Network


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

Hotspot


Dive into the research topics where Amit Dutt is active.

Publication


Featured researches published by Amit Dutt.


Nature | 2008

Somatic mutations affect key pathways in lung adenocarcinoma

Li Ding; Gad Getz; David A. Wheeler; Elaine R. Mardis; Michael D. McLellan; Kristian Cibulskis; Carrie Sougnez; Heidi Greulich; Donna M. Muzny; Margaret Morgan; Lucinda Fulton; Robert S. Fulton; Qunyuan Zhang; Michael C. Wendl; Michael S. Lawrence; David E. Larson; Ken Chen; David J. Dooling; Aniko Sabo; Alicia Hawes; Hua Shen; Shalini N. Jhangiani; Lora Lewis; Otis Hall; Yiming Zhu; Tittu Mathew; Yanru Ren; Jiqiang Yao; Steven E. Scherer; Kerstin Clerc

Determining the genetic basis of cancer requires comprehensive analyses of large collections of histopathologically well-classified primary tumours. Here we report the results of a collaborative study to discover somatic mutations in 188 human lung adenocarcinomas. DNA sequencing of 623 genes with known or potential relationships to cancer revealed more than 1,000 somatic mutations across the samples. Our analysis identified 26 genes that are mutated at significantly high frequencies and thus are probably involved in carcinogenesis. The frequently mutated genes include tyrosine kinases, among them the EGFR homologue ERBB4; multiple ephrin receptor genes, notably EPHA3; vascular endothelial growth factor receptor KDR; and NTRK genes. These data provide evidence of somatic mutations in primary lung adenocarcinoma for several tumour suppressor genes involved in other cancers—including NF1, APC, RB1 and ATM—and for sequence changes in PTPRD as well as the frequently deleted gene LRP1B. The observed mutational profiles correlate with clinical features, smoking status and DNA repair defects. These results are reinforced by data integration including single nucleotide polymorphism array and gene expression array. Our findings shed further light on several important signalling pathways involved in lung adenocarcinoma, and suggest new molecular targets for treatment.


Nature Genetics | 2009

SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas

Adam J. Bass; Hideo Watanabe; Craig H. Mermel; Yu Ss; Sven Perner; Roeland Verhaak; So Young Kim; Leslie Wardwell; Pablo Tamayo; Irit Gat-Viks; Alex H. Ramos; Michele S. Woo; Barbara A. Weir; Gad Getz; Rameen Beroukhim; Michael O'Kelly; Amit Dutt; Orit Rozenblatt-Rosen; Piotr Dziunycz; Justin Komisarof; Lucian R. Chirieac; Christopher J. Lafargue; Veit Scheble; Theresia Wilbertz; Changqing Ma; Shilpa Rao; Hiroshi Nakagawa; Douglas B. Stairs; Lin Lin; Thomas J. Giordano

Lineage-survival oncogenes are activated by somatic DNA alterations in cancers arising from the cell lineages in which these genes play a role in normal development. Here we show that a peak of genomic amplification on chromosome 3q26.33 found in squamous cell carcinomas (SCCs) of the lung and esophagus contains the transcription factor gene SOX2, which is mutated in hereditary human esophageal malformations, is necessary for normal esophageal squamous development, promotes differentiation and proliferation of basal tracheal cells and cooperates in induction of pluripotent stem cells. SOX2 expression is required for proliferation and anchorage-independent growth of lung and esophageal cell lines, as shown by RNA interference experiments. Furthermore, ectopic expression of SOX2 here cooperated with FOXE1 or FGFR2 to transform immortalized tracheobronchial epithelial cells. SOX2-driven tumors show expression of markers of both squamous differentiation and pluripotency. These characteristics identify SOX2 as a lineage-survival oncogene in lung and esophageal SCC.


Cancer Discovery | 2011

Mutations in the DDR2 Kinase Gene Identify a Novel Therapeutic Target in Squamous Cell Lung Cancer

Peter S. Hammerman; Martin L. Sos; Alex H. Ramos; Chunxiao Xu; Amit Dutt; Wenjun Zhou; Lear E. Brace; Brittany A. Woods; Wenchu Lin; Jianming Zhang; Xianming Deng; Sang Min Lim; Stefanie Heynck; Martin Peifer; Jeffrey R. Simard; Michael S. Lawrence; Robert C. Onofrio; Helga B. Salvesen; Danila Seidel; Thomas Zander; Johannes M. Heuckmann; Alex Soltermann; Holger Moch; Mirjam Koker; Frauke Leenders; Franziska Gabler; Silvia Querings; Sascha Ansén; Elisabeth Brambilla; Christian Brambilla

UNLABELLED While genomically targeted therapies have improved outcomes for patients with lung adenocarcinoma, little is known about the genomic alterations which drive squamous cell lung cancer. Sanger sequencing of the tyrosine kinome identified mutations in the DDR2 kinase gene in 3.8% of squamous cell lung cancers and cell lines. Squamous lung cancer cell lines harboring DDR2 mutations were selectively killed by knock-down of DDR2 by RNAi or by treatment with the multi-targeted kinase inhibitor dasatinib. Tumors established from a DDR2 mutant cell line were sensitive to dasatinib in xenograft models. Expression of mutated DDR2 led to cellular transformation which was blocked by dasatinib. A squamous cell lung cancer patient with a response to dasatinib and erlotinib treatment harbored a DDR2 kinase domain mutation. These data suggest that gain-of-function mutations in DDR2 are important oncogenic events and are amenable to therapy with dasatinib. As dasatinib is already approved for use, these findings could be rapidly translated into clinical trials. SIGNIFICANCE DDR2 mutations are present in 4% of lung SCCs, and DDR2 mutations are associated with sensitivity to dasatinib. These findings provide a rationale for designing clinical trials with the FDA-approved drug dasatinib in patients with lung SCCs.


PLOS ONE | 2011

Inhibitor-Sensitive FGFR1 Amplification in Human Non-Small Cell Lung Cancer

Amit Dutt; Alex H. Ramos; Peter S. Hammerman; Craig H. Mermel; Jeonghee Cho; Tanaz Sharifnia; Ajit G. Chande; Kumiko Tanaka; Nicolas Stransky; Heidi Greulich; Nathanael S. Gray; Matthew Meyerson

Background Squamous cell lung carcinomas account for approximately 25% of new lung carcinoma cases and 40,000 deaths per year in the United States. Although there are multiple genomically targeted therapies for lung adenocarcinoma, none has yet been reported in squamous cell lung carcinoma. Methodology/Principal Findings Using SNP array analysis, we found that a region of chromosome segment 8p11-12 containing three genes–WHSC1L1, LETM2, and FGFR1–is amplified in 3% of lung adenocarcinomas and 21% of squamous cell lung carcinomas. Furthermore, we demonstrated that a non-small cell lung carcinoma cell line harboring focal amplification of FGFR1 is dependent on FGFR1 activity for cell growth, as treatment of this cell line either with FGFR1-specific shRNAs or with FGFR small molecule enzymatic inhibitors leads to cell growth inhibition. Conclusions/Significance These studies show that FGFR1 amplification is common in squamous cell lung cancer, and that FGFR1 may represent a promising therapeutic target in non-small cell lung cancer.


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

Drug-sensitive FGFR2 mutations in endometrial carcinoma

Amit Dutt; Helga B. Salvesen; Tzu-Hsiu Chen; Alex H. Ramos; Robert C. Onofrio; Charlie Hatton; Richard Nicoletti; Wendy Winckler; Rupinder Grewal; Megan Hanna; Nicolas Wyhs; Liuda Ziaugra; Daniel J. Richter; Jone Trovik; Ingeborg B. Engelsen; Ingunn Stefansson; Timothy Fennell; Kristian Cibulskis; Michael C. Zody; Lars A. Akslen; Stacey Gabriel; Kwok-Kin Wong; William R. Sellers; Matthew Meyerson; Heidi Greulich

Oncogenic activation of tyrosine kinases is a common mechanism of carcinogenesis and, given the druggable nature of these enzymes, an attractive target for anticancer therapy. Here, we show that somatic mutations of the fibroblast growth factor receptor 2 (FGFR2) tyrosine kinase gene, FGFR2, are present in 12% of endometrial carcinomas, with additional instances found in lung squamous cell carcinoma and cervical carcinoma. These FGFR2 mutations, many of which are identical to mutations associated with congenital craniofacial developmental disorders, are constitutively activated and oncogenic when ectopically expressed in NIH 3T3 cells. Inhibition of FGFR2 kinase activity in endometrial carcinoma cell lines bearing such FGFR2 mutations inhibits transformation and survival, implicating FGFR2 as a novel therapeutic target in endometrial carcinoma.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Integrated genomic profiling of endometrial carcinoma associates aggressive tumors with indicators of PI3 kinase activation

H. B. Salvesen; Scott L. Carter; Monica Mannelqvist; Amit Dutt; Gad Getz; Ingunn Stefansson; Maria B. Ræder; Martin L. Sos; Ingeborg B. Engelsen; Jone Trovik; Elisabeth Wik; Heidi Greulich; Trond Hellem Bø; Inge Jonassen; Roman K. Thomas; Thomas Zander; Levy A. Garraway; Anne Margrete Øyan; William R. Sellers; Karl-Henning Kalland; Matthew Meyerson; Lars A. Akslen; Rameen Beroukhim

Although 75% of endometrial cancers are treated at an early stage, 15% to 20% of these recur. We performed an integrated analysis of genome-wide expression and copy-number data for primary endometrial carcinomas with extensive clinical and histopathological data to detect features predictive of recurrent disease. Unsupervised analysis of the expression data distinguished 2 major clusters with strikingly different phenotypes, including significant differences in disease-free survival. To identify possible mechanisms for these differences, we performed a global genomic survey of amplifications, deletions, and loss of heterozygosity, which identified 11 significantly amplified and 13 significantly deleted regions. Amplifications of 3q26.32 harboring the oncogene PIK3CA were associated with poor prognosis and segregated with the aggressive transcriptional cluster. Moreover, samples with PIK3CA amplification carried signatures associated with in vitro activation of PI3 kinase (PI3K), a signature that was shared by aggressive tumors without PIK3CA amplification. Tumors with loss of PTEN expression or PIK3CA overexpression that did not have PIK3CA amplification also shared the PI3K activation signature, high protein expression of the PI3K pathway member STMN1, and an aggressive phenotype in test and validation datasets. However, mutations of PTEN or PIK3CA were not associated with the same expression profile or aggressive phenotype. STMN1 expression had independent prognostic value. The results affirm the utility of systematic characterization of the cancer genome in clinically annotated specimens and suggest the particular importance of the PI3K pathway in patients who have aggressive endometrial cancer.


Chemistry & Biology | 2010

A Structure-guided Approach to Creating Covalent FGFR Inhibitors

Wenjun Zhou; Wooyoung Hur; Ultan McDermott; Amit Dutt; Wa Xian; Scott B. Ficarro; Jianming Zhang; Sreenath V. Sharma; Joan S. Brugge; Matthew Meyerson; Jeffrey Settleman; Nathanael S. Gray

The fibroblast growth factor receptor tyrosine kinases (FGFR1, 2, 3, and 4) represent promising therapeutic targets in a number of cancers. We have developed the first potent and selective irreversible inhibitor of FGFR1, 2, 3, and 4, which we named FIIN-1 that forms a covalent bond with cysteine 486 located in the P loop of the FGFR1 ATP binding site. We demonstrated that the inhibitor potently inhibits Tel-FGFR1-transformed Ba/F3 cells (EC(50) = 14 nM) as well as numerous FGFR-dependent cancer cell lines. A biotin-derivatized version of the inhibitor, FIIN-1-biotin, was shown to covalently label FGFR1 at Cys486. FIIN-1 is a useful probe of FGFR-dependent cellular phenomena and may provide a starting point of the development of therapeutically relevant irreversible inhibitors of wild-type and drug-resistant forms of FGFR kinases.


Current Opinion in Oncology | 2007

Single nucleotide polymorphism array analysis of cancer.

Amit Dutt; Rameen Beroukhim

Purpose of review Classifying tumors and identifying therapeutic targets requires a description of the genetic changes underlying cancer. Single nucleotide polymorphism (SNP) arrays provide a high-resolution platform for describing several types of genetic changes simultaneously. With the resolution of these arrays increasing exponentially, they are becoming increasingly powerful tools for describing the genetic events underlying cancer. Recent findings The ability to map loss of heterozygosity (LOH) and overall copy number variations using SNP arrays is known. Techniques have recently been developed to map LOH at high resolution in the absence of paired normal data. Copy number variations described by SNP array studies are now reaching resolutions enabling the identification of novel oncogenes and tumor suppressor genes. The ability to determine allele-specific copy number changes has only recently been described. Moreover, SNP arrays offer a high-throughput platform for large-scale association studies that are likely to lead to the identification of multiple germline variants that predispose to cancer. Summary SNP arrays are an ideal platform for identifying both somatic and germline genetic variants that lead to cancer. They provide a basis for DNA-based cancer classification and help to define the genes being modulated, improving understanding of cancer genesis and potential therapeutic targets.


Cancer Biology & Therapy | 2009

Amplification of chromosomal segment 4q12 in non-small cell lung cancer

Alex H. Ramos; Amit Dutt; Craig H. Mermel; Sven Perner; Jeonghee Cho; Christopher J. Lafargue; Laura A. Johnson; Ann Cathrin Stiedl; Kumiko Tanaka; Adam J. Bass; Jordi Barretina; Barbara A. Weir; Rameen Beroukhim; Roman K. Thomas; John D. Minna; Lucian R. Chirieac; Neal I. Lindeman; Thomas J. Giordano; David G. Beer; Patrick L. Wagner; Ignacio I. Wistuba; Mark A. Rubin; Matthew Meyerson

In cancer, proto-oncogenes are often altered by genomic amplification. Here we report recurrent focal amplifications of chromosomal segment 4q12 overlapping the proto-oncogenes PDGFRA and KIT in non-small cell lung cancer (NSCLC). Single nucleotide polymorphism (SNP) array and fluorescent in situ hybridization (FISH) analysis indicate that 4q12 is amplified in 3-7% of lung adenocarcinomas and 8-10% of lung squamous cell carcinomas. In addition, we demonstrate that the NSCLC cell line NCI-H1703 exhibits focal amplification of PDGFRA and is dependent on PDGFRα activity for cell growth. Treatment of NCI-H1703 cells with PDGFRA-specific shRNAs or with the PDGFRα/KIT small molecule inhibitors imatinib or sunitinib leads to cell growth inhibition. However, these observations do not extend to NSCLC cell lines with lower-amplitude and broader gains of chromosome 4q. Together these observations implicate PDGFRA and KIT as potential oncogenes in NSCLC, but further study is needed to define the specific characteristics of those tumors that could respond to PDGFRα/KIT inhibitors.


PLOS Biology | 2004

EGF signal propagation during C. elegans vulval development mediated by ROM-1 rhomboid.

Amit Dutt; Stefano Canevascini; Erika Froehli-Hoier; Alex Hajnal

During Caenorhabditis elegans vulval development, the anchor cell (AC) in the somatic gonad secretes an epidermal growth factor (EGF) to activate the EGF receptor (EGFR) signaling pathway in the adjacent vulval precursor cells (VPCs). The inductive AC signal specifies the vulval fates of the three proximal VPCs P5.p, P6.p, and P7.p. The C. elegans Rhomboid homolog ROM-1 increases the range of EGF, allowing the inductive signal to reach the distal VPCs P3.p, P4.p and P8.p, which are further away from the AC. Surprisingly, ROM-1 functions in the signal-receiving VPCs rather than the signal-sending AC. This observation led to the discovery of an AC–independent activity of EGF in the VPCs that promotes vulval cell fate specification and depends on ROM-1. Of the two previously reported EGF splice variants, the longer one requires ROM-1 for its activity, while the shorter form acts independently of ROM-1. We present a model in which ROM-1 relays the inductive AC signal from the proximal to the distal VPCs by allowing the secretion of the LIN-3L splice variant. These results indicate that, in spite of their structural diversity, Rhomboid proteins play a conserved role in activating EGFR signaling in C. elegans, Drosophila, and possibly also in mammals.

Collaboration


Dive into the Amit Dutt's collaboration.

Top Co-Authors

Avatar

Pratik Chandrani

Homi Bhabha National Institute

View shared research outputs
Top Co-Authors

Avatar

Pawan Upadhyay

Homi Bhabha National Institute

View shared research outputs
Top Co-Authors

Avatar

Nilesh Gardi

Homi Bhabha National Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Prajish Iyer

Homi Bhabha National Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge