Minal Barve
Texas Oncology
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Minal Barve.
Molecular Therapy | 2013
Neil Senzer; John J. Nemunaitis; Derek Nemunaitis; Cynthia Bedell; Gerald Edelman; Minal Barve; Robert Nunan; Kathleen F. Pirollo; Antonina Rait; Esther H. Chang
Selective delivery of therapeutic molecules to primary and metastatic tumors is optimal for effective cancer therapy. A liposomal nanodelivery complex (scL) for systemic, tumor-targeting delivery of anticancer therapeutics has been developed. scL employs an anti-transferrin receptor (TfR), scFv as the targeting molecule. Loss of p53 suppressor function, through mutations or inactivation of the p53 pathway, is present in most human cancers. Rather than being transiently permissive for tumor initiation, persistence of p53 dysfunction is a continuing requirement for maintaining tumor growth. Herein, we report results of a first-in-man Phase I clinical trial of restoration of the normal human tumor suppressor gene p53 using the scL nanocomplex (SGT-53). Minimal side effects were observed in this trial in patients with advanced solid tumors. Furthermore, the majority of patients demonstrated stable disease. One patient with adenoid cystic carcinoma had his status changed from unresectable to resectable after one treatment cycle. More significantly, we observed an accumulation of the transgene in metastatic tumors, but not in normal skin tissue, in a dose-related manner. These results show not only that systemically delivered SGT-53 is well tolerated and exhibits anticancer activity, but also supply evidence of targeted tumor delivery of SGT-53 to metastatic lesions.
Gynecologic Oncology | 2011
Katherine M. Bell-McGuinn; Carolyn M. Matthews; Steffan Ho; Minal Barve; Lucy Gilbert; Richard T. Penson; Ernst Lengyel; Rameshraja Palaparthy; Kye Gilder; Artemios Vassos; William McAuliffe; Sara Weymer; Jeremy Barton; Russell J. Schilder
OBJECTIVE This phase II, multicenter, single-arm, two-stage study in platinum-resistant, advanced epithelial ovarian or primary peritoneal cancer evaluated the efficacy, safety, and tolerability of weekly single-agent volociximab. Pharmacokinetic/pharmacodynamic (PK/PD) studies were also performed. METHODS Sixteen patients were enrolled in Stage 1. Volociximab was administered at 15mg/kg IV qwk until progression of disease or drug intolerability. Tumor response was assessed every 8weeks. Serum samples for PK or whole blood for the evaluation of circulating tumor cells, endothelial cells, and endothelial progenitor cells were obtained on Days 1, 8, 15, 29, and 50. Ascites from one patient was collected for volociximab concentration analysis. Archived tumor tissue was analyzed by immunohistochemistry (IHC) for α5 integrin expression. RESULTS Safety data are available on all 16 patients; 14 were evaluable for efficacy. One patient had stable disease at 8weeks. The remaining 13 progressed on treatment. Twelve patients (75%) experienced study-related adverse events (AEs); the most common (≥20%) were headache and fatigue. Three patients experienced possible study-related serious AEs (SAEs): reversible posterior leukoencephalopathy syndrome, pulmonary embolism, and hyponatremia. Peak serum concentrations of volociximab increased 2-3 fold from Day 1 to Day 50. Clinically relevant trough levels were achieved (>150μg/mL). IHC analysis of archived tumor sections showed low-to-moderate expression of α5 integrin on all ovarian cancer tissue evaluated. CONCLUSION Despite insufficient clinical activity in this refractory patient population to continue the study, weekly volociximab was well tolerated, and the gained understanding of the mechanism of action of volociximab will inform future development efforts.
Molecular Therapy | 2012
Neil Senzer; Minal Barve; Joseph A. Kuhn; Anton Melnyk; Peter Beitsch; Martin Lazar; Samuel Lifshitz; Mitchell Magee; Jonathan Oh; Susan W Mill; Cynthia Bedell; Candice Higgs; Padmasini Kumar; Yang Yu; Fabienne Norvell; Connor Phalon; Nicolas Taquet; Donald Rao; Zhaohui Wang; Chris M. Jay; Beena O. Pappen; Gladice Wallraven; F. Charles Brunicardi; David M. Shanahan; Phillip B. Maples; John Nemunaitis
We performed a phase I trial of FANG vaccine, an autologous tumor-based product incorporating a plasmid encoding granulocyte-macrophage colony-stimulating factor (GMCSF) and a novel bifunctional short hairpin RNAi (bi-shRNAi) targeting furin convertase, thereby downregulating endogenous immunosuppressive transforming growth factors (TGF) β1 and β2. Patients with advanced cancer received up to 12 monthly intradermal injections of FANG vaccine (1 × 107 or 2.5 × 107 cells/ml injection). GMCSF, TGFβ1, TGFβ2, and furin proteins were quantified by enzyme-linked immunosorbent assay (ELISA). Safety and response were monitored. Vaccine manufacturing was successful in 42 of 46 patients of whom 27 received ≥1 vaccine. There were no treatment-related serious adverse events. Most common grade 1, 2 adverse events included local induration (n = 14) and local erythema (n = 11) at injection site. Post-transfection mean product expression GMCSF increased from 7.3 to 1,108 pg/106 cells/ml. Mean TGFβ1 and β2 effective target knockdown was 93.5 and 92.5% from baseline, respectively. Positive enzyme-linked immunospot (ELISPOT) response at month 4 was demonstrated in 9 of 18 patients serially assessed and correlated with survival duration from time of treatment (P = 0.025). Neither dose-adverse event nor dose-response relationship was noted. In conclusion, FANG vaccine was safe and elicited an immune response correlating with prolonged survival. Phase II assessment is justified.We performed a phase I trial of FANG vaccine, an autologous tumor-based product incorporating a plasmid encoding granulocyte-macrophage colony-stimulating factor (GMCSF) and a novel bifunctional short hairpin RNAi (bi-shRNAi) targeting furin convertase, thereby downregulating endogenous immunosuppressive transforming growth factors (TGF) β1 and β2. Patients with advanced cancer received up to 12 monthly intradermal injections of FANG vaccine (1 × 10(7) or 2.5 × 10(7) cells/ml injection). GMCSF, TGFβ1, TGFβ2, and furin proteins were quantified by enzyme-linked immunosorbent assay (ELISA). Safety and response were monitored. Vaccine manufacturing was successful in 42 of 46 patients of whom 27 received ≥1 vaccine. There were no treatment-related serious adverse events. Most common grade 1, 2 adverse events included local induration (n = 14) and local erythema (n = 11) at injection site. Post-transfection mean product expression GMCSF increased from 7.3 to 1,108 pg/10(6) cells/ml. Mean TGFβ1 and β2 effective target knockdown was 93.5 and 92.5% from baseline, respectively. Positive enzyme-linked immunospot (ELISPOT) response at month 4 was demonstrated in 9 of 18 patients serially assessed and correlated with survival duration from time of treatment (P = 0.025). Neither dose-adverse event nor dose-response relationship was noted. In conclusion, FANG vaccine was safe and elicited an immune response correlating with prolonged survival. Phase II assessment is justified.
Molecular Therapy | 2016
Maurizio Ghisoli; Minal Barve; Robert G. Mennel; Carl Lenarsky; Staci Horvath; Gladice Wallraven; Beena O. Pappen; Sam Whiting; Donald Rao; Neil Senzer; John Nemunaitis
Ewings sarcoma is a devastating rare pediatric cancer of the bone. Intense chemotherapy temporarily controls disease in most patients at presentation but has limited effect in patients with progressive or recurrent disease. We previously described preliminary results of a novel immunotherapy, FANG (Vigil) vaccine, in which 12 advanced stage Ewings patients were safely treated and went on to achieve a predicted immune response (IFNγ ELISPOT). We describe follow-up through year 3 of a prospective, nonrandomized study comparing an expanded group of Vigil-treated advanced disease Ewings sarcoma patients (n = 16) with a contemporaneous group of Ewings sarcoma patients (n = 14) not treated with Vigil. Long-term follow-up results show a survival benefit without evidence of significant toxicity (no ≥ grade 3) to Vigil when administered once monthly by intradermal injection (1 × 10e(6) cells/injection to 1 × 10e(7) cells/injection). Specifically, we report a 1-year actual survival of 73% for Vigil-treated patients compared to 23% in those not treated with Vigil. In addition, there was a 17.2-month difference in overall survival (OS; Kaplan-Meier) between the Vigil (median OS 731 days) and no Vigil patient groups (median OS 207 days). In conclusion, these results supply the rational for further testing of Vigil in advanced stage Ewings sarcoma.Ewings sarcoma is a devastating rare pediatric cancer of the bone. Intense chemotherapy temporarily controls disease in most patients at presentation but has limited effect in patients with progressive or recurrent disease. We previously described preliminary results of a novel immunotherapy, FANG (Vigil) vaccine, in which 12 advanced stage Ewings patients were safely treated and went on to achieve a predicted immune response (IFNγ ELISPOT). We describe follow-up through year 3 of a prospective, nonrandomized study comparing an expanded group of Vigil-treated advanced disease Ewings sarcoma patients (n = 16) with a contemporaneous group of Ewings sarcoma patients (n = 14) not treated with Vigil. Long-term follow-up results show a survival benefit without evidence of significant toxicity (no ≥ grade 3) to Vigil when administered once monthly by intradermal injection (1 × 10e6 cells/injection to 1 × 10e7 cells/injection). Specifically, we report a 1-year actual survival of 73% for Vigil-treated patients compared to 23% in those not treated with Vigil. In addition, there was a 17.2-month difference in overall survival (OS; Kaplan-Meier) between the Vigil (median OS 731 days) and no Vigil patient groups (median OS 207 days). In conclusion, these results supply the rational for further testing of Vigil in advanced stage Ewings sarcoma.
Molecular Therapy | 2015
Maurizio Ghisoli; Minal Barve; Reva Schneider; Robert G. Mennel; Carl Lenarsky; Gladice Wallraven; Beena O. Pappen; John LaNoue; Padmasini Kumar; Derek Nemunaitis; Alyssa Roth; James Nemunaitis; Sam Whiting; Neil Senzer; Frederick A. Fletcher; John Nemunaitis
We report on 12 consecutive patients with advanced/metastatic Ewings sarcoma who were treated as a separate cohort of a phase 1 trial of FANG autologous immunotherapy (1 × 106–2.5 × 107 cells/intradermal injection each month for minimum 4 months). Safety and clinical response were monitored. Patient immune response to unmodified autologous tumor cells was assessed by gamma interferon-enzyme-linked immunospot (γIFN-ELISPOT) assay using peripheral blood mononuclear cells from baseline (pretreatment) and multiple postvaccination time points. None of the 12 patients (47 vaccinations) developed grade 2/3/4 drug-related toxicity. Median product release granulocyte-macrophage colony-stimulating factor expression was 1,941 pg/106 cells, and TGFβ1and TGFβ2 knockdown were 99 and 100%, respectively. Eight patients were assessed for ELISPOT response to autologous tumor cells at baseline and all (100%) were negative. In contrast, follow-up ELISPOT response at month 1 or month 4 (one patient) after FANG was positive in all eight patients. One patient achieved a partial tumor response (38% tumor reduction, RECIST 1.1). The Kaplan–Meier estimated survival of these 12 patients at 1 year was 75%. In this phase 1 study in patients with Ewings sarcoma, FANG immunotherapy was well tolerated, elicited a tumor-specific systemic immune response in all patients, and was associated with favorable 1-year survival. Further clinical testing is indicated.
Gynecologic Oncology | 2016
Jonathan Oh; Minal Barve; Carolyn M. Matthews; E. Colin Koon; Thomas P. Heffernan; Bruce Fine; Elizabeth A. Grosen; Melanie K. Bergman; Evelyn L Fleming; Leslie R. DeMars; Loyd West; Daniel L. Spitz; Howard M. Goodman; Kenneth C. Hancock; Gladice Wallraven; Padmasini Kumar; Ernest Bognar; Luisa Manning; Beena O. Pappen; Ned Adams; Neil Senzer; John Nemunaitis
OBJECTIVES The majority of women with Stage III/IV ovarian cancer who achieve clinical complete response with frontline standard of care will relapse within 2years. Vigil immunotherapy, a GMCSF/bi-shRNA furin DNA engineered autologous tumor cell (EATC) product, demonstrated safety and induction of circulating activated T-cells against autologous tumor in Phase I trial Senzer et al. (2012, 2013) . Our objectives for this study include evaluation of safety, immune response and recurrence free survival (RFS). METHODS This is a Phase II crossover trial of Vigil (1.0×107 cells/intradermal injection/month for 4 to 12 doses) in Stage III/IV ovarian cancer patients achieving cCR (normal imaging, CA-125≤35units/ml, physical exam, and no symptoms suggestive of the presence of active disease) following primary surgical debulking and carboplatin/paclitaxel adjuvant or neoadjuvant chemotherapy. Patients received Vigil or standard of care during the maintenance period. RESULTS Forty-two patients were entered into trial, 31 received Vigil and 11 received standard of care. No≥Grade 3 toxicity related to product was observed. A marked induction of circulating activated T-cell population was observed against individual, pre-processed autologous tumor in the Vigil arm as compared to pre-Vigil baseline using IFNγ ELISPOT response (30/31 negative ELISPOT pre Vigil to 31/31 positive ELISPOT post Vigil, median 134 spots). Moreover, in correlation with ELISPOT response, RFS from time of procurement was improved (mean 826days/median 604days in the Vigil arm from mean 481days/median 377days in the control arm, p=0.033). CONCLUSION In conjunction with the demonstrated safety, the high rate of induction of T-cell activation and correlation with improvement in RFS justify further Phase II/III assessment of Vigil.
International Journal of Gynecology & Obstetrics | 2000
Jonathan Oh; Minal Barve; W. Michael Lin; Robert L. Coleman; Joseph T. Santoso; Carolyn Y. Muller; Diana Ferrar; Melanie King; Enoch Lowe; David Scott
Study Methods: 942 with hydatidiform mole admitted to our institution were enrolled in a prospective study from June 1990. The influence of age, parity, and the blood group of husband and wife in the epidemiology of hydatidiform mole were analyzed. A post-molar serum b-hCG regression pattern was established for out patients based on whom diagnosis of persistent trophoblastic disease ({TD) was made. The primary management of our patients with FTD is single agent chemotherapy using methotrexate with folinic acid rescue. Those women who have completed one and a half years of follow-up were advised to attempt pregnancy if they desired so. Results: From June 1990 to May 1998,942 women with hydatidiform mole were admitted to our institution. During this period 146,803 women delivered in our hospital, the incidence of hydatidiform mole being one out of 1.56 deliveries. Nearly 70% of our patients were below 30 years of age and were concerned about their reproductive future. Of the 942 cases of hydatidiform mole, 252 cases developed FTD, an incidence of 26%. Nearly 70% of the FTD patients required 3 or less courses of methotrexate-folinic acid regimen to achieve remission. methotrexate-folinic acid regimen was well tolerated by our patients. After the follow-up, 245 patients, 40 following chemotherapy have conceived and delivered and the outcome was comparable with general population. Conclusions: Follow-up of hydatidiform mole by weekly b-hCG assay will enable identification of malignant changes at an early date after evacuation of hydatidiform mole so that single agent chemotherapy will be curative with conservation of reproductive function.
Molecular Therapy | 2015
Minal Barve; Zhaohui Wang; Padmasini Kumar; Christopher M. Jay; Xiuquan Luo; Cynthia Bedell; Robert G. Mennel; Gladice Wallraven; F.C. Brunicardi; Neil Senzer; John Nemunaitis; Donald Rao
Gynecologic Oncology | 2015
Jonathan Oh; Minal Barve; Elizabeth A. Grosen; B.A. Fine; T.P. Heffernan; Carolyn M. Matthews; C.A. Stringer; E.C. Koon; Howard M. Goodman; E.L. Fleming; Leslie R. DeMars; M.S. Bergman; Neil Senzer; John Nemunaitis
Journal of Clinical Oncology | 2018
Erika Paige Hamilton; Minal Barve; Aditya Bardia; Muralidhar Beeram; Johanna C. Bendell; Rebecca Mosher; Eric Hailman; Donald A. Bergstrom; Howard A. Burris; Hatem Soliman