Network


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

Hotspot


Dive into the research topics where Ali Amin-Mansour is active.

Publication


Featured researches published by Ali Amin-Mansour.


The New England Journal of Medicine | 2014

Response and Acquired Resistance to Everolimus in Anaplastic Thyroid Cancer

Nikhil Wagle; Brian C. Grabiner; Eliezer M. Van Allen; Ali Amin-Mansour; Amaro Taylor-Weiner; Mara Rosenberg; Nathanael S. Gray; Justine A. Barletta; Yanan Guo; Scott J. Swanson; Daniel T. Ruan; Glenn J. Hanna; Robert I. Haddad; Gad Getz; David J. Kwiatkowski; Scott L. Carter; David M. Sabatini; Pasi A. Jänne; Levi A. Garraway; Jochen H. Lorch

Everolimus, an inhibitor of the mammalian target of rapamycin (mTOR), is effective in treating tumors harboring alterations in the mTOR pathway. Mechanisms of resistance to everolimus remain undefined. Resistance developed in a patient with metastatic anaplastic thyroid carcinoma after an extraordinary 18-month response. Whole-exome sequencing of pretreatment and drug-resistant tumors revealed a nonsense mutation in TSC2, a negative regulator of mTOR, suggesting a mechanism for exquisite sensitivity to everolimus. The resistant tumor also harbored a mutation in MTOR that confers resistance to allosteric mTOR inhibition. The mutation remains sensitive to mTOR kinase inhibitors.


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.


Nature Biotechnology | 2014

Assessing the clinical utility of cancer genomic and proteomic data across tumor types

Yuan Yuan; Eliezer M. Van Allen; Larsson Omberg; Nikhil Wagle; Ali Amin-Mansour; Artem Sokolov; Lauren Averett Byers; Yanxun Xu; Kenneth R. Hess; Lixia Diao; Leng Han; Xuelin Huang; Michael S. Lawrence; John N. Weinstein; Josh Stuart; Gordon B. Mills; Levi A. Garraway; Adam A. Margolin; Gad Getz; Han Liang

Molecular profiling of tumors promises to advance the clinical management of cancer, but the benefits of integrating molecular data with traditional clinical variables have not been systematically studied. Here we retrospectively predict patient survival using diverse molecular data (somatic copy-number alteration, DNA methylation and mRNA, microRNA and protein expression) from 953 samples of four cancer types from The Cancer Genome Atlas project. We find that incorporating molecular data with clinical variables yields statistically significantly improved predictions (FDR < 0.05) for three cancers but those quantitative gains were limited (2.2–23.9%). Additional analyses revealed little predictive power across tumor types except for one case. In clinically relevant genes, we identified 10,281 somatic alterations across 12 cancer types in 2,928 of 3,277 patients (89.4%), many of which would not be revealed in single-tumor analyses. Our study provides a starting point and resources, including an open-access model evaluation platform, for building reliable prognostic and therapeutic strategies that incorporate molecular data.


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


Immunity | 2017

Loss of PTEN Is Associated with Resistance to Anti-PD-1 Checkpoint Blockade Therapy in Metastatic Uterine Leiomyosarcoma

Suzanne George; Diana Miao; George D. Demetri; Scott J. Rodig; Sachet A. Shukla; Mikel Lipschitz; Ali Amin-Mansour; Chandrajit P. Raut; Scott L. Carter; Peter S. Hammerman; Gordon J. Freeman; Catherine J. Wu; Patrick A. Ott; Kwok-Kin Wong; Eliezer M. Van Allen

SUMMARY Response to immune checkpoint blockade in mesenchymal tumors is poorly characterized, but immunogenomic dissection of these cancers could inform immunotherapy mediators. We identified a treatment‐naive patient who has metastatic uterine leiomyosarcoma and has experienced complete tumor remission for >2 years on anti‐PD‐1 (pembrolizumab) monotherapy. We analyzed the primary tumor, the sole treatment‐resistant metastasis, and germline tissue to explore mechanisms of immunotherapy sensitivity and resistance. Both tumors stained diffusely for PD‐L2 and showed sparse PD‐L1 staining. PD‐1+ cell infiltration significantly decreased in the resistant tumor (p = 0.039). Genomically, the treatment‐resistant tumor uniquely harbored biallelic PTEN loss and had reduced expression of two neoantigens that demonstrated strong immunoreactivity with patient T cells in vitro, suggesting long‐lasting immunological memory. In this near‐complete response to PD‐1 blockade in a mesenchymal tumor, we identified PTEN mutations and reduced expression of genes encoding neoantigens as potential mediators of resistance to immune checkpoint therapy. Graphical Abstract Figure. No Caption available. HighlightsAnti‐PD‐1 monotherapy can induce complete tumor remission in uterine leiomyosarcomaLongitudinal profiling of patient tumors reveals response and resistance mechanismsTumor neoantigens are patient specific and can induce long‐term immunoreactivityBiallelic PTEN loss is associated with resistance to PD‐1 blockade in sarcoma &NA; George et al. report an exceptional responder to anti‐PD‐1 monotherapy in uterine leiomyosarcoma and propose mediators of treatment sensitivity and resistance. Neoantigen‐directed immunoreactivity was associated with sensitivity to anti‐PD‐1 monotherapy, whereas resistance was associated with reduction in neoantigen expression consistent with immune evasion, and biallelic PTEN loss was associated with induction of an immunosuppressive microenvironment.


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.


Nature | 2016

Genomic evolution and chemoresistance in germ-cell tumours

Amaro Taylor-Weiner; Travis I. Zack; Elizabeth O’Donnell; Jennifer L. Guerriero; Brandon David Bernard; Anita Reddy; G. Celine Han; Saud H. Aldubayan; Ali Amin-Mansour; Steven E. Schumacher; Kevin Litchfield; Clare Turnbull; Stacey Gabriel; Rameen Beroukhim; Gad Getz; Scott L. Carter; Michelle S. Hirsch; Anthony Letai; Christopher Sweeney; Eliezer M. Van Allen

Germ-cell tumours (GCTs) are derived from germ cells and occur most frequently in the testes. GCTs are histologically heterogeneous and distinctly curable with chemotherapy. Gains of chromosome arm 12p and aneuploidy are nearly universal in GCTs, but specific somatic genomic features driving tumour initiation, chemosensitivity and progression are incompletely characterized. Here, using clinical whole-exome and transcriptome sequencing of precursor, primary (testicular and mediastinal) and chemoresistant metastatic human GCTs, we show that the primary somatic feature of GCTs is highly recurrent chromosome arm level amplifications and reciprocal deletions (reciprocal loss of heterozygosity), variations that are significantly enriched in GCTs compared to 19 other cancer types. These tumours also acquire KRAS mutations during the development from precursor to primary disease, and primary testicular GCTs (TGCTs) are uniformly wild type for TP53. In addition, by functional measurement of apoptotic signalling (BH3 profiling) of fresh tumour and adjacent tissue, we find that primary TGCTs have high mitochondrial priming that facilitates chemotherapy-induced apoptosis. Finally, by phylogenetic analysis of serial TGCTs that emerge with chemotherapy resistance, we show how TGCTs gain additional reciprocal loss of heterozygosity and that this is associated with loss of pluripotency markers (NANOG and POU5F1) in chemoresistant teratomas or transformed carcinomas. Our results demonstrate the distinct genomic features underlying the origins of this disease and associated with the chemosensitivity phenotype, as well as the rare progression to chemoresistance. These results identify the convergence of cancer genomics, mitochondrial priming and GCT evolution, and may provide insights into chemosensitivity and resistance in other cancers.


Genetics in Medicine | 2017

Assigning clinical meaning to somatic and germ-line whole-exome sequencing data in a prospective cancer precision medicine study

Arezou A. Ghazani; Nelly Oliver; Joseph P. St. Pierre; Andrea Garofalo; Irene Rainville; Elaine Hiller; Daniel J. Treacy; Vanesa Rojas-Rudilla; Sam Wood; Elizabeth Bair; Michael Parello; Franklin W. Huang; Marios Giannakis; Frederick H. Wilson; Elizabeth H. Stover; Steven M. Corsello; Tom Nguyen; Huma Q. Rana; Alanna Church; Carol Lowenstein; Carrie Cibulskis; Ali Amin-Mansour; Jennifer C. Heng; Lauren K. Brais; Abigail Santos; Patrick Bauer; Amanda Waldron; Peter C. Lo; Megan J. Gorman; Christine A. Lydon

Purpose:Implementing cancer precision medicine in the clinic requires assessing the therapeutic relevance of genomic alterations. A main challenge is the systematic interpretation of whole-exome sequencing (WES) data for clinical care.Methods:One hundred sixty-five adults with metastatic colorectal and lung adenocarcinomas were prospectively enrolled in the CanSeq study. WES was performed on DNA extracted from formalin-fixed paraffin-embedded tumor biopsy samples and matched blood samples. Somatic and germ-line alterations were ranked according to therapeutic or clinical relevance. Results were interpreted using an integrated somatic and germ-line framework and returned in accordance with patient preferences.Results:At the time of this analysis, WES had been performed and results returned to the clinical team for 165 participants. Of 768 curated somatic alterations, only 31% were associated with clinical evidence and 69% with preclinical or inferential evidence. Of 806 curated germ-line variants, 5% were clinically relevant and 56% were classified as variants of unknown significance. The variant review and decision-making processes were effective when the process was changed from that of a Molecular Tumor Board to a protocol-based approach.Conclusion:The development of novel interpretive and decision-support tools that draw from scientific and clinical evidence will be crucial for the success of cancer precision medicine in WES studies.Genet Med advance online publication 26 January 2017


Cancer Discovery | 2017

Exome Sequencing of African-American Prostate Cancer Reveals Loss-of-Function ERF Mutations

Franklin W. Huang; Juan Miguel Mosquera; Andrea Garofalo; Coyin Oh; Maria Baco; Ali Amin-Mansour; Bokang Rabasha; Samira Bahl; Stephanie A. Mullane; Brian D. Robinson; Saud H. Aldubayan; Francesca Khani; Beerinder S. Karir; Eejung Kim; Jeremy Chimene-Weiss; Matan Hofree; Alessandro Romanel; Joseph R. Osborne; Jong Wook Kim; Gissou Azabdaftari; Anna Woloszynska-Read; Karen S. Sfanos; Angelo M. De Marzo; Francesca Demichelis; Stacey Gabriel; Eliezer M. Van Allen; Jill P. Mesirov; Pablo Tamayo; Mark A. Rubin; Isaac J Powell

African-American men have the highest incidence of and mortality from prostate cancer. Whether a biological basis exists for this disparity remains unclear. Exome sequencing (n = 102) and targeted validation (n = 90) of localized primary hormone-naïve prostate cancer in African-American men identified several gene mutations not previously observed in this context, including recurrent loss-of-function mutations in ERF, an ETS transcriptional repressor, in 5% of cases. Analysis of existing prostate cancer cohorts revealed ERF deletions in 3% of primary prostate cancers and mutations or deletions in ERF in 3% to 5% of lethal castration-resistant prostate cancers. Knockdown of ERF confers increased anchorage-independent growth and generates a gene expression signature associated with oncogenic ETS activation and androgen signaling. Together, these results suggest that ERF is a prostate cancer tumor-suppressor gene. More generally, our findings support the application of systematic cancer genomic characterization in settings of broader ancestral diversity to enhance discovery and, eventually, therapeutic applications.Significance: Systematic genomic sequencing of prostate cancer in African-American men revealed new insights into prostate cancer, including the identification of ERF as a prostate cancer gene; somatic copy-number alteration differences; and uncommon PIK3CA and PTEN alterations. This study highlights the importance of inclusion of underrepresented minorities in cancer sequencing studies. Cancer Discov; 7(9); 973-83. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 920.


JAMA Oncology | 2016

Clinical, Molecular, and Immune Analysis of Dabrafenib-Trametinib Combination Treatment for BRAF Inhibitor-Refractory Metastatic Melanoma: A Phase 2 Clinical Trial.

Guo Chen; Jennifer L. McQuade; David J. Panka; Courtney W. Hudgens; Ali Amin-Mansour; Xinmeng Jasmine Mu; Samira Bahl; Judit Jané-Valbuena; Khalida Wani; Alexandre Reuben; Caitlyn A. Creasy; Hong Jiang; Zachary A. Cooper; Jason Roszik; Roland L. Bassett; Aron Joon; Lauren Simpson; Rosalind Mouton; Isabella C. Glitza; Sapna Pradyuman Patel; Wen-Jen Hwu; Rodabe N. Amaria; Adi Diab; Patrick Hwu; Alexander J. Lazar; Jennifer A. Wargo; Levi A. Garraway; Michael T. Tetzlaff; Ryan J. Sullivan; Kevin B. Kim

IMPORTANCE Combined treatment with dabrafenib and trametinib (CombiDT) achieves clinical responses in only about 15% of patients with BRAF inhibitor (BRAFi)-refractory metastatic melanoma in contrast to the higher response rate observed in BRAFi-naïve patients. Identifying correlates of response and mechanisms of resistance in this population will facilitate clinical management and rational therapeutic development. OBJECTIVE To determine correlates of benefit from CombiDT therapy in patients with BRAFi-refractory metastatic melanoma. DESIGN, SETTING, AND PARTICIPANTS Single-center, single-arm, open-label phase 2 trial of CombiDT treatment in patients with BRAF V600 metastatic melanoma resistant to BRAFi monotherapy conducted between September 2012 and October 2014 at the University of Texas MD Anderson Cancer Center. Key eligibility criteria for participants included BRAF V600 metastatic melanoma, prior BRAFi monotherapy, measurable disease (RECIST 1.1), and tumor accessible for biopsy. INTERVENTIONS Patients were treated with dabrafenib (150 mg, twice daily) and trametinib (2 mg/d) continuously until disease progression or intolerance. All participants underwent a mandatory baseline biopsy, and optional biopsy specimens were obtained on treatment and at disease progression. Whole-exome sequencing, reverse transcription polymerase chain reaction analysis for BRAF splicing, RNA sequencing, and immunohistochemical analysis were performed on tumor samples, and blood was analyzed for levels of circulating BRAF V600. MAIN OUTCOMES AND MEASURES The primary end point was overall response rate (ORR). Progression-free survival (PFS) and overall survival (OS) were secondary clinical end points. RESULTS A total of 28 patients were screened, and 23 enrolled. Among evaluable patients, the confirmed ORR was 10%; disease control rate (DCR) was 45%, and median PFS was 13 weeks. Clinical benefit was associated with duration of prior BRAFi therapy greater than 6 months (DCR, 73% vs 11% for ≤6 months; P = .02) and decrease in circulating BRAF V600 at day 8 of cycle 1 (DCR, 75% vs 18% for no decrease; P = .02) but not with pretreatment mitogen-activated protein kinase (MAPK) pathway mutations or activation. Biopsy specimens obtained during treatment demonstrated that CombiDT therapy failed to achieve significant MAPK pathway inhibition or immune infiltration in most patients. CONCLUSIONS AND RELEVANCE The baseline presence of MAPK pathway alterations was not associated with benefit from CombiDT in patients with BRAFi-refractory metastatic melanoma. Failure to inhibit the MAPK pathway provides a likely explanation for the limited clinical benefit of CombiDT in this setting. Circulating BRAF V600 is a promising early biomarker of clinical response. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT01619774.

Collaboration


Dive into the Ali Amin-Mansour'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge