Network


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

Hotspot


Dive into the research topics where James D. Levine is active.

Publication


Featured researches published by James D. Levine.


The New England Journal of Medicine | 1990

Recombinant Human Erythropoietin for Patients with AIDS Treated with Zidovudine

Margaret A. Fischl; Jeffrey E. Galpin; James D. Levine; Jerome E. Groopman; David H. Henry; Peter Kennedy; Steven A. Miles; William Robbins; Barbara Starrett; Ralph Zalusky; Robert I. Abels; Huei C. Tsai; Seth A. Rudnick

Bone marrow suppression and anemia are frequent side effects of treatment of the acquired immunodeficiency syndrome (AIDS) with zidovudine (formerly azidothymidine [AZT]). We conducted a randomized, double-blind, placebo-controlled clinical trial of recombinant human erythropoietin (100 U per kilogram of body weight thrice weekly by intravenous bolus) in 63 patients with AIDS treated with zidovudine (29 in the erythropoietin group and 34 in the placebo group). Reductions in the number of units of red cells transfused and the number of patients given transfusions per month became apparent in the second and third months of the trial. The reductions were observed in patients with endogenous erythropoietin levels less than or equal to 500 IU per liter at base line, but not in patients whose levels were greater than 500 IU per liter at the beginning of the study. Although the hematocrit and hemoglobin level were not used as the primary criteria of efficacy because the patients received transfusions when their physicians decided that they needed them, a significantly higher rate of increase in the hematocrit was observed in the patients treated with recombinant human erythropoietin whose levels of endogenous erythropoietin were less than or equal to 500 IU per liter (0.00353 points per week) than in the patients given placebo (0.00116 points per week). This effect was not seen in patients with higher levels of endogenous erythropoietin. Serious side effects did not occur more often in the group treated with erythropoietin than in the placebo group. We conclude that recombinant human erythropoietin may be useful in patients with AIDS treated with zidovudine, although the indicators for its use remain to be clarified.


Journal of Immunotherapy | 2011

PD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses to autologous dendritic cell/myeloma fusion vaccine.

Jacalyn Rosenblatt; Brett Glotzbecker; Heidi Mills; Baldev Vasir; Dimitrios Tzachanis; James D. Levine; Robin Joyce; Kerry Wellenstein; Whitney Keefe; Michael Schickler; Rinat Rotem-Yehudar; Donald Kufe; David Avigan

We have developed a cancer vaccine in which autologous tumor is fused with dendritic cells (DCs) resulting in the presentation of tumor antigens in the context of DC-mediated costimulation. In clinical trials, immunologic responses have been observed, however responses may be muted by inhibitory pathways. The PD1/PDL1 pathway is an important element contributing to tumor-mediated immune suppression. In this study, we demonstrate that myeloma cells and DC/tumor fusions strongly express PD-L1. Compared with a control population of normal volunteers, increased PD-1 expression was observed on T cells isolated from patients with myeloma. It is interesting to note that after autologous transplantation, T-cell expression of PD-1 returned to levels seen in normal controls. We examined the effect of PD-1 blockade on T-cell response to DC/tumor fusions ex vivo. Presence of CT-011, an anti-PD1 antibody, promoted the vaccine-induced T-cell polarization towards an activated phenotype expressing Th1 compared with Th2 cytokines. A concomitant decrease in regulatory T cells and enhanced killing in a cytotoxicity assay was observed. In summary, we demonstrate that PD-1 expression is increased in T cells of patients with active myeloma, and that CT-011 enhances activated T-cell responses after DC/tumor fusion stimulation.


Blood | 2011

Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma

Jacalyn Rosenblatt; Baldev Vasir; Lynne Uhl; Simona Blotta; Claire MacNamara; Poorvi Somaiya; Zekui Wu; Robin Joyce; James D. Levine; Dilani Dombagoda; Yan Emily Yuan; Karen Francoeur; Donna Fitzgerald; Paul G. Richardson; Edie Weller; Kenneth C. Anderson; Donald Kufe; Nikhil C. Munshi; David Avigan

We have developed a tumor vaccine in which patient-derived myeloma cells are chemically fused with autologous dendritic cells (DCs) such that a broad spectrum of myeloma-associated antigens are presented in the context of DC-mediated costimulation. We have completed a phase 1 study in which patients with multiple myeloma underwent serial vaccination with the DC/multiple myeloma fusions in conjunction with granulocyte-macrophage colony-stimulating factor. DCs were generated from adherent mononuclear cells cultured with granulocyte-macrophage colony-stimulating factor, interleukin-4, and tumor necrosis factor-α and fused with myeloma cells obtained from marrow aspirates. Vaccine generation was successful in 17 of 18 patients. Successive cohorts were treated with 1 × 10(6), 2 × 10(6), and 4 × 10(6) fusion cells, respectively, with 10 patients treated at the highest dose level. Vaccination was well tolerated, without evidence of dose-limiting toxicity. Vaccination resulted in the expansion of circulating CD4 and CD8 lymphocytes reactive with autologous myeloma cells in 11 of 15 evaluable patients. Humoral responses were documented by SEREX (Serologic Analysis of Recombinant cDNA Expression Libraries) analysis. A majority of patients with advanced disease demonstrated disease stabilization, with 3 patients showing ongoing stable disease at 12, 25, and 41 months, respectively. Vaccination with DC/multiple myeloma fusions was feasible and well tolerated and resulted in antitumor immune responses and disease stabilization in a majority of patients.


Clinical Cancer Research | 2013

Vaccination with Dendritic Cell/Tumor Fusions following Autologous Stem Cell Transplant Induces Immunologic and Clinical Responses in Multiple Myeloma Patients

Jacalyn Rosenblatt; Irit Avivi; Baldev Vasir; Lynne Uhl; Nikhil C. Munshi; Tami Katz; Bimalangshu R. Dey; Poorvi Somaiya; Heidi Mills; Federico Campigotto; Edie Weller; Robin Joyce; James D. Levine; Dimitrios Tzachanis; Paul G. Richardson; Jacob P. Laubach; Noopur Raje; Vassiliki A. Boussiotis; Yan Emily Yuan; Lina Bisharat; Viki Held; Jacob M. Rowe; Kenneth C. Anderson; Donald Kufe; David Avigan

Purpose: A multiple myeloma vaccine has been developed whereby patient-derived tumor cells are fused with autologous dendritic cells, creating a hybridoma that stimulates a broad antitumor response. We report on the results of a phase II trial in which patients underwent vaccination following autologous stem cell transplantation (ASCT) to target minimal residual disease. Experimental Design: Twenty-four patients received serial vaccinations with dendritic cell/myeloma fusion cells following posttransplant hematopoietic recovery. A second cohort of 12 patients received a pretransplant vaccine followed by posttransplant vaccinations. Dendritic cells generated from adherent mononuclear cells cultured with granulocyte macrophage colony-stimulating factor, interleukin-4, and TNF-α were fused with autologous bone marrow–derived myeloma fusion cells using polyethylene glycol. Fusion cells were quantified by determining the percentage of cells that coexpress dendritic cell and myeloma fusion antigens. Results: The posttransplant period was associated with reduction in general measures of cellular immunity; however, an increase in CD4 and CD8+ myeloma-specific T cells was observed after ASCT that was significantly expanded following posttransplant vaccination. Seventy-eight percent of patients achieved a best response of complete response (CR)+very good partial response (VGPR) and 47% achieved a CR/near CR (nCR). Remarkably, 24% of patients who achieved a partial response following transplant were converted to CR/nCR after vaccination and at more than 3 months posttransplant, consistent with a vaccine-mediated effect on residual disease. Conclusions: The posttransplant period for patients with multiple myeloma provides a unique platform for cellular immunotherapy in which vaccination with dendritic cell/myeloma fusion fusions resulted in the marked expansion of myeloma-specific T cells and cytoreduction of minimal residual disease. Clin Cancer Res; 19(13); 3640–8. ©2013 AACR.


Clinical Cancer Research | 2008

Phase I and Pharmacokinetic Study of Bortezomib in Combination with Idarubicin and Cytarabine in Patients with Acute Myelogenous Leukemia

Eyal C. Attar; Daniel J. DeAngelo; Jeffrey G. Supko; Ferdinando D'Amato; David Zahrieh; Andres Sirulnik; Martha Wadleigh; Karen K. Ballen; Steve McAfee; Kenneth B. Miller; James D. Levine; Ilene Galinsky; Elizabeth Trehu; David P. Schenkein; Donna Neuberg; Richard Stone; Philip C. Amrein

Purpose: Proteasome inhibition results in cytotoxicity to the leukemia stem cell in vitro. We conducted this phase I study to determine if the proteasome inhibitor bortezomib could be safely added to induction chemotherapy in patients with acute myelogenous leukemia (AML). Experimental Design: Bortezomib was given on days 1, 4, 8, and 11 at doses of 0.7, 1.0, 1.3, or 1.5 mg/m2 with idarubicin 12 mg/m2 on days 1 to 3 and cytarabine 100 mg/m2/day on days 1 to 7. Results: A total of 31 patients were enrolled. The median age was 62 years, and 16 patients were male. Nine patients had relapsed AML (ages, 18-59 years, n = 4 and ≥60 years, n = 5). There were 22 patients of ≥60 years with previously untreated AML (eight with prior myelodysplasia/myeloproliferative disorder or cytotoxic therapy). All doses of bortezomib, up to and including 1.5 mg/m2, were tolerable. Nonhematologic grade 3 or greater toxicities included 12 hypoxia (38%; 11 were grade 3), 4 hyperbilirubinemia (13%), and 6 elevated aspartate aminotransferase (19%). Overall, 19 patients (61%) achieved complete remission (CR) and three had CR with incomplete platelet recovery. Pharmacokinetic studies revealed that the total body clearance of bortezomib decreased significantly (P < 0.01, N = 26) between the first (mean ± SD, 41.9 ± 17.1 L/h/m2) and third (18.4 ± 7.0 L/h/m2) doses. Increased bone marrow expression of CD74 was associated with CR. Conclusions: The combination of bortezomib, idarubicin, and cytarabine showed a good safety profile. The recommended dose of bortezomib for phase II studies with idarubicin and cytarabine is 1.5 mg/m2.


Cancer Immunology, Immunotherapy | 2013

Lenalidomide enhances anti-myeloma cellular immunity

Katarina Luptakova; Jacalyn Rosenblatt; Brett Glotzbecker; Heidi Mills; Dina Stroopinsky; Turner Kufe; Baldev Vasir; Jon Arnason; Dimitri Tzachanis; Jeffrey I. Zwicker; Robin Joyce; James D. Levine; Kenneth C. Anderson; Donald Kufe; David Avigan

Lenalidomide is an effective therapeutic agent for multiple myeloma that exhibits immunomodulatory properties including the activation of T and NK cells. The use of lenalidomide to reverse tumor-mediated immune suppression and amplify myeloma-specific immunity is currently being explored. In the present study, we examined the effect of lenalidomide on T-cell activation and its ability to amplify responses to a dendritic cell-based myeloma vaccine. We demonstrate that exposure to lenalidomide in the context of T-cell expansion with direct ligation of CD3/CD28 complex results in polarization toward a Th1 phenotype characterized by increased IFN-γ, but not IL-10 expression. In vitro exposure to lenalidomide resulted in decreased levels of regulatory T cells and a decrease in T-cell expression of the inhibitory marker, PD-1. Lenalidomide also enhanced T-cell proliferative responses to allogeneic DCs. Most significantly, lenalidomide treatment potentiated responses to the dendritic cell/myeloma fusion vaccine, which were characterized by increased production of inflammatory cytokines and increased cytotoxic lymphocyte-mediated lysis of autologous myeloma targets. These findings indicate that lenalidomide enhances the immunologic milieu in patients with myeloma by promoting T-cell proliferation and suppressing inhibitory factors, and thereby augmenting responses to a myeloma-specific tumor vaccine.


Science Translational Medicine | 2016

Individualized vaccination of AML patients in remission is associated with induction of antileukemia immunity and prolonged remissions

Jacalyn Rosenblatt; Richard Stone; Lynne Uhl; Donna Neuberg; Robin Joyce; James D. Levine; Jon Arnason; Malgorzata McMasters; Katarina Luptakova; Salvia Jain; Jeffrey I. Zwicker; Ayad Hamdan; Vassiliki A. Boussiotis; David P. Steensma; Daniel J. DeAngelo; Ilene Galinsky; Poorvi Somaiya Dutt; Emma Logan; Mary Paty Bryant; Dina Stroopinsky; Lillian Werner; Kristen Palmer; Max Coll; Abigail Washington; Leandra Cole; Donald Kufe; David Avigan

A personalized DC/AML fusion cell vaccine promotes the expansion of leukemia-specific T cells and prolonged remission in patients. Immune cells join leukemia then beat it Acute myeloid leukemia (AML) is an aggressive hematologic cancer. The only curative treatment available for this disease is hematopoietic stem cell transplantation, which can result in donor immune cells helping to eradicate the cancer. Unfortunately, this procedure is not always effective and is itself associated with numerous complications and risk of death. Rosenblatt et al. have identified a potentially better way to stimulate an immune response against AML by fusing patients’ own leukemia cells with dendritic cells. The resulting fusion cells were very effective at presenting tumor antigens to T cells, resulting in a strong antitumor T cell response and prolonged survival in human patients. We developed a personalized cancer vaccine in which patient-derived acute myeloid leukemia (AML) cells are fused with autologous dendritic cells, generating a hybridoma that potently stimulates broad antitumor responses. We report results obtained from the first 17 AML patients, who achieved remission after chemotherapy and were then serially vaccinated to target minimal residual disease and prevent relapse. Vaccination was well tolerated and induced inflammatory responses at the site of administration, characterized by the dense infiltration of T cells. Vaccination was also associated with a marked rise in circulating T cells recognizing whole AML cells and leukemia-specific antigens that persisted for more than 6 months. Twelve of 17 vaccinated patients (71%; 90% confidence interval, 52 to 89%) remain alive without recurrence at a median follow-up of 57 months. The results demonstrate that personalized vaccination of AML patients in remission induces the expansion of leukemia-specific T cells and may be protective against disease relapse.


Bone Marrow Transplantation | 2000

Immune reconstitution following high-dose chemotherapy with stem cell rescue in patients with advanced breast cancer

David Avigan; Zekui Wu; Robin Joyce; Anthony Elias; Paul G. Richardson; David F. McDermott; James D. Levine; L Kennedy; N. Giallombardo; Hurley D; Jianlin Gong; Donald Kufe

The present study examines the nature of humoral and cellular immune reconstitution in 28 patients with advanced breast cancer following high-dose chemotherapy with stem cell rescue. Patients underwent testing of T, B, NK and dendritic cell function at serial time points until 1 year post transplant or until the time of disease progression. Abnormalities in T cell phenotype and function were observed following high-dose chemotherapy that persisted for at least 6–12 months. The vast majority of patients experienced an inversion of the CD4/CD8 ratio and demonstrated an anergic response to candida antigen. Mean T cell proliferation in response to PHA and to co-culture with allogeneic monocytes was significantly compromised. In contrast, mean IgG and IgA levels were normal 6 months post transplant and NK cell yields and function were transiently elevated following high-dose chemotherapy. Dendritic cells generated from peripheral blood progenitors displayed a characteristic phenotype and were potent inducers of allogeneic T cell proliferation in the post-transplant period. The study demonstrates that patients undergoing autologous transplantation for breast cancer experience a prolonged period of T cell dysfunction. In contrast, B, NK, and DC recover more rapidly. These findings carry significant implications for the design of post-transplant immunotherapy. Bone Marrow Transplantation (2000) 26, 169–176.


Cancer Research | 2013

MUC1 Is a Potential Target for the Treatment of Acute Myeloid Leukemia Stem Cells

Dina Stroopinsky; Jacalyn Rosenblatt; Keisuke Ito; Heidi Mills; Li Yin; Hasan Rajabi; Baldev Vasir; Turner Kufe; Katarina Luptakova; Jon Arnason; Caterina Nardella; James D. Levine; Robin Joyce; Ilene Galinsky; Yoram Reiter; Richard Stone; Pier Paolo Pandolfi; Donald Kufe; David Avigan

Acute myeloid leukemia (AML) is a malignancy of stem cells with an unlimited capacity for self-renewal. MUC1 is a secreted, oncogenic mucin that is expressed aberrantly in AML blasts, but its potential uses to target AML stem cells have not been explored. Here, we report that MUC1 is highly expressed on AML CD34(+)/lineage(-)/CD38(-) cells as compared with their normal stem cell counterparts. MUC1 expression was not restricted to AML CD34(+) populations as similar results were obtained with leukemic cells from patients with CD34(-) disease. Engraftment of AML stem cell populations that highly express MUC1 (MUC1(high)) led to development of leukemia in NOD-SCID IL2Rgamma(null) (NSG) immunodeficient mice. In contrast, MUC1(low) cell populations established normal hematopoiesis in the NSG model. Functional blockade of the oncogenic MUC1-C subunit with the peptide inhibitor GO-203 depleted established AML in vivo, but did not affect engraftment of normal hematopoietic cells. Our results establish that MUC1 is highly expressed in AML stem cells and they define the MUC1-C subunit as a valid target for their therapeutic eradication.


American Journal of Clinical Oncology | 1991

GM-CSF as an alternative to dose modification of the combination zidovudine and interferon-alpha in the treatment of AIDS-associated Kaposi's sarcoma.

David T. Scadden; Harriet A. Bering; James D. Levine; Jocelyn Bresnahan; Lydia M. Evans; Carol Epstein; Jerome E. Groopman

Combined zidovudine (ZDV) and interferon-α (IFN) is an appealing therapy for AIDS-associated Kaposis sarcoma because of the antirctroviral as well as antitumor potential of this combination. Overlapping myclotoxicity of these agents, however, frequently complicates their clinical use. This phase I/II study was undertaken to test the safety and efficacy of granulocyte-macrophage colony stimulating factor (GM-CSF) in those patients who became neutropenic while receiving ZDV (1,200 mg/day) and IFN (9 MU/day). Despite a “high-risk” population of patients, the tumor response rate among evaluable patients was 50% (33% overall). Sixty-four percent of patients required GM-CSF and all patients receiving GM-CSF had a prompt improvement in their absolute neutrophil count (ANC). The use of GM-CSF was associated with an improved end of study ANC (p; < 0.05). but was not associated with tumor response, CD4 count improvement, or improved change in hemoglobin concentration. GM-CSF/ZDV/IFN was not associated with increased toxicity over ZDV/IFN; however, two unusual events occurred in the GM-CSF/ZDV/IFN group: erythema multiforme and glucose intolerance. Dose-limiting thrombocytopenia and anemia were seen in two patients and anemia in one patient on GM-CSF/ZDV/IFN. No consistent alterations in serum HIV p24 antigenemia were noted in either group. The use of GM-CSF mitigated the neutropenia of combined ZDV and IFN. Further study evaluating the utility of this hematopoietic growth factor in combination therapies for AIDS patients is warranted.

Collaboration


Dive into the James D. Levine's collaboration.

Top Co-Authors

Avatar

David Avigan

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Robin Joyce

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jacalyn Rosenblatt

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Donald Kufe

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Baldev Vasir

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jon Arnason

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dina Stroopinsky

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heidi Mills

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge