Network


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

Hotspot


Dive into the research topics where Manish R. Patel is active.

Publication


Featured researches published by Manish R. Patel.


Blood | 2017

Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia

Eytan M. Stein; Courtney D. DiNardo; Daniel A. Pollyea; Amir T. Fathi; Gail J. Roboz; Jessica K. Altman; Richard Stone; Daniel J. DeAngelo; Ross L. Levine; Ian W. Flinn; Hagop M. Kantarjian; Robert H. Collins; Manish R. Patel; Arthur E. Frankel; Anthony S. Stein; Mikkael A. Sekeres; Ronan Swords; Bruno C. Medeiros; Christophe Willekens; Paresh Vyas; Alessandra Tosolini; Qiang Xu; Robert Knight; Katharine E. Yen; Sam Agresta; Stéphane de Botton; Martin S. Tallman

Recurrent mutations in isocitrate dehydrogenase 2 (IDH2) occur in ∼12% of patients with acute myeloid leukemia (AML). Mutated IDH2 proteins neomorphically synthesize 2-hydroxyglutarate resulting in DNA and histone hypermethylation, which leads to blocked cellular differentiation. Enasidenib (AG-221/CC-90007) is a first-in-class, oral, selective inhibitor of mutant-IDH2 enzymes. This first-in-human phase 1/2 study assessed the maximum tolerated dose (MTD), pharmacokinetic and pharmacodynamic profiles, safety, and clinical activity of enasidenib in patients with mutant-IDH2 advanced myeloid malignancies. We assessed safety outcomes for all patients and clinical efficacy in the largest patient subgroup, those with relapsed or refractory AML, from the phase 1 dose-escalation and expansion phases of the study. In the dose-escalation phase, an MTD was not reached at doses ranging from 50 to 650 mg per day. Enasidenib 100 mg once daily was selected for the expansion phase on the basis of pharmacokinetic and pharmacodynamic profiles and demonstrated efficacy. Grade 3 to 4 enasidenib-related adverse events included indirect hyperbilirubinemia (12%) and IDH-inhibitor-associated differentiation syndrome (7%). Among patients with relapsed or refractory AML, overall response rate was 40.3%, with a median response duration of 5.8 months. Responses were associated with cellular differentiation and maturation, typically without evidence of aplasia. Median overall survival among relapsed/refractory patients was 9.3 months, and for the 34 patients (19.3%) who attained complete remission, overall survival was 19.7 months. Continuous daily enasidenib treatment was generally well tolerated and induced hematologic responses in patients for whom prior AML therapy had failed. Inducing differentiation of myeloblasts, not cytotoxicity, seems to drive the clinical efficacy of enasidenib. This trial was registered at www.clinicaltrials.gov as #NCT01915498.


Translational Research | 2013

Oncolytic virus therapy for cancer: the first wave of translational clinical trials

Manish R. Patel; Robert A. Kratzke

The field of oncolytic virus therapy, the use of live, replicating viruses for the treatment of cancer, has expanded rapidly over the past decade. Preclinical models have clearly demonstrated anticancer activity against a number of different cancer types. Several agents have entered clinical trials and promising results have led to late stage clinical development for some viruses. The early clinical trials have demonstrated that oncolytic viruses by themselves have potential to result in tumor regression. Engineering of viruses to express novel genes have also led to the use of these vectors as a novel form of gene therapy. As a result, interest in oncolytic virus therapy has gained traction. The following review will focus on the first wave of clinical translation of oncolytic virus therapy, what has been learned so far, and potential challenges ahead for advancing the field.


Translational Research | 2013

Gene Therapy for Human Disease: Clinical Advances and ChallengesReview ArticleOncolytic virus therapy for cancer: the first wave of translational clinical trials

Manish R. Patel; Robert A. Kratzke

The field of oncolytic virus therapy, the use of live, replicating viruses for the treatment of cancer, has expanded rapidly over the past decade. Preclinical models have clearly demonstrated anticancer activity against a number of different cancer types. Several agents have entered clinical trials and promising results have led to late stage clinical development for some viruses. The early clinical trials have demonstrated that oncolytic viruses by themselves have potential to result in tumor regression. Engineering of viruses to express novel genes have also led to the use of these vectors as a novel form of gene therapy. As a result, interest in oncolytic virus therapy has gained traction. The following review will focus on the first wave of clinical translation of oncolytic virus therapy, what has been learned so far, and potential challenges ahead for advancing the field.


