Proof-of-principle studies on colorectal cancer provided insights into immunomodulating effects of aspirin, vitamin D, inflammatory diets and omega-3 polyunsaturated fatty acids. The integrated immunology-MPE model can contribute to better understanding of environment-tumour-immune interactions, and effective immunoprevention and immunotherapy strategies for precision medicine.

Collaboration


Dive into the Marios Giannakis's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge