Jatin Roper
Tufts Medical Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jatin Roper.
Science | 2015
Jihye Yun; Edouard Mullarky; Changyuan Lu; Kaitlyn N. Bosch; Adam Kavalier; Keith Rivera; Jatin Roper; Iok In Christine Chio; Eugenia G. Giannopoulou; Carlo Rago; Ashlesha Muley; John M. Asara; Ji Hye Paik; Olivier Elemento; Zhengming Chen; Darryl Pappin; Lukas E. Dow; Nickolas Papadopoulos; Steven S. Gross; Lewis C. Cantley
Getting all stressed out by vitamin C Few experimental cancer therapies have incited as much debate as vitamin C. Yet the mechanistic effect of vitamin C on cancer cells is still poorly understood. Yun et al. studied human colorectal cancer cells with KRAS or BRAF mutations and found that they “handle” vitamin C in a different way than other cells, ultimately to their detriment (see the Perspective by Reczek and Chandel). Because a certain receptor is up-regulated in the mutant cells, they take up the oxidized form of vitamin C (dehydroascorbate). This leads to oxidative stress, inactivation of a glycolytic enzyme required by the mutant cells for growth, and finally cell death. Whether the selective toxicity of vitamin C to these mutant cells can be exploited therapeutically remains unclear. Science, this issue p. 1391; see also p. 1317 Cancer cells with certain mutations take up the oxidized form of vitamin C, which fatally disrupts their metabolism. [Also see Perspective by Reczek and Chandel] More than half of human colorectal cancers (CRCs) carry either KRAS or BRAF mutations and are often refractory to approved targeted therapies. We found that cultured human CRC cells harboring KRAS or BRAF mutations are selectively killed when exposed to high levels of vitamin C. This effect is due to increased uptake of the oxidized form of vitamin C, dehydroascorbate (DHA), via the GLUT1 glucose transporter. Increased DHA uptake causes oxidative stress as intracellular DHA is reduced to vitamin C, depleting glutathione. Thus, reactive oxygen species accumulate and inactivate glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Inhibition of GAPDH in highly glycolytic KRAS or BRAF mutant cells leads to an energetic crisis and cell death not seen in KRAS and BRAF wild-type cells. High-dose vitamin C impairs tumor growth in Apc/KrasG12D mutant mice. These results provide a mechanistic rationale for exploring the therapeutic use of vitamin C for CRCs with KRAS or BRAF mutations.
Nature | 2016
Semir Beyaz; Miyeko D. Mana; Jatin Roper; Dmitriy Kedrin; Assieh Saadatpour; Sue-Jean Hong; Khristian E. Bauer-Rowe; Michael E. Xifaras; Adam Akkad; Erika Arias; Luca Pinello; Yarden Katz; Shweta Shinagare; Monther Abu-Remaileh; Maria M. Mihaylova; Dudley W. Lamming; Rizkullah Dogum; Guoji Guo; George W. Bell; Martin K. Selig; G. Petur Nielsen; Nitin Gupta; Cristina R. Ferrone; Vikram Deshpande; Guo-Cheng Yuan; Stuart H. Orkin; David M. Sabatini; Ömer H. Yilmaz
Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5+ intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.
PLOS ONE | 2011
Jatin Roper; Michael P. Richardson; Wei Vivian Wang; Larissa Georgeon Richard; Wei-Wei Chen; Erin M. Coffee; Mark J. Sinnamon; Lydia Lee; Peng Chieh Chen; Roderick T. Bronson; Eric Martin; Kenneth E. Hung
Purpose To examine the in vitro and in vivo efficacy of the dual PI3K/mTOR inhibitor NVP-BEZ235 in treatment of PIK3CA wild-type colorectal cancer (CRC). Experimental Design PIK3CA mutant and wild-type human CRC cell lines were treated in vitro with NVP-BEZ235, and the resulting effects on proliferation, apoptosis, and signaling were assessed. Colonic tumors from a genetically engineered mouse (GEM) model for sporadic wild-type PIK3CA CRC were treated in vivo with NVP-BEZ235. The resulting effects on macroscopic tumor growth/regression, proliferation, apoptosis, angiogenesis, and signaling were examined. Results In vitro treatment of CRC cell lines with NVP-BEZ235 resulted in transient PI3K blockade, sustained decreases in mTORC1/mTORC2 signaling, and a corresponding decrease in cell viability (median IC50 = 9.0–14.3 nM). Similar effects were seen in paired isogenic CRC cell lines that differed only in the presence or absence of an activating PIK3CA mutant allele. In vivo treatment of colonic tumor-bearing mice with NVP-BEZ235 resulted in transient PI3K inhibition and sustained blockade of mTORC1/mTORC2 signaling. Longitudinal tumor surveillance by optical colonoscopy demonstrated a 97% increase in tumor size in control mice (p = 0.01) vs. a 43% decrease (p = 0.008) in treated mice. Ex vivo analysis of the NVP-BEZ235-treated tumors demonstrated a 56% decrease in proliferation (p = 0.003), no effects on apoptosis, and a 75% reduction in angiogenesis (p = 0.013). Conclusions These studies provide the preclinical rationale for studies examining the efficacy of the dual PI3K/mTOR inhibitor NVP-BEZ235 in treatment of PIK3CA wild-type CRC.
Cancer Discovery | 2014
Anthony C. Faber; Erin M. Coffee; Carlotta Costa; Anahita Dastur; Hiromichi Ebi; Aaron N. Hata; Alan T. Yeo; Elena J. Edelman; Youngchul Song; Ah Ting Tam; Jessica L. Boisvert; Randy J. Milano; Jatin Roper; David P. Kodack; Rakesh K. Jain; Ryan B. Corcoran; Miguel Rivera; Sridhar Ramaswamy; Kenneth E. Hung; Cyril H. Benes; Jeffrey A. Engelman
Colorectal cancers harboring KRAS or BRAF mutations are refractory to current targeted therapies. Using data from a high-throughput drug screen, we have developed a novel therapeutic strategy that targets the apoptotic machinery using the BCL-2 family inhibitor ABT-263 (navitoclax) in combination with a TORC1/2 inhibitor, AZD8055. This combination leads to efficient apoptosis specifically in KRAS- and BRAF-mutant but not wild-type (WT) colorectal cancer cells. This specific susceptibility results from TORC1/2 inhibition leading to suppression of MCL-1 expression in mutant, but not WT, colorectal cancers, leading to abrogation of BIM/MCL-1 complexes. This combination strategy leads to tumor regressions in both KRAS-mutant colorectal cancer xenograft and genetically engineered mouse models of colorectal cancer, but not in the corresponding KRAS-WT colorectal cancer models. These data suggest that the combination of BCL-2/BCL-XL inhibitors with TORC1/2 inhibitors constitutes a promising targeted therapy strategy to treat these recalcitrant cancers.
Clinical Cancer Research | 2013
Erin M. Coffee; Anthony C. Faber; Jatin Roper; Mark J. Sinnamon; Gautam Goel; Lily Keung; Wei Vivian Wang; Loredana Vecchione; Veerle De Vriendt; Barbara Weinstein; Roderick T. Bronson; Sabine Tejpar; Ramnik J. Xavier; Jeffrey A. Engelman; Eric Martin; Kenneth E. Hung
Purpose: BRAFV600E mutations are associated with poor clinical prognosis in colorectal cancer (CRC). Although selective BRAF inhibitors are effective for treatment of melanoma, comparable efforts in CRC have been disappointing. Here, we investigated potential mechanisms underlying this resistance to BRAF inhibitors in BRAFV600E CRC. Experimental Design: We examined phosphoinositide 3-kinase (PI3K)/mTOR signaling in BRAFV600E CRC cell lines after BRAF inhibition and cell viability and apoptosis after combined BRAF and PI3K/mTOR inhibition. We assessed the efficacy of in vivo combination treatment using a novel genetically engineered mouse model (GEMM) for BRAFV600E CRC. Results: Western blot analysis revealed sustained PI3K/mTOR signaling upon BRAF inhibition. Our BRAFV600E GEMM presented with sessile serrated adenomas/polyps, as seen in humans. Combination treatment in vivo resulted in induction of apoptosis and tumor regression. Conclusions: We have established a novel GEMM to interrogate BRAFV600E CRC biology and identify more efficacious treatment strategies. Combination BRAF and PI3K/mTOR inhibitor treatment should be explored in clinical trials. Clin Cancer Res; 19(10); 2688–98. ©2013 AACR.
Nature Biotechnology | 2017
Jatin Roper; Tuomas Tammela; Naniye Malli Cetinbas; Adam Akkad; Ali Roghanian; Steffen Rickelt; Mohammad Almeqdadi; Katherine Wu; Matthias A. Oberli; Francisco J. Sánchez-Rivera; Yoona Park; Xu Liang; George Eng; Martin S. Taylor; Roxana Azimi; Dmitriy Kedrin; Rachit Neupane; Semir Beyaz; Ewa Sicinska; Yvelisse Suarez; James Yoo; Lillian Chen; Lawrence R. Zukerberg; Pekka Katajisto; Vikram Deshpande; Adam J. Bass; Philip N. Tsichlis; Jacqueline A. Lees; Robert Langer; Richard O. Hynes
In vivo interrogation of the function of genes implicated in tumorigenesis is limited by the need to generate and cross germline mutant mice. Here we describe approaches to model colorectal cancer (CRC) and metastasis, which rely on in situ gene editing and orthotopic organoid transplantation in mice without cancer-predisposing mutations. Autochthonous tumor formation is induced by CRISPR-Cas9-based editing of the Apc and Trp53 tumor suppressor genes in colon epithelial cells and by orthotopic transplantation of Apc-edited colon organoids. ApcΔ/Δ;KrasG12D/+;Trp53Δ/Δ (AKP) mouse colon organoids and human CRC organoids engraft in the distal colon and metastasize to the liver. Finally, we apply the orthotopic transplantation model to characterize the clonal dynamics of Lgr5+ stem cells and demonstrate sequential activation of an oncogene in established colon adenomas. These experimental systems enable rapid in vivo characterization of cancer-associated genes and reproduce the entire spectrum of tumor progression and metastasis.
Trends in Pharmacological Sciences | 2012
Jatin Roper; Kenneth E. Hung
To establish effective drug development for colorectal cancer (CRC), preclinical models that are robust surrogates for human disease are crucial. Mouse models are an attractive platform because of their relatively low cost, short life span, and ease of use. There are two main categories of mouse CRC models: xenografts derived from implantation of CRC cells or tumors in immunodeficient mice; and genetically engineered mouse models (GEMMs) derived from modification of human cancer predisposition genes, resulting in spontaneous tumor formation. Here, we review xenografts and GEMMs and focus on their potential application in translational research. Furthermore, we describe newer GEMMs for sporadic CRC that are particularly suitable for drug testing. Finally, we discuss recent advances in small-animal imaging, such as optical colonoscopy, which allow in vivo assessment of tumors. With the increasing sophistication of GEMMs, our preclinical armamentarium provides new hope for the ongoing war against CRC.
Cancer Letters | 2014
Jatin Roper; Mark J. Sinnamon; Erin M. Coffee; Peter J. Belmont; Lily Keung; Larissa Georgeon-Richard; Wei Vivian Wang; Anthony C. Faber; Jihye Yun; Ömer H. Yilmaz; Roderick T. Bronson; Eric Martin; Philip N. Tsichlis; Kenneth E. Hung
PI3K inhibition in combination with other agents has not been studied in the context of PIK3CA wild-type, KRAS mutant cancer. In a screen of phospho-kinases, PI3K inhibition of KRAS mutant colorectal cancer cells activated the MAPK pathway. Combination PI3K/MEK inhibition with NVP-BKM120 and PD-0325901 induced tumor regression in a mouse model of PIK3CA wild-type, KRAS mutant colorectal cancer, which was mediated by inhibition of mTORC1, inhibition of MCL-1, and activation of BIM. These findings implicate mitochondrial-dependent apoptotic mechanisms as determinants for the efficacy of PI3K/MEK inhibition in the treatment of PIK3CA wild-type, KRAS mutant cancer.
Science Signaling | 2014
Peter J. Belmont; Ping Jiang; Trevor D. McKee; Tao Xie; J. Isaacson; N. E. Baryla; Jatin Roper; Mark J. Sinnamon; N. V. Lee; Julie L.C. Kan; O. Guicherit; Bradly G. Wouters; Catherine O'Brien; D. Shields; P. Olson; Todd VanArsdale; Scott Weinrich; Paul A. Rejto; J. G. Christensen; V. R. Fantin; Kenneth E. Hung; Eric Martin
Combination therapy with EGFR inhibitors may overcome acquired resistance to PI3K pathway inhibitors in some colorectal cancer patients. Drug Resistance Reveals Vulnerability Kinase inhibitors are a common therapy for many cancers, but tumors frequently acquire resistance. Understanding the mechanisms of resistance can reveal new therapeutic options. Using various human and mouse models of KRAS-mutant colorectal cancer (CRC), Belmont et al. found that signaling by the receptor tyrosine kinase EGFR and the related ERBB family members was increased in CRC cells that had acquired resistance to a dual inhibitor of the kinases PI3K and mTOR. Increased EGFR expression was induced by the drug through the release of PI3K pathway–mediated inhibition of the transcription factor FOXO3a. Withdrawing the drug from culture medium returned EGFR abundance and signaling to pretreatment levels. Inhibiting EGFR restored sensitivity to the PI3K/mTOR inhibitor in drug-resistant cells in culture and induced tumor regression in drug-resistant allografts in mice. Thus, CRC patients that develop resistance to PI3K/mTOR inhibitors may benefit from additional treatment with EGFR inhibitors. Targeted blockade of aberrantly activated signaling pathways is an attractive therapeutic strategy for solid tumors, but drug resistance is common. KRAS is a frequently mutated gene in human cancer but remains a challenging clinical target. Inhibitors against KRAS signaling mediators, namely, PI3K (phosphatidylinositol 3-kinase) and mTOR (mechanistic target of rapamycin), have limited clinical efficacy as single agents in KRAS-mutant colorectal cancer (CRC). We investigated potential bypass mechanisms to PI3K/mTOR inhibition in KRAS-mutant CRC. Using genetically engineered mouse model cells that had acquired resistance to the dual PI3K/mTOR small-molecule inhibitor PF-04691502, we determined with chemical library screens that inhibitors of the ERBB [epidermal growth factor receptor (EGFR)] family restored the sensitivity to PF-04691502. Although EGFR inhibitors alone have limited efficacy in reducing KRAS-mutant tumors, we found that PF-04691502 induced the abundance, phosphorylation, and activity of EGFR, ERBB2, and ERBB3 through activation of FOXO3a (forkhead box O 3a), a transcription factor inhibited by the PI3K to AKT pathway. PF-04691502 also induced a stem cell–like gene expression signature. KRAS-mutant patient-derived xenografts from mice treated with PF-04691502 had a similar gene expression signature and exhibited increased EGFR activation, suggesting that this drug-induced resistance mechanism may occur in patients. Combination therapy with dacomitinib (a pan-ERBB inhibitor) restored sensitivity to PF-04691502 in drug-resistant cells in culture and induced tumor regression in drug-resistant allografts in mice. Our findings suggest that combining PI3K/mTOR and EGFR inhibitors may improve therapeutic outcome in patients with KRAS-mutant CRC.
Nature Protocols | 2018
Jatin Roper; Tuomas Tammela; Adam Akkad; Mohammad Almeqdadi; Sebastian B Santos; Tyler Jacks; Ömer H. Yilmaz
Most genetically engineered mouse models (GEMMs) of colorectal cancer are limited by tumor formation in the small intestine, a high tumor burden that limits metastasis, and the need to generate and cross mutant mice. Cell line or organoid transplantation models generally produce tumors in ectopic locations—such as the subcutaneous space, kidney capsule, or cecal wall—that do not reflect the native stromal environment of the colon mucosa. Here, we describe detailed protocols to rapidly and efficiently induce site-directed tumors in the distal colon of mice that are based on colonoscopy-guided mucosal injection. These techniques can be adapted to deliver viral vectors carrying Cre recombinase, CRISPR–Cas9 components, CRISPR-engineered mouse tumor organoids, or human cancer organoids to mice to model the adenoma–carcinoma–metastasis sequence of tumor progression. The colonoscopy injection procedure takes ∼15 min, including preparation. In our experience, anyone with reasonable hand–eye coordination can become proficient with mouse colonoscopy and mucosal injection with a few hours of practice. These approaches are ideal for a wide range of applications, including assessment of gene function in tumorigenesis, examination of tumor–stroma interactions, studies of cancer metastasis, and translational research with patient-derived cancers.