Network


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

Hotspot


Dive into the research topics where Thea M. Friedman is active.

Publication


Featured researches published by Thea M. Friedman.


Blood | 2012

Induction of acute GVHD by sex-mismatched H-Y antigens in the absence of functional radiosensitive host hematopoietic-derived antigen-presenting cells.

Tomomi Toubai; Isao Tawara; Yaping Sun; Chen Liu; Evelyn Nieves; Rebecca Evers; Thea M. Friedman; Robert Korngold; Pavan Reddy

It is currently thought that acute GVHD cannot be elicited in the absence of Ag presentation by radiosensitive host hematopoietic-derived APCs after allogeneic BM transplantation. Because clinical data suggest that sex-mismatched H-Y Ags may be important minor histocompatibility Ags for GVH responses, we directly tested their relevance and ability to initiate GVHD when presented by either the hematopoietic- (host or donor) or the nonhematopoietic-derived APCs. H-Y minor Ag incompatibility elicited both CD4(+) and CD8(+) T-cell driven GVHD lethality. Studies with various well-established BM chimera recipients, in contrast to the current views, have reported that in the absence of functional radiosensitive host hematopoietic-derived APCs, H-Y Ag presentation by either the donor hematopoietic-derived or the host nonhematopoietic-derived APCs is sufficient for inducing GVHD. Our data further suggest that infusion of sufficient numbers of alloreactive donor T cells will induce GVHD in the absence of radiosensitive host hematopoietic-derived APCs.


Journal of Clinical Investigation | 2003

Importance of minor histocompatibility antigen expression by nonhematopoietic tissues in a CD4+ T cell–mediated graft-versus-host disease model

Stephen C. Jones; George F. Murphy; Thea M. Friedman; Robert Korngold

Minor histocompatibility antigens with expression restricted to the recipient hematopoietic compartment represent prospective immunological targets for graft-versus-leukemia therapy. It remains unclear, however, whether donor T cell recognition of these hematopoietically derived minor histocompatibility antigens will induce significant graft-versus-host disease (GVHD). Using established bone marrow irradiation chimeras across the multiple minor histocompatibility antigen-disparate, C57BL/6-->BALB.B combination, we studied the occurrence of lethal GVHD mediated by CD4+ T cells in recipient mice expressing only hematopoietically derived alloantigens. Even substantial dosages of donor C57BL/6 CD4+ T cells were unable to elicit lethal GVHD when transplanted into [BALB.B-->C57BL/6] chimeras. Instead, chimeric mice displayed transient cachexia with reduced target-tissue injury over time, reflecting an early, limited, graft-versus-host response. On the other hand, the importance of minor histocompatibility antigens derived from nonhematopoietic tissues was demonstrated by the finding that [C57BL/6-->BALB.B] chimeric mice succumbed to C57BL/6 CD4+ T cell-mediated GVHD. These data suggest that severe acute CD4+ T cell-mediated GVHD across this minor histocompatibility antigen barrier depends on the expression of nonhematopoietically rather than hematopoietically derived alloantigens for maximal target-tissue infiltration and injury.


Biology of Blood and Marrow Transplantation | 2003

An Epithelial Target Site in Experimental Graft- versus-Host Disease and Cytokine-Mediated Cytotoxicity Is Defined by Cytokeratin 15 Expression

Diana Whitaker-Menezes; Stephen C. Jones; Thea M. Friedman; Robert Korngold; George F. Murphy

The identity of cells within squamous epithelia that represent primary targets in acute graft-versus-host disease (GVHD) has been an enigma. Murine effector T cells implicated in the alloresponse by Vbeta complementarity-determining region-3 spectratype analysis were detected with a Vbeta-specific monoclonal antibody within discrete microdomains of tongue (lingual) squamous epithelium. These microdomains, termed rete-like prominences (RLPs), are similar to the rete ridges of human skin. Cells forming the basal layer of RLPs and of human skin rete ridges were shown to express a distinctive pattern of keratin expression defined by antibodies to cytokeratin 15 (K15). In experimental murine GVHD elicited across minor histocompatibility antigen barriers (miHA), early lesions involved selective apoptosis and loss of K15(+) staining within lingual RLPs. An in vitro organ culture model designed to investigate target cell injury by short-term exposure to tumor necrosis factor-alpha and interleukin-1beta, mediators relevant to GVHD, showed a similar pattern of apoptosis and loss of K15(+) reactivity within RLPs. In aggregate, these findings establish a novel cytoskeletal marker for target epithelial subpopulations that should facilitate evaluation of mechanisms of host cell injury in GVHD. These data may also enable the development of therapeutic approaches to abrogate disease at the level of target cell blockade.


Journal of Immunotherapy | 2010

Generation of Tumor-specific T Lymphocytes Using Dendritic Cell/Tumor Fusions and Anti-CD3/CD28

Jacalyn Rosenblatt; Zekui Wu; Baldev Vasir; Corrine Zarwan; Richard Stone; Heidi Mills; Thea M. Friedman; Panagiotis A. Konstantinopoulos; Dimitrios Spentzos; Musie Ghebremichael; Kristen E. Stevenson; Donna Neuberg; James D. Levine; Robin Joyce; Dimitrios Tzachanis; Vassiliki A. Boussiotis; Donald Kufe; David Avigan

Adoptive immunotherapy with tumor-specific T cells represents a promising treatment strategy for patients with malignancy. However, the efficacy of T-cell therapy has been limited by the ability to expand tumor-reactive cells with an activated phenotype that effectively target malignant cells. We have developed an anticancer vaccine in which patient-derived tumor cells are fused with autologous dendritic cells (DCs), such that a wide array of tumor antigens are presented in the context of DC-mediated costimulation. In this study, we demonstrate that DC/tumor fusions induce T cells that react with tumor and are dramatically expanded by subsequent ligation of the CD3/CD28 costimulatory complex. These T cells exhibit a predominantly activated phenotype as manifested by an increase in the percentage of cells expressing CD69 and interferon γ. In addition, the T cells upregulate granzyme B expression and are highly effective in lysing autologous tumor targets. Targeting of tumor-specific antigen was demonstrated by the expansion of T cells with specificity for the MUC1 tetramer. Stimulation with anti-CD3/CD28 followed by DC/tumor fusions or either agent alone failed to result in a similar expansion of tumor-reactive T cells. Consistent with these findings, spectratyping analysis demonstrates selective expansion of T-cell clones as manifested by considerable skewing of the Vβ repertoire following sequential stimulation with DC/tumor fusions and anti-CD3/CD28. Gene expression analysis was notable for the upregulation of inflammatory pathways. These findings indicate that stimulation with DC/tumor fusions provides a unique platform for subsequent expansion with anti-CD3/CD28 in adoptive T-cell therapy of cancer.


Blood | 2008

Overlap between in vitro donor antihost and in vivo posttransplantation TCR Vβ use: a new paradigm for designer allogeneic blood and marrow transplantation

Thea M. Friedman; Kira Goldgirsh; Stephanie A. Berger; Jenny Zilberberg; Joanne Filicko-O'Hara; Neal Flomenberg; Michele Donato; Scott D. Rowley; Robert Korngold

Following allogeneic blood and marrow transplantation (BMT), mature donor T cells can enhance engraftment, counteract opportunistic infections, and mount graft-versus-tumor (GVT) responses, but at the risk of developing graft-versus-host disease (GVHD). With the aim of separating the beneficial effects of donor T cells from GVHD, one approach would be to selectively deplete subsets of alloreactive T cells in the hematopoietic cell inoculum. In this regard, TCR Vbeta repertoire analysis by CDR3-size spectratyping can be a powerful tool for the characterization of alloreactive T-cell responses. We investigated the potential of this spectratype approach by comparing the donor T-cell alloresponses generated in vitro against patient peripheral blood lymphocytes (PBLs) with those detected in vivo posttransplantation. The results indicated that for most Vbeta families that exhibited alloreactive CDR3-size skewing, there was a robust overlap between the in vitro antipatient and in vivo spectratype histograms. Thus, in vitro spectratype analysis may be useful for determining the alloreactive T-cell response involved in GVHD development and, thereby, could serve to guide select Vbeta family depletion for designer transplants to improve outcomes.


Journal of Immunology | 2009

The Immunological Impact of Genetic Drift in the B10.BR Congenic Inbred Mouse Strain

Stacey L. Fanning; Michael Y. Appel; Stephanie A. Berger; Robert Korngold; Thea M. Friedman

The MHC-matched, minor histocompatibility Ag (miHA)-mismatched B10.BR→CBA strain combination has been used to elucidate the immunobiology of graft-vs-host disease (GVHD) following allogeneic bone marrow transplantation. Studies conducted in the 1980s had established that B10.BR CD8+ T cells were capable of mediating GVHD in the absence of CD4+ T cells, and that CD4+ T cells were unable to induce lethal disease. In more recent studies with this GVHD model, we detected etiological discrepancies with the previously published results, which suggested that genetic drift might have occurred within the B10.BR strain. In particular, there was increased allorecognition of CBA miHA by B10.BR CD4+ T cells, as determined by both TCR Vβ spectratype analysis and the induction of lethal GVHD in CBA recipients. Additionally, alloreactivity was observed between the genetically drifted mice (B10.BR/Jdrif) and mice rederived from frozen embryos of the original strain (B10.BR/Jrep) using Vβ spectratype analysis and IFN-γ ELISPOT assays, suggesting that new miHA differences had arisen between the mice. Furthermore, T cell-depleted B10.BR/Jdrif bone marrow cells were unable to provide long-term survival following either allogeneic or syngeneic bone marrow transplantation. Gene expression analysis revealed several genes involved in hematopoiesis that were overexpressed in the lineage-negative fraction of B10.BR/Jdrif bone marrow, as compared with B10.BR/Jrep mice. Taken together, these results suggest that genetic drift in the B10.BR strain has significantly impacted the immune alloreactive response in the GVHD model by causing altered expression of miHA and diminished capacity for survival following transplantation into lethally irradiated recipients.


Journal of Immunology | 2008

Inter-Strain Tissue-Infiltrating T Cell Responses to Minor Histocompatibility Antigens Involved in Graft-Versus-Host Disease as Determined by Vβ Spectratype Analysis

Jenny Zilberberg; Danielle McElhaugh; Loise N. Gichuru; Robert Korngold; Thea M. Friedman

Lethal graft-vs-host disease (GVHD) can be induced between MHC-matched murine strains expressing multiple minor histocompatibility Ag differences. In the B6–>BALB.B model, both CD4+ and CD8+ donor T cells can mediate lethal GVHD, whereas in the B6–>CXB-2 model, only CD8+ T cells are lethal. TCR Vβ CDR3-size spectratyping was previously used to analyze CD8+ and CD4+ T cell responses in lethally irradiated BALB.B and CXB-2 recipients, which showed significant overlap in the reacting repertoires. However, CD4+ T cells exhibited unique skewing of the Vβ2 and 11 families in only BALB.B recipients. These Vβ family reactivities were confirmed by immunohistochemical staining of lingual epithelial infiltrates, and by positive and negative selection Vβ family transfer experiments for GVHD induction in BALB.B recipients. We have now extended these studies to examine the T cell repertoire responses involved in target tissue damage. Infiltrating B6 host-presensitized CD8+ and CD4+ T cells were isolated 8–10 days post-transplant from the spleens, intestines and livers of CXB-2 and BALB.B transplant recipients. For both T cell subsets, the results indicated overlapping tissue skewings between the recipients, also between the tissues sampled within the respective recipients as well as tissue specific responses unique to both the BALB.B and CXB-2 infiltrates. Most notably, the CD4+ Vβ 11+ family was skewed in the intestines of BALB.B but not CXB-2 recipients. Taken together, these data suggest that there are likely to be target tissue-related anti-multiple minor histocompatibility Ag-specific responses in each of the strain recipients, which may also differ from those found in peripheral lymphoid organs.


Journal of Immunology | 2013

Unraveling Graft-versus-Host Disease and Graft-versus-Leukemia Responses Using TCR Vβ Spectratype Analysis in a Murine Bone Marrow Transplantation Model

S.L. Fanning; Jenny Zilberberg; Johann Stein; Kristin Vazzana; Stephanie A. Berger; Robert Korngold; Thea M. Friedman

The optimum use of allogeneic blood and marrow transplantation (BMT) as a curative therapy for hematological malignancies lies in the successful separation of mature donor T cells that are host reactive and induce graft-versus-host disease (GVHD) from those that are tumor reactive and mediate graft-versus-leukemia (GVL) effects. To study whether this separation was possible in an MHC-matched murine BMT model (B10.BR→CBA) with a CBA-derived myeloid leukemia line, MMC6, we used TCR Vβ CDR3-size spectratype analysis to first show that the Vβ13 family was highly skewed in the B10.BR anti-MMC6 CD8+ T cell response but not in the alloresponse against recipient cells alone. Transplantation of CD8+Vβ13+ T cells at the dose equivalent of their constituency in 1 × 107 CD8+ T cells, a dose that had been shown to mediate lethal GVHD in recipient mice, induced a slight GVL response with no concomitant GVHD. Increasing doses of CD8+Vβ13+ T cells led to more significant GVL responses but also increased GVHD symptoms and associated mortality. Subsequent spectratype analysis of GVHD target tissues revealed involvement of gut-infiltrating CD8+Vβ13+ T cells accounting for the observed in vivo effects. When BMT recipients were given MMC6-presensitized CD8+Vβ13+ T cells, they displayed a significant GVL response with minimal GVHD. Spectratype analysis of tumor-presensitized, gut-infiltrating CD8+Vβ13+ T cells showed preferential usage of tumor-reactive CDR3-size lengths, and these cells expressed increased effector memory phenotype (CD44+CD62L−/lo). Thus, Vβ spectratyping can identify T cells involved in antihost and antitumor reactivity and tumor presensitization can aid in the separation of GVHD and GVL responses.


Biology of Blood and Marrow Transplantation | 2009

Antiviral Responses following L-Leucyl-L-Leucine Methyl Esther (LLME)-Treated Lymphocyte Infusions: Graft-versus-Infection without Graft-versus-Host Disease

Joanne Filicko-O'Hara; Dolores Grosso; Phyllis Flomenberg; Thea M. Friedman; Janet Brunner; William R. Drobyski; Andres Ferber; Irina Kakhniashvili; Carolyn A. Keever-Taylor; Bijoyesh Mookerjee; Julie-An Talano; John I. Wagner; Robert Korngold; Neal Flomenberg

Although allogeneic hematopoietic progenitor cell transplant (HPCT) is curative therapy for many disorders, it is associated with significant morbidity and mortality, which can be related to graft-versus-host disease (GVHD) and the immunosuppressive measures required for its prevention and/or treatment. Whether the immunosuppression is pharmacologic or secondary to graft manipulation, the graft recipient is left at increased risk of the threatening opportunistic infection. Refractory viral diseases in the immunocompromised host have been treated by infusion of virus-specific lymphotyces and by unmanipulated donor lymphocyte infusion (DLI) therapy. L-leucyl-L-leucine methyl ester (LLME) is a compound that induces programmed cell death of natural killer (NK) cells, monocytes, granulocytes, most CD8(+) T cells, and a small fraction of CD4(+) T cells. We have undertaken a study of the use of LLME-treated DLI following T cell-depleted allogeneic HPCT, specifically to aid with immune reconstitution. In this ongoing clinical trial, we have demonstrated the rapid emergence of virus-specific responses following LLME DLI with minimal associated GVHD. This paper examines the pace of immune recovery and the rapid development of antiviral responses in 6 patients who developed viral infections during the time period immediately preceding or coincident with the administration of the LLME DLI.


Clinical Immunology (Third Edition)#R##N#Principles and Practice | 2008

82 – Hematopoietic stem cell transplantation for malignant diseases

Scott D. Rowley; Thea M. Friedman; Robert Korngold

Abstract Hematopoietic stem cell transplantation (HSCT) is an effective treatment for most hematological malignancies. Autologous HSCT targets malignancies in a dose-responsive manner, but regimen-related toxicities limit the intensity of the treatment regimen that can be given. Autologous HSCT may serve as a platform for subsequent therapies. Allogeneic HSCT offers a form of immunotherapy against malignancies, as demonstrated by a powerful graft-versus-tumor (GvT) effect in some malignancies. Allogeneic HSCT, however, has a higher incidence of morbidity and mortality from the similar graft-versus-host disease (GvHD), and maintaining a fine balance between GvT and GvHD is a continuing area of evolution, expertise, and research. The subtle nuances of donor selection, degree of human leukocyte antigen (HLA) disparity, type of conditioning regimen, and the choice of stem cell source all influence the outcome of allogeneic HSCT. Relapse is the most common cause of failure of autologous and allogeneic HSCT, and the use of immunotherapy during and/or after HSCT is the focus of research in this decade.

Collaboration


Dive into the Thea M. Friedman's collaboration.

Top Co-Authors

Avatar

Robert Korngold

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jenny Zilberberg

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

George F. Murphy

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Neal Flomenberg

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Scott D. Rowley

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stephen C. Jones

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Kira Goldgirsh

Hackensack University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Song Li

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Swati Choksi

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Andrea E. Edling

Thomas Jefferson University

View shared research outputs
Researchain Logo
Decentralizing Knowledge