Network


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

Hotspot


Dive into the research topics where Chitra Hosing is active.

Publication


Featured researches published by Chitra Hosing.


Blood | 2009

Plerixafor and G-CSF versus placebo and G-CSF to mobilize hematopoietic stem cells for autologous stem cell transplantation in patients with multiple myeloma.

John F. DiPersio; Edward A. Stadtmauer; Auayporn Nademanee; Ivana N. Micallef; Patrick J. Stiff; Jonathan L. Kaufman; Richard T. Maziarz; Chitra Hosing; Stefan Fruehauf; Mitchell E. Horwitz; Dennis L. Cooper; Gary Bridger; Gary Calandra

This phase 3, multicenter, randomized (1:1), double-blind, placebo-controlled study evaluated the safety and efficacy of plerixafor with granulocyte colony-stimulating factor (G-CSF) in mobilizing hematopoietic stem cells in patients with multiple myeloma. Patients received G-CSF (10 microg/kg) subcutaneously daily for up to 8 days. Beginning on day 4 and continuing daily for up to 4 days, patients received either plerixafor (240 microg/kg) or placebo subcutaneously. Starting on day 5, patients began daily apheresis for up to 4 days or until more than or equal to 6 x 10(6) CD34(+) cells/kg were collected. The primary endpoint was the percentage of patients who collected more than or equal to 6 x 10(6) CD34(+) cells/kg in less than or equal to 2 aphereses. A total of 106 of 148 (71.6%) patients in the plerixafor group and 53 of 154 (34.4%) patients in the placebo group met the primary endpoint (P < .001). A total of 54% of plerixafor-treated patients reached target after one apheresis, whereas 56% of the placebo-treated patients required 4 aphereses to reach target. The most common adverse events related to plerixafor were gastrointestinal disorders and injection site reactions. Plerixafor and G-CSF were well tolerated, and significantly more patients collected the optimal CD34(+) cell/kg target for transplantation earlier compared with G-CSF alone. This study is registered at www.clinicaltrials.gov as #NCT00103662.


Journal of Clinical Oncology | 2013

Disabling Immune Tolerance by Programmed Death-1 Blockade With Pidilizumab After Autologous Hematopoietic Stem-Cell Transplantation for Diffuse Large B-Cell Lymphoma: Results of an International Phase II Trial

Philippe Armand; Arnon Nagler; Edie Weller; Steven M. Devine; David Avigan; Yi-Bin Chen; Mark S. Kaminski; H. Kent Holland; Jane N. Winter; James Mason; Joseph W. Fay; David A. Rizzieri; Chitra Hosing; Edward D. Ball; Joseph P. Uberti; Hillard M. Lazarus; Markus Y. Mapara; Stephanie A. Gregory; John M. Timmerman; David J. Andorsky; Reuven Or; Edmund K. Waller; Rinat Rotem-Yehudar; Leo I. Gordon

PURPOSE The Programmed Death-1 (PD-1) immune checkpoint pathway may be usurped by tumors, including diffuse large B-cell lymphoma (DLBCL), to evade immune surveillance. The reconstituting immune landscape after autologous hematopoietic stem-cell transplantation (AHSCT) may be particularly favorable for breaking immune tolerance through PD-1 blockade. PATIENTS AND METHODS We conducted an international phase II study of pidilizumab, an anti-PD-1 monoclonal antibody, in patients with DLBCL undergoing AHSCT, with correlative studies of lymphocyte subsets. Patients received three doses of pidilizumab beginning 1 to 3 months after AHSCT. RESULTS Sixty-six eligible patients were treated. Toxicity was mild. At 16 months after the first treatment, progression-free survival (PFS) was 0.72 (90% CI, 0.60 to 0.82), meeting the primary end point. Among the 24 high-risk patients who remained positive on positron emission tomography after salvage chemotherapy, the 16-month PFS was 0.70 (90% CI, 0.51 to 0.82). Among the 35 patients with measurable disease after AHSCT, the overall response rate after pidilizumab treatment was 51%. Treatment was associated with increases in circulating lymphocyte subsets including PD-L1E-bearing lymphocytes, suggesting an on-target in vivo effect of pidilizumab. CONCLUSION This is the first demonstration of clinical activity of PD-1 blockade in DLBCL. Given these results, PD-1 blockade after AHSCT using pidilizumab may represent a promising therapeutic strategy in this disease.


The New England Journal of Medicine | 2012

Cord-blood engraftment with ex vivo mesenchymal-cell coculture.

Marcos de Lima; Ian McNiece; Simon N. Robinson; Mark F. Munsell; Mary Eapen; Mary M. Horowitz; Amin M. Alousi; Rima M. Saliba; John McMannis; Indreshpal Kaur; Partow Kebriaei; Simrit Parmar; Uday Popat; Chitra Hosing; Richard E. Champlin; Catherine M. Bollard; Jeffrey J. Molldrem; Roy B. Jones; Yago Nieto; Borje S. Andersson; Nina Shah; Betul Oran; Laurence J.N. Cooper; Laura L. Worth; Muzaffar H. Qazilbash; Martin Korbling; Gabriela Rondon; Stefan O. Ciurea; Doyle Bosque; I. Maewal

BACKGROUND Poor engraftment due to low cell doses restricts the usefulness of umbilical-cord-blood transplantation. We hypothesized that engraftment would be improved by transplanting cord blood that was expanded ex vivo with mesenchymal stromal cells. METHODS We studied engraftment results in 31 adults with hematologic cancers who received transplants of 2 cord-blood units, 1 of which contained cord blood that was expanded ex vivo in cocultures with allogeneic mesenchymal stromal cells. The results in these patients were compared with those in 80 historical controls who received 2 units of unmanipulated cord blood. RESULTS Coculture with mesenchymal stromal cells led to an expansion of total nucleated cells by a median factor of 12.2 and of CD34+ cells by a median factor of 30.1. With transplantation of 1 unit each of expanded and unmanipulated cord blood, patients received a median of 8.34×10(7) total nucleated cells per kilogram of body weight and 1.81×10(6) CD34+ cells per kilogram--doses higher than in our previous transplantations of 2 units of unmanipulated cord blood. In patients in whom engraftment occurred, the median time to neutrophil engraftment was 15 days in the recipients of expanded cord blood, as compared with 24 days in controls who received unmanipulated cord blood only (P<0.001); the median time to platelet engraftment was 42 days and 49 days, respectively (P=0.03). On day 26, the cumulative incidence of neutrophil engraftment was 88% with expansion versus 53% without expansion (P<0.001); on day 60, the cumulative incidence of platelet engraftment was 71% and 31%, respectively (P<0.001). CONCLUSIONS Transplantation of cord-blood cells expanded with mesenchymal stromal cells appeared to be safe and effective. Expanded cord blood in combination with unmanipulated cord blood significantly improved engraftment, as compared with unmanipulated cord blood only. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT00498316.).


Blood | 2008

Eight-year experience with allogeneic stem cell transplantation for relapsed follicular lymphoma after nonmyeloablative conditioning with fludarabine, cyclophosphamide, and rituximab

Issa F. Khouri; Peter McLaughlin; Rima M. Saliba; Chitra Hosing; Martin Korbling; Ming S. Lee; L. Jeffrey Medeiros; Luis Fayad; Felipe Samaniego; Amin M. Alousi; Paolo Anderlini; Daniel R. Couriel; Marcos de Lima; Sergio Giralt; Sattva S. Neelapu; Naoto Ueno; Barry I. Samuels; Fredrick B. Hagemeister; Larry W. Kwak; Richard E. Champlin

Nonmyeloablative stem cell transplantation in patients with follicular lymphoma has been designed to exploit the graft-versus-lymphoma immunity. The long-term effectiveness and toxicity of this strategy, however, is unknown. In this prospective study, we analyzed our 8-year experience. Patients received a conditioning regimen of fludarabine (30 mg/m(2) daily for 3 days), cyclophosphamide (750 mg/m(2) daily for 3 days), and rituximab (375 mg/m(2) for 1 day plus 1000 mg/m(2) for 3 days). They were then given an infusion of human leukocyte antigen-matched hematopoietic cells from related (n = 45) or unrelated donors (n = 2). Tacrolimus and methotrexate were used for graft-versus-host disease (GVHD) prophylaxis. Forty-seven patients were included. All patients experienced complete remission, with only 2 relapses. With a median follow-up time of 60 months (range, 19-94), the estimated survival and progression-free survival rates were 85% and 83%, respectively. All 18 patients who were tested and had evidence of JH/bcl-2 fusion transcripts in the bone marrow at study entry experienced continuous molecular remission. The incidence of grade 2-IV acute GVHD was 11%. Only 5 patients were still undergoing immunosuppressive therapy at the time of last follow-up. We believe that the described results are a step forward toward developing a curative strategy for recurrent follicular lymphoma.


Blood | 2013

Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study

Conrad Russell Y. Cruz; Kenneth P. Micklethwaite; Barbara Savoldo; Carlos A. Ramos; Sharon Lam; Stephanie Ku; Oumar Diouf; Enli Liu; A. John Barrett; Sawa Ito; Elizabeth J. Shpall; Robert A. Krance; Rammurti T. Kamble; George Carrum; Chitra Hosing; Adrian P. Gee; Zhuyong Mei; Bambi Grilley; Helen E. Heslop; Cliona M. Rooney; Malcolm K. Brenner; Catherine M. Bollard; Gianpietro Dotti

Autologous T cells expressing a CD19-specific chimeric antigen receptor (CD19.CAR) are active against B-cell malignancies, but it is unknown whether allogeneic CD19.CAR T cells are safe or effective. After allogeneic hematopoietic stem cell transplantation (HSCT), infused donor-derived virus-specific T cells (VSTs) expand in vivo, persist long term, and display antiviral activity without inducing graft-vs-host disease; therefore, we determined whether donor VSTs, engineered to express CD19.CAR, retained the characteristics of nonmanipulated allogeneic VSTs while gaining antitumor activity. We treated 8 patients with allogeneic (donor-derived) CD19.CAR-VSTs 3 months to 13 years after HSCT. There were no infusion-related toxicities. VSTs persisted for a median of 8 weeks in blood and up to 9 weeks at disease sites. Objective antitumor activity was evident in 2 of 6 patients with relapsed disease during the period of CD19.CAR-VST persistence, whereas 2 patients who received cells while in remission remain disease free. In 2 of 3 patients with viral reactivation, donor CD19.CAR-VSTs expanded concomitantly with VSTs. Hence CD19.CAR-VSTs display antitumor activity and, because their number may be increased in the presence of viral stimuli, earlier treatment post-HSCT (when lymphodepletion is greater and the incidence of viral infection is higher) or planned vaccination with viral antigens may enhance disease control.


Bone Marrow Transplantation | 2008

Transplantation of ex vivo expanded cord blood cells using the copper chelator tetraethylenepentamine: a phase I/II clinical trial.

M. de Lima; John McMannis; Adrian P. Gee; Krishna V. Komanduri; Daniel R. Couriel; Borje S. Andersson; Chitra Hosing; Issa F. Khouri; Roy B. Jones; Richard E. Champlin; S. Karandish; Tara Sadeghi; T. Peled; F. Grynspan; Y. Daniely; Arnon Nagler; Elizabeth J. Shpall

The copper chelator tetraethylenepentamine (TEPA; StemEx) was shown to attenuate the differentiation of ex vivo cultured hematopoietic cells resulting in preferential expansion of early progenitors. A phase I/II trial was performed to test the feasibility and safety of transplantation of CD133+ cord blood (CB) hematopoietic progenitors cultured in media containing stem cell factor, FLT-3 ligand, interleukin-6, thrombopoietin and TEPA. Ten patients with advanced hematological malignancies were transplanted with a CB unit originally frozen in two fractions. The smaller fraction was cultured ex vivo for 21 days and transplanted 24 h after infusion of the larger unmanipulated fraction. All but two units contained <2 × 107 total nucleated cells (TNCs) per kilogram pre-expansion. All donor–recipient pairs were mismatched for one or two HLA loci. Nine patients were beyond first remission; median age and weight were 21 years and 68.5 kg. The average TNCs fold expansion was 219 (range, 2–620). Mean increase of CD34+ cell count was 6 (over the CD34+ cell content in the entire unit). Despite the low TNCs per kilogram infused (median=1.8 × 107/kg), nine patients engrafted. Median time to neutrophil and platelet engraftment was 30 (range, 16–46) and 48 (range, 35–105) days. There were no cases of grades 3–4 acute graft-versus-host disease (GVHD) and 100-day survival was 90%. This strategy is feasible.


Blood | 2009

Mature results of the M. D. Anderson Cancer Center risk-adapted transplantation strategy in mantle cell lymphoma

Constantine S. Tam; Roland L. Bassett; Celina Ledesma; Martin Korbling; Amin M. Alousi; Chitra Hosing; Partow Kebraei; Robyn Harrell; Gabriela Rondon; Sergio Giralt; Paolo Anderlini; Uday Popat; Barbara Pro; Barry I. Samuels; Fredrick B. Hagemeister; L. Jeffrey Medeiros; Richard E. Champlin; Issa F. Khouri

In this study, we analyzed the long-term outcome of a risk-adapted transplantation strategy for mantle cell lymphoma in 121 patients enrolled in sequential transplantation protocols. Notable developments over the 17-year study period were the addition of rituximab to chemotherapy and preparative regimens and the advent of nonmyeloablative allogeneic stem cell transplantation (NST). In the autologous transplantation group (n = 86), rituximab resulted in a marked improvement in progression-free survival for patients who received a transplant in their first remission (where a plateau emerged at 3-8 years) but did not change the outcomes for patients who received a transplant beyond their first remission. In the NST group, composed entirely of patients who received a transplant beyond their first remission, durable remissions also emerged in progression-free survival at 5 to 9 years. The major determinants of disease control after NST were the use of a peripheral blood stem cell graft and donor chimerism of at least 95%, whereas the major determinant of death was immunosuppression for chronic graft-versus-host disease. Our results show that long-term disease-free survival in mantle cell lymphoma is possible after rituximab-containing autologous transplantation for patients in first remission and after NST for patients with relapsed or refractory disease.


Biology of Blood and Marrow Transplantation | 2012

Improved Early Outcomes Using a T Cell Replete Graft Compared with T Cell Depleted Haploidentical Hematopoietic Stem Cell Transplantation

Stefan O. Ciurea; Victor E. Mulanovich; Rima M. Saliba; Ulas D. Bayraktar; Ying Jiang; Roland L. Bassett; Sa Wang; Marina Konopleva; Marcelo Fernandez-Vina; Nivia Montes; Doyle Bosque; Julianne Chen; Gabriela Rondon; Gheath Alatrash; Amin M. Alousi; Qaiser Bashir; Martin Korbling; Muzaffar H. Qazilbash; Simrit Parmar; Elizabeth J. Shpall; Yago Nieto; Chitra Hosing; Partow Kebriaei; Issa F. Khouri; Uday Popat; Marcos de Lima; Richard E. Champlin

Haploidentical stem cell transplantation (SCT) has been generally performed using a T cell depleted (TCD) graft; however, a high rate of nonrelapse mortality (NRM) has been reported, particularly in adult patients. We hypothesized that using a T cell replete (TCR) graft followed by effective posttransplantation immunosuppressive therapy would reduce NRM and improve outcomes. We analyzed 65 consecutive adult patients with hematologic malignancies who received TCR (N = 32) or TCD (N = 33) haploidentical transplants. All patients received a preparative regimen consisting of melphalan, fludarabine, and thiotepa. The TCR group received posttransplantation treatment with cyclophosphamide (Cy), tacrolimus (Tac), and mycophenolate mofetil (MMF). Patients with TCD received antithymocyte globulin followed by infusion of CD34+ selected cells with no posttransplantation immunosuppression. The majority of patients in each group had active disease at the time of transplantation. Outcomes are reported for the TCR and TCD recipients, respectively. Engraftment was achieved in 94% versus 81% (P = NS). NRM at 1 year was 16% versus 42% (P = .02). Actuarial overall survival (OS) and progression-free survival (PFS) rates at 1 year posttransplantation were 64% versus 30% (P = .02) and 50% versus 21% (P = .02). The cumulative incidence of grade II-IV acute graft-versus-host disease (aGVHD) was 20% versus 11% (P = .20), and chronic GVHD (cGVHD) 7% versus 18% (P = .03). Improved reconstitution of T cell subsets and a lower rate of infection were observed in the TCR group. These results indicate that a TCR graft followed by effective control of GVHD posttransplantation may lower NRM and improve survival after haploidentical SCT.


British Journal of Haematology | 2005

Sirolimus in combination with tacrolimus and corticosteroids for the treatment of resistant chronic graft-versus-host disease.

Daniel R. Couriel; Rima M. Saliba; M. P. Escalón; Yvonne Hsu; S. Ghosh; C. Ippoliti; K. Hicks; M. Donato; Sergio Giralt; Issa F. Khouri; Chitra Hosing; M. de Lima; Borje S. Andersson; J. Neumann; R. Champlin

Chronic graft‐versus‐host disease (cGVHD) remains a major cause of morbidity and mortality in haematopoietic transplant recipients. Sirolimus is a macrocyclic triene antibiotic with immunosuppressive, antifungal and antitumour properties, that has activity in the prevention and treatment of acute GVHD. We conducted a phase II trial of sirolimus combined with tacrolimus and methylprednisolone in patients with steroid‐resistant cGVHD. Thirty‐five patients who developed GVHD after day 100 post‐transplant were studied. Six patients had a complete response and 16 a partial response with an overall response rate of 63%. Major adverse events related to the combination of tacrolimus and sirolimus were hyperlipidaemia, renal dysfunction and cytopenias. Four patients had thrombotic microangiopathy (TMA) and 27 (77%) had infectious complications. The median survival for the whole group was 15 months. A significantly better outcome was observed in patients with a platelet count ≥100 × 109/l, as well as in those with true chronic manifestations of GVHD compared to those with acute GVHD beyond day 100. Controlled trials comparing this approach with alternative strategies to determine which can best achieve the goal of GVHD‐free survival are warranted.


Journal of Clinical Oncology | 2005

Concurrent Administration of High-Dose Rituximab Before and After Autologous Stem-Cell Transplantation for Relapsed Aggressive B-Cell Non-Hodgkin’s Lymphomas

Issa F. Khouri; Rima M. Saliba; Chitra Hosing; Grace Julia Okoroji; Sandra Acholonu; Paolo Anderlini; Daniel R. Couriel; Marcos de Lima; Michele Donato; Luis Fayad; Segio Giralt; Roy B. Jones; Martin Korbling; Farzaneh Maadani; John T. Manning; Barbara Pro; Elizabeth J. Shpall; Anas Younes; Peter McLaughlin; Richard E. Champlin

PURPOSE We investigated the efficacy and safety of administering high-dose rituximab (HD-R) in combination with high-dose carmustine, cytarabine, etoposide, and melphalan chemotherapy and autologous stem-cell transplantation (SCT) in patients with recurrent B-cell aggressive non-Hodgkins lymphoma (NHL). PATIENTS AND METHODS Sixty-seven consecutive patients were treated. Rituximab was administered during stem-cell mobilization (1 day before chemotherapy at 375 mg/m(2) and 7 days after chemotherapy at 1,000 mg/m(2)), together with granulocyte colony-stimulating factor 10 mug/kg and granulocyte-macrophage colony-stimulating factor 250 microg/m(2) administered subcutaneously daily. HD-R of 1,000 mg/m(2) was administered again days 1 and 8 after transplantation. The results of this treatment were retrospectively compared with those of a historical control group receiving the same preparative regimen without rituximab. RESULTS With a median follow-up time for the study group of 20 months, the overall survival rate at 2-years was 80% (95% CI, 65% to 89%) for the study group and 53% (95% CI, 34% to 69%) for the control group (P = .002). Disease-free survival was 67% (95% CI, 51% to 79%) for the study group and 43% (95% CI, 26% to 60%) for the control group (P = .004). The median time to recovery of absolute neutrophil count to >/= 500 cells/microL was 11 days (range, 8 to 37 days) for the rituximab group and 10 days (range, 8 to 17 days) for the matched control group (P = .001). However, infections were not significantly increased in patients treated with rituximab. CONCLUSION The results of this study suggest that using HD-R and autologous SCT is a feasible and promising treatment for patients with B-cell aggressive NHL.

Collaboration


Dive into the Chitra Hosing's collaboration.

Top Co-Authors

Avatar

Richard E. Champlin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Uday Popat

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Muzaffar H. Qazilbash

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Partow Kebriaei

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth J. Shpall

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Issa F. Khouri

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amin M. Alousi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rima M. Saliba

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sergio Giralt

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Marcos de Lima

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge