Network


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

Hotspot


Dive into the research topics where Bijay S. Jaiswal is active.

Publication


Featured researches published by Bijay S. Jaiswal.


Nature | 2010

RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth

Georgia Hatzivassiliou; Kyung Song; Ivana Yen; Barbara J. Brandhuber; Daniel J. Anderson; Ryan Alvarado; Mary J. C. Ludlam; David Stokoe; Susan L. Gloor; Guy Vigers; Tony Morales; Ignacio Aliagas; Bonnie Liu; Steve Sideris; Klaus P. Hoeflich; Bijay S. Jaiswal; Somasekar Seshagiri; Hartmut Koeppen; Marcia Belvin; Lori S. Friedman; Shiva Malek

Activating mutations in KRAS and BRAF are found in more than 30% of all human tumours and 40% of melanoma, respectively, thus targeting this pathway could have broad therapeutic effects. Small molecule ATP-competitive RAF kinase inhibitors have potent antitumour effects on mutant BRAF(V600E) tumours but, in contrast to mitogen-activated protein kinase kinase (MEK) inhibitors, are not potent against RAS mutant tumour models, despite RAF functioning as a key effector downstream of RAS and upstream of MEK. Here we show that ATP-competitive RAF inhibitors have two opposing mechanisms of action depending on the cellular context. In BRAF(V600E) tumours, RAF inhibitors effectively block the mitogen-activated protein kinase (MAPK) signalling pathway and decrease tumour growth. Notably, in KRAS mutant and RAS/RAF wild-type tumours, RAF inhibitors activate the RAF–MEK–ERK pathway in a RAS-dependent manner, thus enhancing tumour growth in some xenograft models. Inhibitor binding activates wild-type RAF isoforms by inducing dimerization, membrane localization and interaction with RAS–GTP. These events occur independently of kinase inhibition and are, instead, linked to direct conformational effects of inhibitors on the RAF kinase domain. On the basis of these findings, we demonstrate that ATP-competitive kinase inhibitors can have opposing functions as inhibitors or activators of signalling pathways, depending on the cellular context. Furthermore, this work provides new insights into the therapeutic use of ATP-competitive RAF inhibitors.


Nature | 2010

Diverse somatic mutation patterns and pathway alterations in human cancers.

Zhengyan Kan; Bijay S. Jaiswal; Jeremy Stinson; Vasantharajan Janakiraman; Deepali Bhatt; Howard M. Stern; Peng Yue; Peter M. Haverty; Richard Bourgon; Jianbiao Zheng; Martin Moorhead; Subhra Chaudhuri; Lynn P. Tomsho; Brock A. Peters; Kanan Pujara; Shaun Cordes; David P. Davis; Victoria Carlton; Wenlin Yuan; Li Li; Weiru Wang; Charles Eigenbrot; Joshua S. Kaminker; David A. Eberhard; Paul Waring; Stephan C. Schuster; Zora Modrusan; Zemin Zhang; David Stokoe; Frederic J. de Sauvage

The systematic characterization of somatic mutations in cancer genomes is essential for understanding the disease and for developing targeted therapeutics. Here we report the identification of 2,576 somatic mutations across approximately 1,800 megabases of DNA representing 1,507 coding genes from 441 tumours comprising breast, lung, ovarian and prostate cancer types and subtypes. We found that mutation rates and the sets of mutated genes varied substantially across tumour types and subtypes. Statistical analysis identified 77 significantly mutated genes including protein kinases, G-protein-coupled receptors such as GRM8, BAI3, AGTRL1 (also called APLNR) and LPHN3, and other druggable targets. Integrated analysis of somatic mutations and copy number alterations identified another 35 significantly altered genes including GNAS, indicating an expanded role for galpha subunits in multiple cancer types. Furthermore, our experimental analyses demonstrate the functional roles of mutant GNAO1 (a Galpha subunit) and mutant MAP2K4 (a member of the JNK signalling pathway) in oncogenesis. Our study provides an overview of the mutational spectra across major human cancers and identifies several potential therapeutic targets.


Nature | 2012

Recurrent R-spondin fusions in colon cancer

Somasekar Seshagiri; Eric Stawiski; Steffen Durinck; Zora Modrusan; Elaine E. Storm; Caitlin B. Conboy; Subhra Chaudhuri; Yinghui Guan; Vasantharajan Janakiraman; Bijay S. Jaiswal; Joseph Guillory; Connie Ha; Gerrit J. P. Dijkgraaf; Jeremy Stinson; Florian Gnad; Melanie A. Huntley; Jeremiah D. Degenhardt; Peter M. Haverty; Richard Bourgon; Weiru Wang; Hartmut Koeppen; Robert Gentleman; Timothy K. Starr; Zemin Zhang; David A. Largaespada; Thomas D. Wu; Frederic J. de Sauvage

Identifying and understanding changes in cancer genomes is essential for the development of targeted therapeutics. Here we analyse systematically more than 70 pairs of primary human colon tumours by applying next-generation sequencing to characterize their exomes, transcriptomes and copy-number alterations. We have identified 36,303 protein-altering somatic changes that include several new recurrent mutations in the Wnt pathway gene TCF7L2, chromatin-remodelling genes such as TET2 and TET3 and receptor tyrosine kinases including ERBB3. Our analysis for significantly mutated cancer genes identified 23 candidates, including the cell cycle checkpoint kinase ATM. Copy-number and RNA-seq data analysis identified amplifications and corresponding overexpression of IGF2 in a subset of colon tumours. Furthermore, using RNA-seq data we identified multiple fusion transcripts including recurrent gene fusions involving R-spondin family members RSPO2 and RSPO3 that together occur in 10% of colon tumours. The RSPO fusions were mutually exclusive with APC mutations, indicating that they probably have a role in the activation of Wnt signalling and tumorigenesis. Consistent with this we show that the RSPO fusion proteins were capable of potentiating Wnt signalling. The R-spondin gene fusions and several other gene mutations identified in this study provide new potential opportunities for therapeutic intervention in colon cancer.


Nature Genetics | 2012

Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer

Charles M. Rudin; Steffen Durinck; Eric Stawiski; John T. Poirier; Zora Modrusan; David S. Shames; Emily Bergbower; Yinghui Guan; James Shin; Joseph Guillory; Celina Sanchez Rivers; Catherine K. Foo; Deepali Bhatt; Jeremy Stinson; Florian Gnad; Peter M. Haverty; Robert Gentleman; Subhra Chaudhuri; Vasantharajan Janakiraman; Bijay S. Jaiswal; Chaitali Parikh; Wenlin Yuan; Zemin Zhang; Hartmut Koeppen; Thomas D. Wu; Howard M. Stern; Robert L. Yauch; Kenneth Huffman; Diego D Paskulin; Peter B. Illei

Small-cell lung cancer (SCLC) is an exceptionally aggressive disease with poor prognosis. Here, we obtained exome, transcriptome and copy-number alteration data from approximately 53 samples consisting of 36 primary human SCLC and normal tissue pairs and 17 matched SCLC and lymphoblastoid cell lines. We also obtained data for 4 primary tumors and 23 SCLC cell lines. We identified 22 significantly mutated genes in SCLC, including genes encoding kinases, G protein–coupled receptors and chromatin-modifying proteins. We found that several members of the SOX family of genes were mutated in SCLC. We also found SOX2 amplification in ∼27% of the samples. Suppression of SOX2 using shRNAs blocked proliferation of SOX2-amplified SCLC lines. RNA sequencing identified multiple fusion transcripts and a recurrent RLF-MYCL1 fusion. Silencing of MYCL1 in SCLC cell lines that had the RLF-MYCL1 fusion decreased cell proliferation. These data provide an in-depth view of the spectrum of genomic alterations in SCLC and identify several potential targets for therapeutic intervention.


Cancer Cell | 2009

Somatic mutations in p85α promote tumorigenesis through class IA PI3K activation

Bijay S. Jaiswal; Vasantharajan Janakiraman; Noelyn M. Kljavin; Subhra Chaudhuri; Howard M. Stern; Weiru Wang; Zhengyan Kan; Hashem A. Dbouk; Brock A. Peters; Paul Waring; Trisha Dela Vega; Denise M. Kenski; Krista K. Bowman; Maria N. Lorenzo; Hong Li; Jiansheng Wu; Zora Modrusan; Jeremy Stinson; Michael Eby; Peng Yue; Josh Kaminker; Frederic J. de Sauvage; Jonathan M. Backer; Somasekar Seshagiri

Members of the mammalian phosphoinositide-3-OH kinase (PI3K) family of proteins are critical regulators of various cellular process including cell survival, growth, proliferation, and motility. Oncogenic activating mutations in the p110alpha catalytic subunit of the heterodimeric p110/p85 PI3K enzyme are frequent in human cancers. Here we show the presence of frequent mutations in p85alpha in colon cancer, a majority of which occurs in the inter-Src homology-2 (iSH2) domain. These mutations uncouple and retain p85alphas p110-stabilizing activity, while abrogating its p110-inhibitory activity. The p85alpha mutants promote cell survival, AKT activation, anchorage-independent cell growth, and oncogenesis in a p110-dependent manner.


Cancer Cell | 2013

Oncogenic ERBB3 Mutations in Human Cancers

Bijay S. Jaiswal; Noelyn M. Kljavin; Eric Stawiski; Emily Chan; Chaitali Parikh; Steffen Durinck; Subhra Chaudhuri; Kanan Pujara; Joseph Guillory; Kyle A. Edgar; Vasantharajan Janakiraman; Rolf-Peter Scholz; Krista K. Bowman; Maria N. Lorenzo; Hong Li; Jiansheng Wu; Wenlin Yuan; Brock A. Peters; Zhengyan Kan; Jeremy Stinson; Michelle Mak; Zora Modrusan; Charles Eigenbrot; Ron Firestein; Howard M. Stern; Krishnaraj Rajalingam; Gabriele Schaefer; Mark Merchant; Mark X. Sliwkowski; Frederic J. de Sauvage

The human epidermal growth factor receptor (HER) family of tyrosine kinases is deregulated in multiple cancers either through amplification, overexpression, or mutation. ERBB3/HER3, the only member with an impaired kinase domain, although amplified or overexpressed in some cancers, has not been reported to carry oncogenic mutations. Here, we report the identification of ERBB3 somatic mutations in ~11% of colon and gastric cancers. We found that the ERBB3 mutants transformed colonic and breast epithelial cells in a ligand-independent manner. However, the mutant ERBB3 oncogenic activity was dependent on kinase-active ERBB2. Furthermore, we found that anti-ERBB antibodies and small molecule inhibitors effectively blocked mutant ERBB3-mediated oncogenic signaling and disease progression in vivo.


Nature Genetics | 2016

Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations

Raphael Bueno; Eric Stawiski; Leonard D. Goldstein; Steffen Durinck; Assunta De Rienzo; Zora Modrusan; Florian Gnad; Thong T. Nguyen; Bijay S. Jaiswal; Lucian R. Chirieac; Daniele Sciaranghella; Nhien Dao; Corinne E. Gustafson; Kiara J. Munir; Jason A. Hackney; Amitabha Chaudhuri; Ravi Gupta; Joseph Guillory; Karen Toy; Connie Ha; Ying-Jiun Chen; Jeremy Stinson; Subhra Chaudhuri; Na Zhang; Thomas D. Wu; David J. Sugarbaker; Frederic J. de Sauvage; William G. Richards; Somasekar Seshagiri

We analyzed transcriptomes (n = 211), whole exomes (n = 99) and targeted exomes (n = 103) from 216 malignant pleural mesothelioma (MPM) tumors. Using RNA-seq data, we identified four distinct molecular subtypes: sarcomatoid, epithelioid, biphasic-epithelioid (biphasic-E) and biphasic-sarcomatoid (biphasic-S). Through exome analysis, we found BAP1, NF2, TP53, SETD2, DDX3X, ULK2, RYR2, CFAP45, SETDB1 and DDX51 to be significantly mutated (q-score ≥ 0.8) in MPMs. We identified recurrent mutations in several genes, including SF3B1 (∼2%; 4/216) and TRAF7 (∼2%; 5/216). SF3B1-mutant samples showed a splicing profile distinct from that of wild-type tumors. TRAF7 alterations occurred primarily in the WD40 domain and were, except in one case, mutually exclusive with NF2 alterations. We found recurrent gene fusions and splice alterations to be frequent mechanisms for inactivation of NF2, BAP1 and SETD2. Through integrated analyses, we identified alterations in Hippo, mTOR, histone methylation, RNA helicase and p53 signaling pathways in MPMs.


Nature Genetics | 2015

Spectrum of diverse genomic alterations define non–clear cell renal carcinoma subtypes

Steffen Durinck; Eric Stawiski; Andrea Pavia-Jimenez; Zora Modrusan; Payal Kapur; Bijay S. Jaiswal; Na Zhang; Vanina Toffessi-Tcheuyap; Thong T. Nguyen; Kanika Bajaj Pahuja; Ying Jiun Chen; Sadia Saleem; Subhra Chaudhuri; Sherry Heldens; Marlena Jackson; Samuel Peña-Llopis; Joseph Guillory; Karen Toy; Connie Ha; Corissa J. Harris; Eboni Holloman; Haley Hill; Jeremy Stinson; Celina Sanchez Rivers; Vasantharajan Janakiraman; Weiru Wang; Lisa N. Kinch; Nick V. Grishin; Peter M. Haverty; Bernard Chow

To further understand the molecular distinctions between kidney cancer subtypes, we analyzed exome, transcriptome and copy number alteration data from 167 primary human tumors that included renal oncocytomas and non–clear cell renal cell carcinomas (nccRCCs), consisting of papillary (pRCC), chromophobe (chRCC) and translocation (tRCC) subtypes. We identified ten significantly mutated genes in pRCC, including MET, NF2, SLC5A3, PNKD and CPQ. MET mutations occurred in 15% (10/65) of pRCC samples and included previously unreported recurrent activating mutations. In chRCC, we found TP53, PTEN, FAAH2, PDHB, PDXDC1 and ZNF765 to be significantly mutated. Gene expression analysis identified a five-gene set that enabled the molecular classification of chRCC, renal oncocytoma and pRCC. Using RNA sequencing, we identified previously unreported gene fusions, including ACTG1-MITF fusion. Ectopic expression of the ACTG1-MITF fusion led to cellular transformation and induced the expression of downstream target genes. Finally, we observed upregulation of the anti-apoptotic factor BIRC7 in MiTF-high RCC tumors, suggesting a potential therapeutic role for BIRC7 inhibitors.


PLOS ONE | 2009

Combined targeting of BRAF and CRAF or BRAF and PI3K effector pathways is required for efficacy in NRAS mutant tumors.

Bijay S. Jaiswal; Vasantharajan Janakiraman; Noelyn M. Kljavin; Jeffrey Eastham-Anderson; James E. Cupp; Yuxin Liang; David P. Davis; Klaus P. Hoeflich; Somasekar Seshagiri

Background Oncogenic RAS is a highly validated cancer target. Attempts at targeting RAS directly have so far not succeeded in the clinic. Understanding downstream RAS-effectors that mediate oncogenesis in a RAS mutant setting will help tailor treatments that use RAS-effector inhibitors either alone or in combination to target RAS-driven tumors. Methodology/Principal Findings In this study, we have investigated the sufficiency of targeting RAS-effectors, RAF, MEK and PI3-Kinase either alone or in combination in RAS mutant lines, using an inducible shRNA in vivo mouse model system. We find that in colon cancer cells harboring a KRASG13D mutant allele, knocking down KRAS alone or the RAFs in combination or the RAF effectors, MEK1 and MEK2, together is effective in delaying tumor growth in vivo. In melanoma cells harboring an NRASQ61L or NRASQ61K mutant allele, we find that targeting NRAS alone or both BRAF and CRAF in combination or both BRAF and PIK3CA together showed efficacy. Conclusion/Significance Our data indicates that targeting oncogenic NRAS-driven melanomas require decrease in both pERK and pAKT downstream of RAS-effectors for efficacy. This can be achieved by either targeting both BRAF and CRAF or BRAF and PIK3CA simultaneously in NRAS mutant tumor cells.


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

Regulation of Class IA PI 3-kinases: C2 domain-iSH2 domain contacts inhibit p85/p110α and are disrupted in oncogenic p85 mutants

Haiyan Wu; S. Chandra Shekar; Rory J. Flinn; Mirvat El-Sibai; Bijay S. Jaiswal; K. Ilker Sen; Vasantharajan Janakiraman; Somasekar Seshagiri; Gary J. Gerfen; Mark E. Girvin; Jonathan M. Backer

We previously proposed a model of Class IA PI3K regulation in which p85 inhibition of p110α requires (i) an inhibitory contact between the p85 nSH2 domain and the p110α helical domain, and (ii) a contact between the p85 nSH2 and iSH2 domains that orients the nSH2 so as to inhibit p110α. We proposed that oncogenic truncations of p85 fail to inhibit p110 due to a loss of the iSH2-nSH2 contact. However, we now find that within the context of a minimal regulatory fragment of p85 (the nSH2-iSH2 fragment, termed p85ni), the nSH2 domain rotates much more freely (τc ≈12.7 ns) than it could if it were interacting rigidly with the iSH2 domain. These data are not compatible with our previous model. We therefore tested an alternative model in which oncogenic p85 truncations destabilize an interface between the p110α C2 domain (residue N345) and the p85 iSH2 domain (residues D560 and N564). p85ni-D560K/N564K shows reduced inhibition of p110α, similar to the truncated p85ni-572STOP. Conversely, wild-type p85ni poorly inhibits p110αN345K. Strikingly, the p110αN345K mutant is inhibited to the same extent by the wild-type or truncated p85ni, suggesting that mutation of p110α-N345 is not additive with the p85ni-572STOP mutation. Similarly, the D560K/N564K mutation is not additive with the p85ni-572STOP mutant for downstream signaling or cellular transformation. Thus, our data suggests that mutations at the C2-iSH2 domain contact and truncations of the iSH2 domain, which are found in human tumors, both act by disrupting the C2-iSH2 domain interface.

Collaboration


Dive into the Bijay S. Jaiswal's collaboration.

Researchain Logo
Decentralizing Knowledge