Oncotarget | 2015

Vesicular stomatitis virus expressing interferon-β is oncolytic and promotes antitumor immune responses in a syngeneic murine model of non-small cell lung cancer

Manish R. Patel; Blake A. Jacobson; Yan Ji; Jeremy Drees; Shaogeng Tang; Kerry Xiong; Hengbing Wang; Jennifer E. Prigge; Alexander S. Dash; Andrea K. Kratzke; Emily Mesev; Ryan G. Etchison; Mark J. Federspiel; Stephen J. Russell; Robert A. Kratzke

Vesicular stomatitis virus (VSV) is a potent oncolytic virus for many tumors. VSV that produces interferon-β (VSV-IFNβ) is now in early clinical testing for solid tumors. Here, the preclinical activity of VSV and VSV-IFNβ against non-small cell lung cancer (NSCLC) is reported. NSCLC cell lines were treated in vitro with VSV expressing green fluorescence protein (VSV-GFP) and VSV-IFNβ. VSV-GFP and VSV-IFNβ were active against NSCLC cells. JAK/STAT inhibition with ruxolitinib re-sensitized resistant H838 cells to VSV-IFNβ mediated oncolysis. Intratumoral injections of VSV-GFP and VSV-IFNβ reduced tumor growth and weight in H2009 nude mouse xenografts (p < 0.01). A similar trend was observed in A549 xenografts. Syngeneic LM2 lung tumors grown in flanks of A/J mice were injected with VSV-IFNβ intratumorally. Treatment of LM2 tumors with VSV-IFNβ resulted in tumor regression, prolonged survival (p < 0.0001), and cure of 30% of mice. Intratumoral injection of VSV-IFNβ resulted in decreased tumor-infiltrating regulatory T cells (Treg) and increased CD8+ T cells. Tumor cell expression of PDL-1 was increased after VSV-IFNβ treatment. VSV-IFNβ has potent antitumor effects and promotes systemic antitumor immunity. These data support further clinical investigation of VSV-IFNβ for NSCLC.


Journal of Thoracic Oncology | 2007

Ras pathway activation in malignant mesothelioma

Manish R. Patel; Blake A. Jacobson; Arpita De; Sandra P. Frizelle; Pasi A. Jänne; Saritha C. Thumma; Brian A. Whitson; Faris Farassati; Robert A. Kratzke

Introduction: Mutations in Ras family genes are rare in malignant mesothelioma. The role of activation of the Ras signaling pathway in the pathogenesis of mesothelioma is not clear. Methods: We studied the activation status of the Ras pathway and the status of other Ras-associated kinases in a panel of human mesothelioma cell lines. In addition, we tested the effect of inhibition of several kinase pathways on mesothelioma cell proliferation. The potential role of kinase signaling on the regulation of cap-dependent translation was also studied. Results: In general, Ras-guanosine triphosphate (GTP) was higher in mesothelioma cell lines when compared with a nontransformed mesothelial cell line (LP9). Furthermore, known Ras effectors such as extracellular-regulated kinase 1/2, p38 mitogen-activated protein kinase, and c-Jun N-terminal kinase were found to be active in most of the mesothelioma cell lines tested. Exposure to specific inhibitors of extracellular-regulated kinase 1/2 (U0126) and c-Jun N-terminal kinase (SP600125) significantly decreased the proliferation of H2596 and H2373 cells compared with mock-treated cells. SP600125-mediated c-Jun N-terminal kinase inhibition, but not extracellular-regulated kinase 1/2 inhibition, resulted in a decrease in phosphorylation of 4E-BP1, consequently decreasing cap-dependent activation. Conclusions: These experiments provide a rationale for targeting Ras and associated signaling pathways in mesothelioma and also suggest cap-dependent translation as one mechanism by which Ras induces proliferation in this disease.


British Journal of Cancer | 2009

Activated 4E-BP1 represses tumourigenesis and IGF-I-mediated activation of the eIF4F complex in mesothelioma

Blake A. Jacobson; Arpita De; Marian G. Kratzke; Manish R. Patel; Joseph Jay-Dixon; Bryan A. Whitson; Ahad A. Sadiq; Peter B. Bitterman; Vitaly A. Polunovsky; Robert A. Kratzke

Background:Insulin-like growth factor (IGF)-I signalling stimulates proliferation, survival, and invasion in malignant mesothelioma and other tumour types. Studies have found that tumourigenesis is linked to dysregulation of cap-dependent protein translation.Methods:The effect of IGF stimulation on cap-mediated translation activation in mesothelioma cell lines was studied using binding assays to a synthetic 7-methyl GTP-cap analogue. In addition, cap-mediated translation was genetically repressed in these cells with a dominant active motive of 4E-BP1.Results:In most mesothelioma cell lines, IGF-I stimulation resulted in a hyperphosphorylation-mediated inactivation of 4E-BP1 compared with that in normal mesothelial cells. An inhibitor of Akt diminished IGF-I-mediated phosphorylation of 4E-BP1, whereas inhibiting MAPK signalling had no such effect. IGF-I stimulation resulted in the activation of the cap-mediated translation complex as indicated by an increased eIF4G/eIF4E ratio in cap-affinity assays. Akt inhibition reversed the eIF4G/eIF4E ratio. Mesothelioma cells transfected with an activated 4E-BP1 protein (4E-BP1A37/A46) were resistant to IGF-I-mediated growth, motility, and colony formation. In a murine xenograft model, mesothelioma cells expressing the dominant active 4E-BP1A37/A46 repressor protein showed abrogated tumourigenicity compared with control tumours.Conclusion:IGF-I signalling in mesothelioma cells drives cell proliferation, motility, and tumourigenesis through its ability to activate cap-mediated protein translation complex through PI3K/Akt/mTOR signalling.


PLOS ONE | 2013

Targeting eukaryotic translation in mesothelioma cells with an eIF4E-specific antisense oligonucleotide.

Blake A. Jacobson; Saritha C. Thumma; Joseph Jay-Dixon; Manish R. Patel; K. Dubear Kroening; Marian G. Kratzke; Ryan G. Etchison; Bruce W. Konicek; Jeremy R. Graff; Robert A. Kratzke

Background Aberrant cap-dependent translation is implicated in tumorigenesis in multiple tumor types including mesothelioma. In this study, disabling the eIF4F complex by targeting eIF4E with eIF4E-specific antisense oligonucleotide (4EASO) is assessed as a therapy for mesothelioma. Methods Mesothelioma cells were transfected with 4EASO, designed to target eIF4E mRNA, or mismatch-ASO control. Cell survival was measured in mesothelioma treated with 4EASO alone or combined with either gemcitabine or pemetrexed. Levels of eIF4E, ODC, Bcl-2 and β-actin were assessed following treatment. Binding to a synthetic cap-analogue was used to study the strength of eIF4F complex activation following treatment. Results eIF4E level and the formation of eIF4F cap-complex decreased in response to 4EASO, but not mismatch control ASO, resulting in cleavage of PARP indicating apoptosis. 4EASO treatment resulted in dose dependent decrease in eIF4E levels, which corresponded to cytotoxicity of mesothelioma cells. 4EASO resulted in decreased levels of eIF4E in non-malignant LP9 cells, but this did not correspond to increased cytotoxicity. Proteins thought to be regulated by cap-dependent translation, Bcl-2 and ODC, were decreased upon treatment with 4EASO. Combination therapy of 4EASO with pemetrexed or gemcitabine further reduced cell number. Conclusion 4EASO is a novel drug that causes apoptosis and selectively reduces eIF4E levels, eIF4F complex formation, and proliferation of mesothelioma cells. eIF4E knockdown results in decreased expression of anti-apoptotic and pro-growth proteins and enhances chemosensitivity.


European Journal of Cancer | 2015

2746 A Phase 2 open-label, multicenter study of single-agent rucaparib in the treatment of patients with relapsed ovarian cancer and a deleterious BRCA mutation

Ronnie Shapira-Frommer; Amit M. Oza; Susan M. Domchek; Judith Balmaña; Manish R. Patel; Lee-may Chen; Yvette Drew; Howard A. Burris; Jacob Korach; Michael J. Flynn; Valerie Bowering; Mark Aloysuis Morgan; Simon Watkins; Dayna Simpson; Sandra Goble; Lara Maloney; Rebecca Kristeleit

5513 Background: Rucaparib is a potent oral PARP inhibitor that has shown robust activity in patients (pts) with homologous recombination deficient (HRD) tumors, with the highest response rates observed in ovarian and breast cancer pts harboring deleterious germline BRCA mutations (gBRCAmut). This phase II study (NCT 01891344) evaluated the efficacy and safety of rucaparib in pts with relapsed gBRCAmut high-grade epithelial ovarian, fallopian tube, or primary peritoneal cancer (EOC/FTC/PPC). Methods: Women with gBRCAmut EOC/FTC/PPC who received 2-4 prior chemotherapy (chemo) regimens and had a progression-free interval (PFI) ≥ 6 months after their last platinum dose were enrolled. ECOG PS 0-1 and adequate organ function were required. Pts received oral rucaparib 600 mg BID in 21 day cycles until disease progression. The primary endpoint was objective response rate (ORR) by RECIST 1.1. Results: 35 pts have been enrolled. Median age was 55 years (range 44-84). The median number of prior chemo regimens was 2...


Journal of Macromolecular Science, Part A | 1986

Synthesis and Study of Polymeric Ultraviolet Absorbers. II

Mahendra R. Patel; J. S. Parmar; Manish R. Patel; Mohan M. Patel

Abstract Polymeric UV absorbers have been prepared by free-radical solution copolymerization at 75°C of methyl methacrylate and 2-hydroxy-4-methacryloyloxybenzophenone monomers at low conversion (around 10%). The composition of the copolymers was determined by UV, IR, and NMR studies. The molecular weight was estimated by GPC. The reactivity ratios were determined by several methods. Viscosity was used to study the effect of copolymer composition and solvents. The copolymers were also analyzed by TGA and DSC, and DSC was used to study the effect of copolymer composition on Tg.


Lancet Oncology | 2018

ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial

John Wrangle; Vamsidhar Velcheti; Manish R. Patel; Elizabeth Garrett-Mayer; Elizabeth G. Hill; James G. Ravenel; Jeffrey S. Miller; Mohammad Farhad; Kate Anderton; Kathryn G. Lindsey; Michele Taffaro-Neskey; Carol A. Sherman; Samantha Suriano; Marzena Swiderska-Syn; Amy Sion; Joni Harris; Andie R Edwards; Julie A. Rytlewski; Catherine Sanders; Erik Yusko; Mark D. Robinson; Carsten Krieg; William L. Redmond; Jack O. Egan; Peter R. Rhode; Emily K. Jeng; Amy Rock; Hing C. Wong; Mark P. Rubinstein

BACKGROUND Immunotherapy with PD-1 or PD-L1 blockade fails to induce a response in about 80% of patients with unselected non-small cell lung cancer (NSCLC), and many of those who do initially respond then develop resistance to treatment. Agonists that target the shared interleukin-2 (IL-2) and IL-15Rβγ pathway have induced complete and durable responses in some cancers, but no studies have been done to assess the safety or efficacy of these agonists in combination with anti-PD-1 immunotherapy. We aimed to define the safety, tolerability, and activity of this drug combination in patients with NSCLC. METHODS In this non-randomised, open-label, phase 1b trial, we enrolled patients (aged ≥18 years) with previously treated histologically or cytologically confirmed stage IIIB or IV NSCLC from three academic hospitals in the USA. Key eligibility criteria included measurable disease, eligibility to receive anti-PD-1 immunotherapy, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients received the anti-PD-1 monoclonal antibody nivolumab intravenously at 3 mg/kg (then 240 mg when US Food and Drug Administration [FDA]-approved dosing changed) every 14 days (either as new treatment or continued treatment at the time of disease progression) and the IL-15 superagonist ALT-803 subcutaneously once per week on weeks 1-5 of four 6-week cycles for 6 months. ALT-803 was administered at one of four escalating dose concentrations: 6, 10, 15, or 20 μg/kg. The primary endpoint was to define safety and tolerability and to establish a recommended phase 2 dose of ALT-803 in combination with nivolumab. Analyses were per-protocol and included any patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, number NCT02523469; phase 2 enrolment of patients is ongoing. FINDINGS Between Jan 18, 2016, and June 28, 2017, 23 patients were enrolled and 21 were treated at four dose levels of ALT-803 in combination with nivolumab. Two patients did not receive treatment because of the development of inter-current illness during enrolment, one patient due to leucopenia and one patient due to pulmonary dysfunction. No dose-limiting toxicities were recorded and the maximum tolerated dose was not reached. The most common adverse events were injection-site reactions (in 19 [90%] of 21 patients) and flu-like symptoms (15 [71%]). The most common grade 3 adverse events, occurring in two patients each, were lymphocytopenia and fatigue. A grade 3 myocardial infarction occurred in one patient. No grade 4 or 5 adverse events were recorded. The recommended phase 2 dose of ALT-803 is 20 μg/kg given once per week subcutaneously in combination with 240 mg intravenous nivolumab every 2 weeks. INTERPRETATION ALT-803 in combination with nivolumab can be safely administered in an outpatient setting. The promising clinical activity observed with the addition of ALT-803 to the regimen of patients with PD-1 monoclonal antibody relapsed and refractory disease shows evidence of anti-tumour activity for a new class of agents in NSCLC. FUNDING Altor BioScience (a NantWorks company), National Institutes of Health, and Medical University of South Carolina Hollings Cancer Center.

Collaboration


Dive into the Manish R. Patel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey R. Infante

Sarah Cannon Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shubham Pant

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Howard A. Burris

Sarah Cannon Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ian W. Flinn

Sarah Cannon Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Todd Michael Bauer

Sarah Cannon Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ahmad Raza

University of Minnesota

View shared research outputs
Researchain Logo
Decentralizing Knowledge