Network


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

Hotspot


Dive into the research topics where Rammurti T. Kamble is active.

Publication


Featured researches published by Rammurti T. Kamble.


Journal of Clinical Investigation | 2011

CD28 costimulation improves expansion and persistence of chimeric antigen receptor–modified T cells in lymphoma patients

Barbara Savoldo; Carlos A. Ramos; Enli Liu; Martha P. Mims; Michael J. Keating; George Carrum; Rammurti T. Kamble; Catherine M. Bollard; Adrian P. Gee; Zhuyong Mei; Hao Liu; Bambi Grilley; Cliona M. Rooney; Helen E. Heslop; Malcolm K. Brenner; Gianpietro Dotti

Targeted T cell immunotherapies using engineered T lymphocytes expressing tumor-directed chimeric antigen receptors (CARs) are designed to benefit patients with cancer. Although incorporation of costimulatory endodomains within these CARs increases the proliferation of CAR-redirected T lymphocytes, it has proven difficult to draw definitive conclusions about the specific effects of costimulatory endodomains on the expansion, persistence, and antitumor effectiveness of CAR-redirected T cells in human subjects, owing to the lack of side-by-side comparisons with T cells bearing only a single signaling domain. We therefore designed a study that allowed us to directly measure the consequences of adding a costimulatory endodomain to CAR-redirected T cells. Patients with B cell lymphomas were simultaneously infused with 2 autologous T cell products expressing CARs with the same specificity for the CD19 antigen, present on most B cell malignancies. One CAR encoded both the costimulatory CD28 and the ζ-endodomains, while the other encoded only the ζ-endodomain. CAR+ T cells containing the CD28 endodomain showed strikingly enhanced expansion and persistence compared with CAR+ T cells lacking this endodomain. These results demonstrate the superiority of CARs with dual signal domains and confirm a method of comparing CAR-modified T cells within individual patients, thereby avoiding patient-to-patient variability and accelerating the development of optimal T cell immunotherapies.


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.


Journal of Clinical Oncology | 2010

Hematopoietic Stem-Cell Transplantation for Acute Leukemia in Relapse or Primary Induction Failure

Michel Duval; John P. Klein; Wensheng He; Jean Yves Cahn; Mitchell S. Cairo; Bruce M. Camitta; Rammurti T. Kamble; Edward A. Copelan; Marcos de Lima; Vikas Gupta; Armand Keating; Hillard M. Lazarus; Mark R. Litzow; David I. Marks; Richard T. Maziarz; David A. Rizzieri; Gary J. Schiller; Kirk R. Schultz; Martin S. Tallman; Daniel J. Weisdorf

PURPOSE Patients with acute leukemia refractory to induction or reinduction chemotherapy have poor prognoses if they do not undergo hematopoietic stem-cell transplantation (HSCT). However, HSCT when a patient is not in complete remission (CR) is of uncertain benefit. We hypothesized that pretransplantation variables may define subgroups that have a better prognosis. PATIENTS AND METHODS Overall, 2,255 patients who underwent transplantation for acute leukemia in relapse or with primary induction failure after myeloablative conditioning regimen between 1995 and 2004 were reported to the Center for International Blood and Marrow Transplant Research. The median follow-up of survivors was 61 months. We performed multivariate analysis of pretransplantation variables and developed a predictive scoring system for survival. RESULTS The 3-year overall survival (OS) rates were 19% for acute myeloid leukemia (AML) and 16% for acute lymphoblastic leukemia (ALL). For AML, five adverse pretransplantation variables significantly influenced survival: first CR duration less than 6 months, circulating blasts, donor other than HLA-identical sibling, Karnofsky or Lansky score less than 90, and poor-risk cytogenetics. For ALL, survival was worse with the following: first refractory or second or greater relapse, > or = 25% marrow blasts, cytomegalovirus-seropositive donor, and age of 10 years or older. Patients with AML who had a predictive score of 0 had 42% OS at 3 years, whereas OS was 6% for a score > or = 3. Patients with ALL who had a score of 0 or 1 had 46% 3-year OS but only 10% OS rate for a score > or = 3. CONCLUSION Pretransplantation variables delineate subgroups with different outcomes. HSCT during relapse can achieve long-term survival in selected patients with acute leukemia.


Reviews in Medical Virology | 2012

Chromosomally integrated human herpesvirus 6: questions and answers

Philip E. Pellett; Dharam V. Ablashi; Peter F. Ambros; Henri Agut; Mary T. Caserta; Vincent Descamps; Louis Flamand; Agnès Gautheret-Dejean; Caroline B. Hall; Rammurti T. Kamble; Uwe Kuehl; Dirk Lassner; Irmeli Lautenschlager; Kristin S. Loomis; Mario Luppi; Paolo Lusso; Peter G. Medveczky; Jose G. Montoya; Yasuko Mori; Masao Ogata; Joshua C. Pritchett; Sylvie Rogez; Edward Seto; Katherine N. Ward; Tetsushi Yoshikawa; Raymund R. Razonable

Chromosomally integrated human herpesvirus 6 (ciHHV‐6) is a condition in which the complete HHV‐6 genome is integrated into the host germ line genome and is vertically transmitted in a Mendelian manner. The condition is found in less than 1% of controls in the USA and UK, but has been found at a somewhat higher prevalence in transplant recipients and other patient populations in several small studies. HHV‐6 levels in whole blood that exceed 5.5 log10 copies/ml are strongly suggestive of ciHHV‐6. Monitoring DNA load in plasma and serum is unreliable, both for identifying and for monitoring subjects with ciHHV‐6 due to cell lysis and release of cellular DNA. High HHV‐6 DNA loads associated with ciHHV‐6 can lead to erroneous diagnosis of active infection. Transplant recipients with ciHHV‐6 may be at increased risk for bacterial infection and graft rejection. ciHHV‐6 can be induced to a state of active viral replication in vitro. It is not known whether ciHHV‐6 individuals are put at clinical risk by the use of drugs that have been associated with HHV‐6 reactivation in vivo or in vitro. Nonetheless, we urge careful observation when use of such drugs is indicated in individuals known to have ciHHV‐6. Little is known about whether individuals with ciHHV‐6 develop immune tolerance for viral proteins. Further research is needed to determine the role of ciHHV‐6 in disease. Copyright


Blood | 2013

Better leukemia-free and overall survival in AML in first remission following cyclophosphamide in combination with busulfan compared with TBI

Edward A. Copelan; Betty K. Hamilton; Belinda R. Avalos; Kwang Woo Ahn; Brian J. Bolwell; Xiaochun Zhu; Mahmoud Aljurf; Koen van Besien; Christopher Bredeson; Jean-Yves Cahn; Luciano J. Costa; Marcos de Lima; Robert Peter Gale; Gregory A. Hale; Joerg Halter; Mehdi Hamadani; Yoshihiro Inamoto; Rammurti T. Kamble; Mark R. Litzow; Alison W. Loren; David I. Marks; Eduardo Olavarria; Vivek Roy; Mitchell Sabloff; Bipin N. Savani; Matthew D. Seftel; Harry C. Schouten; Celalettin Ustun; Edmund K. Waller; Daniel J. Weisdorf

Cyclophosphamide combined with total body irradiation (Cy/TBI) or busulfan (BuCy) are the most widely used myeloablative conditioning regimens for allotransplants. Recent data regarding their comparative effectiveness are lacking. We analyzed data from the Center for International Blood and Marrow Transplant Research for 1230 subjects receiving a first hematopoietic cell transplant from a human leukocyte antigen-matched sibling or from an unrelated donor during the years 2000 to 2006 for acute myeloid leukemia (AML) in first complete remission (CR) after conditioning with Cy/TBI or oral or intravenous (IV) BuCy. Multivariate analysis showed significantly less nonrelapse mortality (relative risk [RR] = 0.58; 95% confidence interval [CI]: 0.39-0.86; P = .007), and relapse after, but not before, 1 year posttransplant (RR = 0.23; 95% CI: 0.08-0.65; P = .006), and better leukemia-free survival (RR = 0.70; 95% CI: 0.55-0.88; P = .003) and survival (RR = 0.68; 95% CI: 0.52-0.88; P = .003) in persons receiving IV, but not oral, Bu compared with TBI. In combination with Cy, IV Bu is associated with superior outcomes compared with TBI in patients with AML in first CR.


Blood | 2015

Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation.

Xiaoou Zhou; Gianpietro Dotti; Robert A. Krance; Caridad Martinez; Swati Naik; Rammurti T. Kamble; April G. Durett; Olga Dakhova; Barbara Savoldo; Antonio Di Stasi; David M. Spencer; Yu-Feng Lin; Hao Liu; Bambi Grilley; Adrian P. Gee; Cliona M. Rooney; Helen E. Heslop; Malcolm K. Brenner

To test the feasibility of a single T-cell manipulation to eliminate alloreactivity while sparing antiviral and antitumor T cells, we infused 12 haploidentical hematopoietic stem cell transplant patients with increasing numbers of alloreplete haploidentical T cells expressing the inducible caspase 9 suicide gene (iC9-T cells). We determined whether the iC9-T cells produced immune reconstitution and if any resultant graft-versus-host disease (GVHD) could be controlled by administration of a chemical inducer of dimerization (CID; AP1903/Rimiducid). All patients receiving >10(4) alloreplete iC9-T lymphocytes per kilogram achieved rapid reconstitution of immune responses toward 5 major pathogenic viruses and concomitant control of active infections. Four patients received a single AP1903 dose. CID infusion eliminated 85% to 95% of circulating CD3(+)CD19(+) T cells within 30 minutes, with no recurrence of GVHD within 90 days. In one patient, symptoms and signs of GVHD-associated cytokine release syndrome (CRS-hyperpyrexia, high levels of proinflammatory cytokines, and rash) resolved within 2 hours of AP1903 infusion. One patient with varicella zoster virus meningitis and acute GVHD had iC9-T cells present in the cerebrospinal fluid, which were reduced by >90% after CID. Notably, virus-specific T cells recovered even after AP1903 administration and continued to protect against infection. Hence, alloreplete iC9-T cells can reconstitute immunity posttransplant and administration of CID can eliminate them from both peripheral blood and the central nervous system (CNS), leading to rapid resolution of GVHD and CRS. The approach may therefore be useful for the rapid and effective treatment of toxicities associated with infusion of engineered T lymphocytes. This trial was registered at www.clinicaltrials.gov as #NCT01494103.


Bone Marrow Transplantation | 2007

Human herpesvirus-6 encephalitis following allogeneic hematopoietic stem cell transplantation

T Vu; George Carrum; George J. Hutton; Helen E. Heslop; Malcolm K. Brenner; Rammurti T. Kamble

Immunosuppressive monoclonal antibodies directed to immune system cells may reduce rejection and graft versus host disease (GvHD) after allogeneic stem cell transplantation (SCT), but can increase the risks of viral infection. Here, we report human herpes virus-6 (HHV-6) encephalitis despite antiviral prophylaxis in 5 of 43 (11.6%) patients receiving alemtuzumab supported conditioning. Encephalitis occurred at 41–103 days (median 60 days) presenting with confusion in all patients, combined with amnesia (n=3) or seizures (n=2). MRI revealed non-specific white matter changes in two and a non-enhancing medial temporal lobe lesion in three patients. Cerebrospinal fluid (CSF) PCR amplification for HHV-6 was positive in all five patients, (600–2 25 000 (median 4700) copies/ml CSF), while analysis of peripheral blood revealed 100–22 500 (median 1200) viral copies/ml plasma. CSF protein was elevated in four patients, with minimal CSF pleocytosis. Intravenous foscarnet produced neurological improvement at 8–13 (median 11) days and negative plasma PCR at 30–66 (median 50) days. Four patients had complete neurological recovery, but one patient with persistent viral DNA in the CSF succumbed to progressive encephalopathy. Given this high incidence of HHV-6 and the possibility of successful outcome with prompt treatment, a high index of suspicion of this disorder is required in recipients of monoclonal antibody supported allografts.


Transfusion | 2005

Microbial contamination of hematopoietic progenitor cell grafts—incidence, clinical outcome, and cost-effectiveness: an analysis of 735 grafts

Rammurti T. Kamble; Shubham Pant; George B. Selby; Mohamed A. Kharfan-Dabaja; Sanjay Sethi; Kristen Kratochvil; Nancy Kohrt; Howard Ozer

BACKGROUND: Screening of progenitor cell grafts (marrow, peripheral blood, and cord blood) for microbial contamination is required by the standards of AABB. Clinical sequelae from infusion of these contaminated grafts, however, is uncommon.


Journal of Clinical Oncology | 2015

Improved Outcomes After Autologous Hematopoietic Cell Transplantation for Light Chain Amyloidosis: A Center for International Blood and Marrow Transplant Research Study

Anita D'Souza; Angela Dispenzieri; Baldeep Wirk; Mei-Jie Zhang; Jiaxing Huang; Morie A. Gertz; Robert A. Kyle; Shaji Kumar; Raymond L. Comenzo; Robert Peter Gale; Hillard M. Lazarus; Bipin N. Savani; Robert F. Cornell; Brendan M. Weiss; Dan T. Vogl; Cesar O. Freytes; Emma C. Scott; Heather Landau; Jan S. Moreb; Luciano J. Costa; Muthalagu Ramanathan; Natalie S. Callander; Rammurti T. Kamble; Richard Olsson; Siddhartha Ganguly; Taiga Nishihori; Tamila L. Kindwall-Keller; William A. Wood; Tomer Mark; Parameswaran Hari

PURPOSE Autologous hematopoietic cell transplantation, or autotransplantation, is effective in light-chain amyloidosis (AL), but it is associated with a high risk of early mortality (EM). In a multicenter randomized comparison against oral chemotherapy, autotransplantation was associated with 24% EM. We analyzed trends in outcomes after autologous hematopoietic cell transplantation for AL in North America. PATIENTS AND METHODS Between 1995 and 2012, 1,536 patients with AL who underwent autotransplantation at 134 centers were identified in the Center for International Blood and Marrow Transplant Research database. EM and overall survival (OS) were analyzed in three time cohorts: 1995 to 2000 (n = 140), 2001 to 2006 (n = 596), and 2007 to 2012 (n = 800). Hematologic and renal responses and factors associated with EM, relapse and/or progression, progression-free survival and OS were analyzed in more recent subgroups from 2001 to 2006 (n = 197) and from 2007 to 2012 (n = 157). RESULTS Mortality at 30 and 100 days progressively declined over successive time periods from 11% and 20%, respectively, in 1995 to 2000 to 5% and 11%, respectively, in 2001 to 2006, and to 3% and 5%, respectively, in 2007 to 2012. Correspondingly, 5-year OS improved from 55% in 1995 to 2000 to 61% in 2001 to 2006 and to 77% in 2007 to 2012. Hematologic response to transplantation improved in the latest cohort. Renal response rate was 32%. Centers performing more than four AL transplantations per year had superior survival outcomes. In the multivariable analysis, cardiac AL was associated with high EM and inferior progression-free survival and OS. Autotransplantation in 2007 to 2012 and use of higher dosages of melphalan were associated with a lowered relapse risk. A Karnofsky score less than 80 and creatinine levels 2 mg/m(2) or greater were associated with worsened OS. CONCLUSION Post-transplantation survival in AL has improved, with a dramatic reduction in early post-transplantation mortality and excellent 5-year survival. The risk-benefit ratio for autotransplantation has changed, and randomized comparison with nontransplantation approaches is again warranted.


Journal of Clinical Investigation | 2016

Clinical responses with T lymphocytes targeting malignancy-associated κ light chains

Carlos A. Ramos; Barbara Savoldo; Vicky Torrano; Brandon Ballard; Huimin Zhang; Olga Dakhova; Enli Liu; George Carrum; Rammurti T. Kamble; Adrian P. Gee; Zhuyong Mei; Meng Fen Wu; Hao Liu; Bambi Grilley; Cliona M. Rooney; Malcolm K. Brenner; Helen E. Heslop; Gianpietro Dotti

BACKGROUND Treatment of B cell malignancies with adoptive transfer of T cells with a CD19-specific chimeric antigen receptor (CAR) shows remarkable clinical efficacy. However, long-term persistence of T cells targeting CD19, a pan-B cell marker, also depletes normal B cells and causes severe hypogammaglobulinemia. Here, we developed a strategy to target B cell malignancies more selectively by taking advantage of B cell light Ig chain restriction. We generated a CAR that is specific for the κ light chain (κ.CAR) and therefore recognizes κ-restricted cells and spares the normal B cells expressing the nontargeted λ light chain, thus potentially minimizing humoral immunity impairment. METHODS We conducted a phase 1 clinical trial and treated 16 patients with relapsed or refractory κ+ non-Hodgkin lymphoma/chronic lymphocytic leukemia (NHL/CLL) or multiple myeloma (MM) with autologous T cells genetically modified to express κ.CAR (κ.CARTs). Other treatments were discontinued in 11 of the 16 patients at least 4 weeks prior to T cell infusion. Six patients without lymphopenia received 12.5 mg/kg cyclophosphamide 4 days before κ.CART infusion (0.2 × 108 to 2 × 108 κ.CARTs/m2). No other lymphodepletion was used. RESULTS κ.CART expansion peaked 1-2 weeks after infusion, and cells remained detectable for more than 6 weeks. Of 9 patients with relapsed NHL or CLL, 2 entered complete remission after 2 and 3 infusions of κ.CARTs, and 1 had a partial response. Of 7 patients with MM, 4 had stable disease lasting 2-17 months. No toxicities attributable to κ.CARTs were observed. CONCLUSION κ.CART infusion is feasible and safe and can lead to complete clinical responses. TRIAL REGISTRATION ClinicalTrials.gov NCT00881920. FUNDING National Cancer Institute (NCI) grants 3P50CA126752 and 5P30CA125123 and Leukemia and Lymphoma Society (LLS) Specialized Centers of Research (SCOR) grant 7018.

Collaboration


Dive into the Rammurti T. Kamble's collaboration.

Top Co-Authors

Avatar

Hillard M. Lazarus

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

George Carrum

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bipin N. Savani

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David I. Marks

University Hospitals Bristol NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cesar O. Freytes

University of Texas Health Science Center at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge