Network


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

Hotspot


Dive into the research topics where Richard F. Ambinder is active.

Publication


Featured researches published by Richard F. Ambinder.


Blood | 2015

Risk-stratified outcomes of nonmyeloablative HLA-haploidentical BMT with high-dose posttransplantation cyclophosphamide

Shannon R. McCurdy; Jennifer A. Kanakry; Margaret M. Showel; Hua Ling Tsai; Javier Bolaños-Meade; Gary L. Rosner; Christopher G. Kanakry; Karlo Perica; Heather J. Symons; Robert A. Brodsky; Douglas E. Gladstone; Carol Ann Huff; Keith W. Pratz; Gabrielle T. Prince; Amy E. DeZern; Ivana Gojo; William Matsui; Ivan Borrello; Michael A. McDevitt; Lode J. Swinnen; B. Douglas Smith; Mark Levis; Richard F. Ambinder; Leo Luznik; Richard J. Jones; Ephraim J. Fuchs; Yvette L. Kasamon

Related HLA-haploidentical blood or marrow transplantation (BMT) with high-dose posttransplantation cyclophosphamide (PTCy) is being increasingly used because of its acceptable safety profile. To better define outcomes of nonmyeloablative (NMA) HLA-haploidentical BMT with PTCy, 372 consecutive adult hematologic malignancy patients who underwent this procedure were retrospectively studied. Risk-stratified outcomes were evaluated using the refined Disease Risk Index (DRI), developed to stratify disease risk across histologies and allogeneic BMT regimens. Patients received uniform conditioning, T-cell-replete allografting, then PTCy, mycophenolate mofetil, and tacrolimus. Six-month probabilities of nonrelapse mortality and severe acute graft-versus-host disease were 8% and 4%. With 4.1-year median follow-up, 3-year probabilities of relapse, progression-free survival (PFS), and overall survival (OS) were 46%, 40%, and 50%, respectively. By refined DRI group, low (n = 71), intermediate (n = 241), and high/very high (n = 60) risk groups had 3-year PFS estimates of 65%, 37%, and 22% (P < .0001), with corresponding 3-year OS estimates of 71%, 48%, and 35% (P = .0001). On multivariable analyses, the DRI was statistically significantly associated with relapse, PFS, and OS (each P < .001). This analysis demonstrates that the DRI effectively risk stratifies recipients of NMA HLA-haploidentical BMT with PTCy and also suggests that this transplantation platform yields similar survivals to those seen with HLA-matched BMT.


The Journal of Infectious Diseases | 1999

Heterogeneity of Viral IL-6 Expression in HHV-8—Associated Diseases

Jennifer S. Cannon; John Nicholas; Jan M. Orenstein; Risa B. Mann; Paul G. Murray; Philip J. Browning; Joseph A. DiGiuseppe; Ethel Cesarman; Gary S. Hayward; Richard F. Ambinder

In order to characterize the expression of the viral interleukin-6 (vIL-6) homologue in various human herpesvirus 8 (HHV-8)-associated diseases, in situ hybridization and immunohistochemistry were applied to formalin-fixed specimens. These assays showed consistent expression of vIL-6 in primary effusion lymphomas and in a case of human immunodeficiency virus (HIV)-associated lymphadenopathy with a Castlemans disease-like appearance. In contrast, Kaposis sarcoma specimens showed marked differences among specimens. In a consecutive series of specimens from the Johns Hopkins archives, vIL-6 expression was demonstrated in one of 13 cases. However, among 7 specimens selected from the AIDS Malignancy Bank because of their high levels of the T1.1 lytic transcript and virion production, vIL-6 expression was consistently demonstrated in infiltrating mononuclear cells and occasional spindle-shaped cells. Thus vIL-6 expression in clinical specimens correlates with other measures of the lytic viral cycle. Both assays generally give congruent results and are consistent with the possibility that vIL-6 expression plays a role in the pathogenesis of a variety of HHV-8-associated diseases.


Blood | 2014

Single-agent GVHD prophylaxis with posttransplantation cyclophosphamide after myeloablative, HLA-matched BMT for AML, ALL, and MDS

Christopher G. Kanakry; Hua Ling Tsai; Javier Bolaños-Meade; B. Douglas Smith; Ivana Gojo; Jennifer A. Kanakry; Yvette L. Kasamon; Douglas E. Gladstone; William Matsui; Ivan Borrello; Carol Ann Huff; Lode J. Swinnen; Jonathan D. Powell; Keith W. Pratz; Amy E. DeZern; Margaret M. Showel; Michael A. McDevitt; Robert A. Brodsky; Mark Levis; Richard F. Ambinder; Ephraim J. Fuchs; Gary L. Rosner; Richard J. Jones; Leo Luznik

High-dose, posttransplantation cyclophosphamide (PTCy) reduces severe graft-versus-host disease (GVHD) after allogeneic blood or marrow transplantation (alloBMT), but the impact of PTCy on long-term, disease-specific outcomes is unclear. We conducted a retrospective study of 209 consecutive adult patients transplanted for acute myeloid leukemia (AML, n = 138), myelodysplastic syndrome (n = 28), or acute lymphoblastic leukemia (ALL, n = 43) using PTCy as sole GVHD prophylaxis after myeloablative conditioning and HLA-matched-related or -unrelated T-cell-replete allografting. At alloBMT, 30% of patients were not in morphologic complete remission. The cumulative incidences of grades II to IV and III to IV acute GVHD at 100 days and chronic GVHD at 2 years were 45%, 11%, and 13%, respectively. Forty-three percent of patients did not require immunosuppression for any reason beyond PTCy. At 3 years, relapse cumulative incidence was 36%, disease-free survival was 46%, survival free of disease and chronic GVHD was 39%, and overall survival was 58%. Lack of remission at alloBMT, adverse cytogenetics, and low allograft nucleated cell dose were associated with inferior survival for AML patients. Minimal residual disease but not t(9;22) was associated with inferior outcomes for ALL patients. The ability to limit posttransplantation immunosuppression makes PTCy a promising transplantation platform for the integration of postgrafting strategies to prevent relapse.


Lancet Oncology | 2011

Use of antineoplastic agents in patients with cancer who have HIV/AIDS

Michelle A. Rudek; Charles Flexner; Richard F. Ambinder

Highly active antiretroviral therapy (HAART) has substantially reduced morbidity and mortality of AIDS-related complications in patients with HIV; however, the prevalence of AIDS-defining cancers and non-AIDS-defining cancers has increased. In this Review we discuss the management of HAART pharmacotherapy in relation to cytotoxic chemotherapy or targeted antineoplastic agents. We will review potential pharmacological interactions between antiretroviral and antineoplastic therapies and consider how to combine antiretroviral and antineoplastic agents in patients with HIV who are receiving HAART therapy.


Blood | 2011

Bortezomib induction of C/EBPβ mediates Epstein-Barr virus lytic activation in Burkitt lymphoma

Courtney M Shirley; Jianmeng Chen; Meir Shamay; Huili Li; Cynthia A. Zahnow; S. Diane Hayward; Richard F. Ambinder

Epstein-Barr virus (EBV) is associated with a variety of lymphoid malignancies. Bortezomib activates EBV lytic gene expression. Bortezomib, a proteasome inhibitor, leads to increased levels of CCAAT/enhancer-binding proteinβ (C/EBPβ) in a variety of tumor cell lines. C/EBPβ activates the promoter of the EBV lytic switch gene ZTA. Bortezomib treatment leads to increased binding of C/EBP to previously recognized binding sites in the ZTA promoter. Knockdown of C/EBPβ inhibits bortezomib activation of EBV lytic gene expression. Bortezomib also induces the unfolded protein response (UPR), as evidenced by increases in ATF4, CHOP10, and XBP1s and cleavage of ATF6. Thapsigargin, an inducer of the UPR that does not interfere with proteasome function, also induces EBV lytic gene expression. The effects of thapsigargin on EBV lytic gene expression are also inhibited by C/EBPβ knock-down. Therefore, C/EBPβ mediates the activation of EBV lytic gene expression associated with bortezomib and another UPR inducer.


Science Translational Medicine | 2015

Adoptive transfer of activated marrow-infiltrating lymphocytes induces measurable antitumor immunity in the bone marrow in multiple myeloma

Kimberly A. Noonan; Carol Ann Huff; Janice M. Davis; M. Victor Lemas; Susan Fiorino; Jeffrey Bitzan; Anna Ferguson; Amy Emerling; Leo Luznik; William Matsui; Jonathan D. Powell; Ephraim J. Fuchs; Gary L. Rosner; Caroline Epstein; Lakshmi Rudraraju; Richard F. Ambinder; Richard J. Jones; Drew M. Pardoll; Ivan Borrello

Marrow-infiltrating lymphocytes (MILs) may be used as adoptive cell therapy for multiple myeloma. ACT against multiple myeloma Adoptive T cell therapy (ACT) has had some success in treating certain types of cancer; however, widespread use is limited in part by the lack of tumor-specific targets. Tumor-infiltrating T cells may overcome this limitation for solid tumors. Noonan et al. now show in a phase 1 clinical trial that bone marrow can be a source of ACT for hematologic malignancies such as multiple myeloma. Marrow-infiltrating lymphocytes (MILs) demonstrated myeloma-specific immunity in the bone marrow up to 1 year after ACT, and a significant increase was observed in progression-free survival. If these results can be replicated in larger studies, MILs may represent a source for ACT for hematologic malignancies and bone marrow–infiltrating solid tumors. Successful adoptive T cell therapy (ACT) requires the ability to activate tumor-specific T cells with the ability to traffic to the tumor site and effectively kill their target as well as persist over time. We hypothesized that ACT using marrow-infiltrating lymphocytes (MILs) in multiple myeloma (MM) could impart greater antitumor immunity in that they were obtained from the tumor microenvironment. We describe the results from the first clinical trial using MILs in MM. Twenty-five patients with either newly diagnosed or relapsed disease had their MILs harvested, activated and expanded, and subsequently infused on the third day after myeloablative therapy. Cells were obtained and adequately expanded in all patients with anti-CD3/CD28 beads plus interleukin-2, and a median of 9.5 × 108 MILs were infused. Factors indicative of response to MIL ACT included (i) the presence of measurable myeloma-specific activity of the ex vivo expanded product, (ii) low endogenous bone marrow T cell interferon-γ production at baseline, (iii) a CD8+ central memory phenotype at baseline, and (iv) the generation and persistence of myeloma-specific immunity in the bone marrow at 1 year after ACT. Achieving at least a 90% reduction in disease burden significantly increased the progression-free survival (25.1 months versus 11.8 months; P = 0.01). This study demonstrates the feasibility and efficacy of MILs as a form of ACT with applicability across many hematologic malignancies and possibly solid tumors infiltrating the bone marrow.


Haematologica | 2017

Comparable composite endpoints after HLA-matched and HLA-haploidentical transplantation with post-transplantation cyclophosphamide

Shannon R. McCurdy; Yvette L. Kasamon; Christopher G. Kanakry; Javier Bolaños-Meade; Hua Ling Tsai; Margaret M. Showel; Jennifer A. Kanakry; Heather J. Symons; Ivana Gojo; B. Douglas Smith; Maria P. Bettinotti; William Matsui; Amy E. DeZern; Carol Ann Huff; Ivan Borrello; Keith W. Pratz; Douglas E. Gladstone; Lode J. Swinnen; Robert A. Brodsky; Mark Levis; Richard F. Ambinder; Ephraim J. Fuchs; Gary L. Rosner; Richard J. Jones; Leo Luznik

Composite endpoints that not only encompass mortality and relapse, but other critical post-transplant events such as graft-versus-host disease, are being increasingly utilized to quantify survival without significant morbidity after allogeneic blood or marrow transplantation. High-dose, post-transplantation cyclophosphamide reduces severe graft-versus-host disease with allogeneic marrow transplantation, making composite endpoints after this management particularly interesting. We retrospectively analyzed 684 adults with hematologic malignancies who received T-cell-replete bone marrow grafts and cyclophosphamide after myeloablative HLA-matched related (n=192) or unrelated (n=120), or non-myeloablative HLA-haploidentical (n=372) donor transplantation. The median follow up was 4 (range, 0.02–11.4) years. Graft-versus-host disease-free, relapse-free survival was defined as the time after transplantation without grade III–IV acute graft-versus-host disease, chronic graft-versus-host disease requiring systemic treatment, relapse, or death. Chronic graft-versus-host disease-free, relapse-free survival was defined as the time after transplantation without moderate or severe chronic graft-versus-host disease, relapse, or death. One-year graft-versus-host disease-free, relapse-free survival and chronic graft-versus-host disease-free, relapse-free survival estimates were, respectively, 47% (95% CI: 41–55%) and 53% (95% CI: 46–61%) after myeloablative HLA-matched related, 42% (95% CI: 34–52%) and 52% (95% CI: 44–62%) after myeloablative HLA-matched unrelated, and 45% (95% CI: 40–50%) and 50% (95% CI: 45–55%) after non-myeloablative HLA-haploidentical donor transplantation. In multivariable models, there were no differences in graft-versus-host disease-free, or chronic graft-versus-host disease-free, relapse-free survival after either myeloablative HLA-matched unrelated or non-myeloablative HLA-haploidentical, compared with myeloablative HLA-matched related donor transplantation. Although limited by inclusion of dissimilar cohorts, we found that post-transplantation cyclophosphamide-based platforms yield comparable composite endpoints across conditioning intensity, donor type, and HLA match.


Archive | 2007

Clinical and pathological aspects of EBV and KSHV infection

Richard F. Ambinder; Ethel Cesarman

The human γ-herpesviruses Epstein–Barr virus (EBV) and Kaposi’s sarcoma herpesvirus (KSHV) establish latency in cellular reservoirs that are maintained for the life of the infected individual. Intermittent reactivation leads to infection of new cells within the host and secretion of virions in saliva. Primary infections are usually asymptomatic. However, in immunocompromised patients and in other special but poorly understood circumstances, tumors and other virus-associated diseases may manifest. Both human γ-herpesviruses were first identified in tumors and primary effusion lymphomas are typically dually infected (Cesarman et al., ; Chang et al., ; Epstein, 2001). For all their similarities, however, there are also striking differences between the viruses. EBV is nearly ubiquitous whereas KSHV is restricted to particular populations. EBV is most commonly associated with B-, T- and NK-cell tumors and epithelial tumors, whereas KSHV is associated with endothelial and B-cell tumors. In this chapter, aspects of virus–disease associations and therapies will be explored.


Biology of Blood and Marrow Transplantation | 2018

Grade II Acute Graft-versus-Host Disease and Higher Nucleated Cell Graft Dose Improve Progression-Free Survival after HLA-Haploidentical Transplant with Post-Transplant Cyclophosphamide

Shannon R. McCurdy; Christopher G. Kanakry; Hua Ling Tsai; Yvette L. Kasamon; Margaret M. Showel; Javier Bolaños-Meade; Carol Ann Huff; Ivan Borrello; William Matsui; Robert A. Brodsky; Richard F. Ambinder; Maria P. Bettinotti; Ephraim J. Fuchs; Gary L. Rosner; Richard J. Jones; Leo Luznik

Compared with standard graft-versus-host disease (GVHD) prophylaxis platforms, post-transplantation cyclophosphamide (PTCy) after T cell-replete HLA-haploidentical (haplo) bone marrow transplantation (BMT) reduces the risk of grades III to IV acute (a) and chronic (c) GVHD, but maintains similar rates of grade II aGVHD. Given that mild GVHD has been associated with reduced treatment failure in HLA-matched BMT, we evaluated the risk factors for and effects of GVHD on survival in 340 adults with hematologic malignancies who engrafted after nonmyeloablative haplo-BMT with PTCy, mycophenolate mofetil, and tacrolimus. The cumulative incidence at 100 days of grade II and grades III to IV aGVHD were 30% (95% confidence interval [CI], 25% to 35%) and 2% (95% CI, 1% to 4%), respectively. The 1-year cumulative incidence of cGVHD was 10% (95% CI, 7% to 13%). In landmark analyses at 100 days, the 4-year probabilities of overall survival (OS) and progression-free survival (PFS) were, 48% (95% CI, 41% to 56%) and 39% (95% CI, 32% to 47%) for patients without grades II to IV aGVHD, compared with 63% (95% CI, 53% to 73%) and 59% (95% CI, 50% to 71%) for patients with grade II aGVHD (P = .05 and P = .009). In multivariable modeling, when compared with patients who never experienced GVHD, the hazard ratio (HR) for OS and PFS in patients with grade II aGVHD was .78 (95% CI, .54 to 1.13; P = .19) and .69 (95% CI, .48 to .98; P = .04). Higher nucleated cell graft dose was also associated with improved OS (HR, .88; 95% CI, .78 to 1.00; P = .05) and PFS (HR, .89; 95% CI, .79 to 1.0; P = .05) and decreased risk of grades III to IV aGVHD (subdistribution HR, .66; 95% CI, .46 to .96; P = .03). PTCy reduces grades III to IV aGVHD and cGVHD, but retains similar incidence of grade II aGVHD, the development of which improves PFS. Higher nucleated cell graft dose goals may also improve survival after nonmyeloablative haplo-BMT with PTCy.


Biology of Blood and Marrow Transplantation | 2016

Functionally Active HIV-Specific T Cells that Target Gag and Nef Can Be Expanded from Virus-Naïve Donors and Target a Range of Viral Epitopes: Implications for a Cure Strategy after Allogeneic Hematopoietic Stem Cell Transplantation

Shabnum Patel; Sharon Lam; Conrad Russell Y. Cruz; Kaylor Wright; Christina Cochran; Richard F. Ambinder; Catherine M. Bollard

Allogeneic hematopoietic stem cell transplantation (HSCT) can potentially cure human immunodeficiency virus (HIV) by eliminating infected recipient cells, particularly in the context of technologies that may confer HIV resistance to these stem cells. But, to date, the Berlin patient remains the only case of HIV cure despite multiple attempts to eradicate infection with HSCT. One approach to improve this is to administer virus-specific T cells, a strategy that has proven success in preventing other infections after transplantation. Although we have reported that broadly HIV-specific T cells can be expanded from HIV+ patients, allogeneic transplantations only contain virus-naïve T cells. Modifying this approach for the allogeneic setting requires a robust, reproducible platform that can expand HIV-specific cells from the naïve pool. Hence, we hypothesized that HIV-specific T cells could be primed ex vivo from seronegative individuals to effectively target HIV. Here, we show that ex vivo-primed and expanded HIV-specific T cells released IFNγ in response to HIV antigens and that these cells have enhanced ability to suppress replication in vitro. This is the first demonstration of ex vivo priming and expansion of functional, multi-HIV antigen-specific T cells from HIV-negative donors, which has implications for use of allogeneic HSCT as a functional HIV cure.

Collaboration


Dive into the Richard F. Ambinder's collaboration.

Top Co-Authors

Avatar

Leo Luznik

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ivan Borrello

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Robert A. Brodsky

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carol Ann Huff

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge