Network


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

Hotspot


Dive into the research topics where Manoj Maniar is active.

Publication


Featured researches published by Manoj Maniar.


Journal of Clinical Oncology | 2008

Phase I Study of ON 01910.Na, a Novel Modulator of the Polo-Like Kinase 1 Pathway, in Adult Patients With Solid Tumors

Antonio Jimeno; Jing Li; Wells A. Messersmith; Daniel A. Laheru; Michelle A. Rudek; Manoj Maniar; Manuel Hidalgo; Sharyn D. Baker; Ross C. Donehower

PURPOSE We conducted a first-in-man (to our knowledge) phase I study to determine the dose-limiting toxicities (DLTs), characterize the pharmacokinetic profile, and document any antitumor activity of ON 01910.Na, a new chemical entity that arrests cancer cells in G(2)/M by modulating mitotic regulatory pathways including polo-like kinase 1 (Plk1). PATIENTS AND METHODS Patients had solid tumors refractory to standard therapy. ON 01910.Na was administered as a 2-hour infusion on days 1, 4, 8, 11, 15, and 18 in 28-day cycles. The starting dose was 80 mg, and an accelerated titration schedule (single-patient cohorts) was used for escalation. Pharmacokinetics were studied on days 1 and 15 of cycle 1. RESULTS Twenty patients (11 women and nine men; age 46 to 73 years) were enrolled onto the study. Dose levels of 80, 160, 320, 480, 800, 1,280, 2,080, and 3,120 mg were evaluated in single-patient cohorts. A DLT and additional grade 2 toxicities made the 4,370-mg dose (n = 6) not tolerable, and the next lower dose cohort (3,120 mg) was expanded to six assessable patients. Toxicities were skeletal, abdominal, and tumor pain; nausea; urge to defecate; and fatigue. Hematologic toxicity was infrequent and mild. ON 01910.Na pharmacokinetics were characterized by a rapid distribution phase (distribution half-life, 1 hour) and a relatively slow elimination phase (elimination half-life, 27 hours). A refractory ovarian cancer patient had an objective response after four cycles and remained progression free for 24 months. CONCLUSION ON 01910.Na showed a distinct but moderate toxicity pattern. The recommended phase II dose of ON 01910.Na with this schedule of administration is 3,120 mg. Single-agent activity was documented in an ovarian cancer patient.


British Journal of Haematology | 2013

Phase I clinical trial of oral rigosertib in patients with myelodysplastic syndromes.

Rami S. Komrokji; Azra Raza; Jeffrey E. Lancet; Chen Ren; David R. Taft; Manoj Maniar; Francois Wilhelm; Alan F. List

The multi‐kinase inhibitor rigosertib (ON 01910.Na) induces mitotic arrest and apoptosis in myeloblasts, while sparing normal cells. The purpose of this study was to determine the pharmacokinetic profile, maximum‐tolerated dose (MTD), safety, and clinical activity of an oral formulation of rigosertib in patients with myelodysplastic syndromes (MDS). For pharmacokinetic studies, patients received rigosertib in single escalating weekly doses. To determine the MTD, patient cohorts received escalating doses of rigosertib twice daily for 14 d of a 21‐d cycle. Overall, 37 patients were treated. Rigosertib exposure increased with escalating oral doses. Mean absolute oral bioavailability ranged from 13·9% (fed) to 34·8% (fasting) in 12 patients treated at the 560 mg b.i.d. dose level. Dose‐limiting toxicity (grade 3 dysuria and shortness of breath) occurred at the 700 mg b.i.d. dose. Five patients experienced grade 3 non‐haematological toxicity, including symptoms of urothelial inflammation, hypotension and syncope, fatigue and abdominal pain. Encouraging signs of clinical activity included two bone marrow complete remissions in refractory anaemia with excess blasts type 1 patients previously treated with azacitidine. In addition, four patients each achieved transfusion independence and haematological improvements. In conclusion, oral rigosertib is bioavailable and well tolerated, and has clinical activity in patients with MDS.


Hematological Oncology | 2015

Clinical activity and safety of the dual pathway inhibitor rigosertib for higher risk myelodysplastic syndromes following DNA methyltransferase inhibitor therapy.

Lewis R. Silverman; Peter L. Greenberg; Azra Raza; Matthew J. Olnes; James F. Holland; Premkumar E Reddy; Manoj Maniar; Francois Wilhelm

Rigosertib (ON 01910.Na) is an inhibitor of the phosphoinositide 3‐kinase and polo‐like kinase pathways that induces mitotic arrest and apoptosis in neoplastic cells, while sparing normal cells. Our purpose is to summarize the clinical activity and safety of intravenous (IV) rigosertib delivered by an external ambulatory infusion pump in patients with refractory anemia with excess blasts‐1, ‐2, or, ‐t myelodysplastic syndromes (MDS) following prior treatment with DNA methyltransferase (DNMT) inhibitors. A total of 39 patients with MDS who fulfilled these criteria were enrolled in four phase 1‐2 clinical trials of IV rigosertib. Thirty five (88%) had higher risk disease according to the Revised International Prognostic Scoring System. Median overall survival for this group of 39 patients was 35 weeks. Of 30 evaluable patients with follow‐up bone marrow biopsies, 12 (40%) achieved complete (n = 5) or partial (n = 7) bone marrow blast responses. In addition, 15 patients achieved stabilization of bone marrow blasts. One patient with a complete bone marrow response also achieved a complete cytogenetic response. A second patient with stable bone marrow blasts achieved a partial cytogenetic response. Two of the responding patients and three patients with stable disease had hematological improvements. Rigosertib‐induced bone marrow blast decreases and stability appeared to be predictive of prolonged survival. IV rigosertib had a favorable safety profile without significant myelosuppression. Most common drug‐related toxicities included fatigue, diarrhea, nausea, dysuria, and hematuria. In summary, IV rigosertib is well tolerated and has clinical activity in patients with higher risk MDS following DNMT inhibitor treatment. A multinational pivotal phase 3 randomized clinical trial of rigosertib versus best supportive care for patients with MDS with excess blasts following prior treatment with DNMT inhibitors (ONTIME: ON 01910.Na Trial In Myelodysplastic SyndromE) has recently completed enrollment.


Clinical Cancer Research | 2012

Phase I Study of Rigosertib, an Inhibitor of the Phosphatidylinositol 3-Kinase and Polo-like Kinase 1 Pathways, Combined with Gemcitabine in Patients with Solid Tumors and Pancreatic Cancer

Wen Wee Ma; Wells A. Messersmith; Grace K. Dy; Colin D. Weekes; Amy Whitworth; Chen Ren; Manoj Maniar; Francois Wilhelm; S. Gail Eckhardt; Alex A. Adjei; Antonio Jimeno

Purpose: Rigosertib, a dual non-ATP inhibitor of polo-like kinase 1 (Plk1) and phosphoinositide 3-kinase pathways (PI3K), and gemcitabine have synergistic antitumor activity when combined in preclinical studies. This phase I study aimed to determine the recommended phase II dose (RPTD) of the combination of rigosertib and gemcitabine in patients with cancer. Experimental Design: Patients with solid tumors who failed standard therapy or were candidates for gemcitabine-based therapy were eligible. Gemcitabine was administered on days 1, 8, and 15 on a 28-day cycle and rigosertib on days 1, 4, 8, 11, 15, and 18. Pharmacokinetic studies were conducted during an expansion cohort of patients with advanced pancreatic ductal adenocarcinoma (PDA). Results: Forty patients were treated, 19 in the dose-escalation phase and 21 in the expansion cohort. Dose levels evaluated were (gemcitabine/rigosertib mg/m2): 750/600 (n = 4), 750/1,200 (n = 3), 1,000/600 (n = 3), 1,000/1,200 (n = 3), and 1,000/1,800 (n = 6 + 21). One dose-limiting toxicity (death) occurred at the highest dose level (1,000/1,800) tested. Non–dose-limiting ≥grade II/III toxicities included neutropenia, lymphopenia, thrombocytopenia, fatigue, and nausea. Grade III/IV neutropenia, thrombocytopenia, and fatigue were seen in two, one, and two patients in the expansion cohort. Partial responses were observed in PDA, thymic cancer, and Hodgkin lymphoma, including gemcitabine-pretreated PDA. The pharmacokinetic profile of rigosertib was not affected by gemcitabine. Conclusion: The RPTD established in this study is rigosertib 1,800 mg/m2 and gemcitabine 1,000 mg/m2. This regimen is well tolerated with a toxicity profile of the combination similar to the profile of gemcitabine alone. Antitumor efficacy was observed in patients who previously progressed on gemcitabine-based therapy. Clin Cancer Res; 18(7); 2048–55. ©2012 AACR.


Clinical Cancer Research | 2014

Phase I study of oral rigosertib (ON 01910.Na), a dual inhibitor of the PI3K and Plk1 pathways, in adult patients with advanced solid malignancies.

Daniel W. Bowles; Jennifer R. Diamond; Elaine T. Lam; Colin D. Weekes; David P. Astling; Ryan T. Anderson; Stephen Leong; Lia Gore; Marileila Varella-Garcia; Brian W. Vogler; Stephen B. Keysar; Elizabeth Freas; Dara L. Aisner; Chen Ren; Aik Choon Tan; Francois Wilhelm; Manoj Maniar; S. Gail Eckhardt; Wells A. Messersmith; Antonio Jimeno

Purpose: To determine the pharmacokinetics (PK), maximum tolerated dose (MTD), safety, and antitumor activity of an oral formulation of rigosertib, a dual phosphoinositide 3-kinase (PI3K) and polo-like kinase 1 (Plk1) pathway inhibitor, in patients with advanced solid malignancies. Experimental Design: Patients with advanced solid malignancies received rigosertib twice daily continuously in 21-day cycles. Doses were escalated until intolerable grade ≥2 toxicities, at which point the previous dose level was expanded to define the MTD. All patients were assessed for safety, PK, and response. Urinary PK were performed at the MTD. Archival tumors were assessed for potential molecular biomarkers with multiplex mutation testing. A subset of squamous cell carcinomas (SCC) underwent exome sequencing. Results: Forty-eight patients received a median of 2 cycles of therapy at 5 dose levels. Rigosertib exposure increased with escalating doses. Dose-limiting toxicities were hematuria and dysuria. The most common grade ≥2 drug-related toxicities involved urothelial irritation. The MTD is 560 mg twice daily. Activity was seen in head and neck SCCs (1 complete response, 1 partial response) and stable disease for ≥12 weeks was observed in 8 additional patients. Tumors experiencing ≥partial response had PI3K pathway activation, inactivated p53, and unique variants in ROBO3 and FAT1, two genes interacting with the Wnt/β-catenin pathway. Conclusions: The recommended phase II dose of oral rigosertib is 560 mg twice daily given continuously. Urinary toxicity is the dose-limiting and most common toxicity. Alterations in PI3K, p53, and Wnt/β-catenin pathway signaling should be investigated as potential biomarkers of response in future trials. Clin Cancer Res; 20(6); 1656–65. ©2014 AACR.


Journal of Pharmaceutical Sciences | 2013

Disposition of ON 01210.Na (Ex-RAD(R)), a novel radioprotectant, in the isolated perfused rat liver: Probing metabolic inhibition to increase systemic exposure

Mitalee Tamhane; Manoj Maniar; Chen Ren; Kenza E. Benzeroual; David R. Taft

ON 01210.Na (Ex-RAD) is a novel benzyl styryl sulfone analog, developed as a radioprotectant by Onconova Therapeutics Inc. The objectives of this research were to evaluate the hepatobiliary disposition of ON 01210.Na in the isolated perfused rat liver (IPRL) and to determine the effect of coadministration of ethacrynic acid (EA) on the pharmacokinetic profile of ON 01210.Na. EA acid was used as a prototypical inhibitor of glutathione-S-transferase inhibitor. ON 01210.Na was highly bound in IPRL perfusate proteins, and binding was significantly lower in the presence of EA. Dose-escalation studies (bolus dose, target concentrations 10-250 μg/mL) showed that ON 01210.Na followed nonlinear pharmacokinetics with hepatic clearance decreasing from 3.14 to 1.99 mL/min with increasing dose. ON 01210.Na underwent extensive metabolic degradation to its glutathione (GSH) adduct in liver. The GSH metabolite was mainly excreted into the bile. Coadministration of EA (1 mM) significantly inhibited the conversion of ON 01210.Na to its GSH conjugate, resulting in decreased clearance (approx. fivefold lower), and prolonged elimination from the perfusate. These preclinical studies suggest that EA is a potential pharmacoenhancer that can reduce the metabolism of ON 01210.Na in vivo, thereby increasing drug exposure and boosting radioprotective activity.


Cancer Research | 2017

Abstract 2172: ON 123300, an orally administered novel CDK4/6 + ARK5 inhibitor, exhibits potent antitumor activity in vivo: comparative studies with Palbociclib

Shraddha Patel; Priya Pancholi; Tanvi Visal; Amruta Samant; Dhvanir Kansara; V. J. Rajadhyaksha; Benjamin S. Hoffman; Manoj Maniar; Vikas Sehdev

Background: The overexpression of cyclin-dependent kinases 4/6 (CDK4/6) is known to cause cell cycle dysregulation in certain cancer types, making these cell cycle kinases attractive targets for pharmacological inhibition. The effectiveness of first-generation non-selective cyclin-dependent kinases, such as roscovitine and flavopiridol, was hampered by toxicities, leading to the development of second-generation compounds like IBRANCE®/Palbociclib that specifically inhibit CDK4 and 6. ON 123300 is a third-generation potent CDK4/6 inhibitor that also inhibits ARK5 with low nanomolar potency and has the potential to improve upon second-generation compounds. Previous studies have demonstrated the inhibitory effect of single-agent ON 123300 in various pre-clinical cancer models of MM and leukemia. In this study, we investigated the comparative therapeutic potential of ON 123300 as an oral anticancer agent and a second-generation inhibitor, Palbociclib, in xenografted Rb+ve mouse models. Methods: MDA-MB-435S xenografted mice were treated once a day for 21 days with ON 123300 (125mg/kg) or Palbociclib (125mg/kg). Tumor volumes were measured and peripheral blood was gathered to evaluate the effects on hematological parameters. Separately, Western blot analyses were performed to determine the effect of CDK4/6 inhibition on p-Rb following intra-tumoral treatment with ON 123300 (2.5µM) or Palbociclib (2.5µM). Results: ON 123300 and Palbociclib reduced tumor growth with an equivalent magnitude during the 21-day treatment period, suggesting that the two compounds were equally effective in this model. Both compounds decreased RBC and platelet counts, however Palbociclib had a more prominent and statistically significant (P≤0.05) inhibitory effect on neutrophil counts when compared to ON 123300 (30.70 ± 3.55 vs. 45.10 ± 2.04). Western blot analysis of tumor tissues demonstrated equivalent effects on p-Rb for both compounds. Conclusions: Xenograft data indicates that a third-generation CDK4/6 inhibitor, ON 123300, is as effective as Palbociclib in an Rb+ve xenograft model. Moreover, this study also suggests that ON 123300 may have the added advantage of reduced neutropenia compared to Palbociclib. Prior preclinical data suggest that ON 123300 may be efficacious in Rb-ve tumors, where second-generation compounds have diminished single-agent activity, and our ongoing studies are aimed at further characterizing the in vivo activity of ON 123300 in this setting. Citation Format: Shraddha Patel, Priya Pancholi, Tanvi Visal, Amruta Samant, Dhvanir Kansara, V J. Rajadhyaksha, Benjamin S. Hoffman, Manoj Maniar, Vikas Sehdev. ON 123300, an orally administered novel CDK4/6 + ARK5 inhibitor, exhibits potent antitumor activity in vivo: comparative studies with Palbociclib [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2172. doi:10.1158/1538-7445.AM2017-2172


Cancer Research | 2016

Abstract 4687: Rigosertib synergistically enhances the anticancer activity of Cisplatin in various preclinical models of upper gastrointestinal cancers

Priya Pancholi; Tanmay Dichwalkar; Samhita Bapat; V. J. Rajadhyaksha; Benjamin S. Hoffman; Manoj Maniar; Vikas Sehdev

Background: Upper Gastrointestinal Cancers (UGCs) respond poorly to conventional chemotherapy due to overactive intrinsic mechanisms that mediate drug resistance.] Rigosertib is an investigational anticancer agent, that inhibits cellular signaling by acting as a Ras mimetic to effect the activation of multiple pathways associated with oncogenic transformation and drug resistance. Previous studies have demonstrated the inhibitory effect of rigosertib with conventional platinum based anticancer agents in various pre-clinical cancer models. We hypothesized that the novel mechanism of action of rigosertib would complement the DNA-damaging activity of Cisplatin (CDDP), the standard of care in UGC. To test this hypothesis, we investigated the potential therapeutic benefit of rigosertib alone and in combination with CDDP in P53 wild type and mutant models of UGCs. Methods: For this study, we evaluated the effect of rigosertib treatment alone and/or in combination with CDDP on AGS (P53 wild type) and FLO-1 (P53 mutant) UGC cell viability, survival, and expression of apoptotic markers. The MTT cell viability assay and Compusyn mediated median effect plot analysis (MEPA)(Chou and Talaly) were used to determine synergistic drug combinations of rigosertib and CDDP in AGS and FLO-1 UGC cells, respectively. Results: The cell viability data and MEPA indicated that rigosertib and CDDP show optimal synergistic anticancer activity in both AGS and FLO-1 UGC cells at a ratio of 1:10, respectively. The clonogenic cell survival assay data showed that in comparison to treatment with Rigosertib (AGS: 100nM; FLO-1: 50nM) or CDDP (AGS: 1000nM; FLO-1: 500nM) alone, the combination treatment at a ratio of 1:10 significantly increased (p Citation Format: Priya Pancholi, Tanmay Dichwalkar, Samhita Bapat, V. J. Rajadhyaksha, Benjamin S. Hoffman, Manoj Maniar, Vikas Sehdev. Rigosertib synergistically enhances the anticancer activity of Cisplatin in various preclinical models of upper gastrointestinal cancers. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4687.


Molecular Cancer Therapeutics | 2015

Abstract LB-A21: Single-agent activity and favorable pharmaceutical properties of orally bioavailable next-generation CDK4/6 inhibitor, ON 123300

Benjamin S. Hoffman; Ramana Reddy; Muralidhar R. Mallireddigari; Daniel Fox; Chen Ren; Manoj Maniar

Introduction: Recent proof-of-concept for targeting cyclin dependent kinases 4 and 6 (CDK4/6) culminated in the approval for breast cancer of the first-in-class CDK4/6 inhibitor, palbociclib (Ibrance®). Palbociclib and other potent but narrowly targeted CDK4/6 inhibitors are cytostatic rather than cytotoxic (Leonard et al, 2012), and require combination therapy for optimal activity. ON 123300 is a next-generation CDK4/6 inhibitor with improved single-agent cytotoxicity. This novel compound is a potent CDK4 and CDK6 inhibitor (IC50 = 3.9 nM and 9.8 nM, respectively) that is cytotoxic against breast and other cancer cell lines (Reddy et al, 2014). We present pre-IND profiling pharmacology, physicochemical and ADME studies to support ON 123300 as a clinical candidate and next-generation CDK4/6 inhibitor. Methods and Results: Non-clinical studies were conducted in order to assess the drug-like properties of ON 123300 and its pharmaceutical salts. Physicochemical parameters were determined for the compound. ON 123300 was highly soluble below pH 4.0, with reduced solubility in more basic conditions. Predicted intestinal absorption, as measured by Caco-2 permeability, was 1.95×10-5 cm/s with a low efflux ratio. Based on these characteristics, the compound was advanced to lead profiling against 68 primary molecular targets. Radioligand binding studies revealed limited receptor inhibition (11/68 targets) with 10 μM of ON 123300. Human Ether-a-go-go (hERG) inhibition testing revealed no significant effect (IC50 > 25 μM). In vitro metabolism studies indicated metabolism through CYP3A4 and CYP2C8. Finally, in silico based modeling (Cloe® PK) integrating all key non-clinical parameters was carried out in order to estimate the potential for oral absorption in humans. Preliminary analysis suggests good oral bioavailability and the potential to establish an acceptable therapeutic window. Conclusions: The well differentiated biochemical, anticancer and pharmaceutical properties of ON 123300 provide the basis for pre-clinical toxicology testing, leading to an investigational new drug (IND) filing and clinical exploration of this compound across tumor types that share targets accessed by this next-generation CDK inhibitor. Citation Format: Benjamin Hoffman, Ramana Reddy, Muralidhar Mallireddigari, Daniel Fox, Chen Ren, Manoj Maniar. Single-agent activity and favorable pharmaceutical properties of orally bioavailable next-generation CDK4/6 inhibitor, ON 123300. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr LB-A21.


Cancer Research | 2015

Abstract 1649: Potent anticancer activity of an orally bioavailable small molecule, ON 013100, and its water soluble derivative, briciclib, a clinical-stage eIF4E-targeted agent

Neel Jasani; Bina Desai; Justine M. Betzu; Tanmay Dichwalkar; Samhita Bapat; V. J. Rajadhayksha; Benjamin S. Hoffman; Manoj Maniar; Vikas Sehdev

Introduction: Eukaryotic translation initiation factor 4E (eIF4E) is a master regulator that controls translation of mRNA in mammalian cells. eIF4E is a proto-oncogene that promotes translation of several genes essential for cellular proliferation (cyclin D1, c-Myc, mTOR), survival (Akt, survivin), angiogenesis (VEGF), and metastasis (MMP9). Overexpression of eIF4E has been observed in almost all major groups of cancers and has been shown to induce increased expression of cyclin D1 and c-Myc. Briciclib is a small molecule, water soluble derivative of ON 013100 that binds to eIF4E. An intravenous formulation of briciclib is currently being investigated in a Phase 1 clinical trial. Recent advancements in formulation technology have made feasible a stable, orally bioavailable version of ON 013100, which may allow for more convenient administration. In this study we investigated and compared the anticancer activity of briciclib to ON 013100. We determined the susceptibility of various breast, mantle cell leukemia (MCL), gastric, and esophageal cancer cell lines to treatment with briciclib or ON 013100. In addition, we investigated the effect of briciclib and ON 013100 on expression of markers associated with eIF4E activity (cyclin D1 and c-Myc) and apoptosis (P53 and Cleaved Caspase 3). Methods: MTT cell viability assays, Western blot analysis, and ELISA assays were used to evaluate cellular viability, survival, and protein expression levels. Results: Briciclib and ON 013100 inhibited the proliferation of MCL (JEKO-1 and MINO), breast (MCF7 and MDA-MB-231), gastric (AGS), and esophageal (OE19, OE33, and FLO-1) cancer cell lines at nanomolar concentrations (Briciclib: GI50 = 9.8 - 12.2 nM; ON 013100 GI50 = 6.7 - 11.2 nM). By comparison, briciclib and ON 013100 were relatively non-toxic to normal endothelial cells. Western blot analysis indicated that treatment with briciclib or ON 013100 significantly reduced the expression of cyclin D1 and c-Myc in breast and MCL cancer cell lines within 8 hours and in a dose-dependent manner. These observations were supported by ELISA analysis of cyclin D1 and c-Myc protein levels. Furthermore, treatment with these agents enhanced the expression of P53 and Cleaved Caspase 3 pro-apoptotic proteins in breast and MCL cancer cell lines. Our ongoing tumor xenograft experiments are in agreement with the aforementioned in vitro observations. Conclusions: Our findings suggest that both an orally bioavailable ON 013100, and its water soluble derivative, briciclib, have the same novel mechanism of action involving translation. Our in vitro and in vivo data demonstrate the potential of briciclib in targeting eIF4E for hematopoietic and solid cancers and the possibility for developing an oral version of this promising clinical agent. Citation Format: Neel Jasani, Bina Desai, Justine M. Betzu, Tanmay Dichwalkar, Samhita Bapat, V. J. Rajadhayksha, Benjamin S. Hoffman, Manoj Maniar, Vikas Sehdev. Potent anticancer activity of an orally bioavailable small molecule, ON 013100, and its water soluble derivative, briciclib, a clinical-stage eIF4E-targeted agent. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1649. doi:10.1158/1538-7445.AM2015-1649

Collaboration


Dive into the Manoj Maniar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonio Jimeno

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dara L. Aisner

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wells A. Messersmith

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Wen Wee Ma

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Azra Raza

Columbia University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge