Network


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

Hotspot


Dive into the research topics where Jeffrey J. Roszkowski is active.

Publication


Featured researches published by Jeffrey J. Roszkowski.


Cancer Research | 2005

Simultaneous Generation of CD8+ and CD4+ Melanoma-Reactive T Cells by Retroviral-Mediated Transfer of a Single T-Cell Receptor

Jeffrey J. Roszkowski; Gretchen E. Lyons; W. Martin Kast; Cassian Yee; Koen van Besien; Michael I. Nishimura

Adoptive immunotherapy of cancer requires the generation of large numbers of tumor antigen-reactive T cells for transfer into cancer patients. Genes encoding tumor antigen-specific T-cell receptors can be introduced into primary human T cells by retroviral mediated gene transfer as a potential method of providing any patient with a source of autologous tumor-reactive T cells. A T-cell receptor-specific for a class I MHC (HLA-A2)-restricted epitope of the melanoma antigen tyrosinase was isolated from a CD4(+) tumor-infiltrating lymphocyte (TIL 1383I) and introduced into normal human peripheral blood lymphocytes by retroviral transduction. T-cell receptor-transduced T cells secreted various cytokines when cocultured with tyrosinase peptide-loaded antigen-presenting cells as well as melanoma cells in an HLA-A2-restricted manner, and could also lyse target cells. Furthermore, T-cell clones isolated from these cultures showed both CD8(+) and CD4(+) transduced T cells could recognize HLA-A2(+) melanoma cells, giving us the possibility of engineering class I MHC-restricted effector and T helper cells against melanoma. The ability to confer class I MHC-restricted tumor cell recognition to CD4(+) T cells makes the TIL 1383I TCR an attractive candidate for T-cell receptor gene transfer-based immunotherapy.


Journal of Immunology | 2003

CD8-Independent Tumor Cell Recognition Is a Property of the T Cell Receptor and Not the T Cell

Jeffrey J. Roszkowski; David C. Yu; Mark P. Rubinstein; Mark D. McKee; David J. Cole; Michael I. Nishimura

The CD8 coreceptor enhances T cell function by stabilizing the TCR/peptide/MHC complex and/or increasing T cell avidity via interactions with the intracellular kinases Lck and LAT. We previously reported a CD4+ T cell (TIL 1383I), which recognizes the tumor-associated Ag tyrosinase in the context of HLA-A2. To determine whether CD8 independent tumor cell recognition is a property of the TCR, we used retroviral transduction to express the TIL 1383I TCR in the CD8− murine lymphoma, 58 α−/β−. Immunofluorescent staining of TCR-transduced cells with human TCR Vβ subfamily-specific and mouse CD3-specific Abs confirmed surface expression of the transferred TCR and coexpression of mouse CD3. Transduced effector cells secreted significant amounts of IL-2 following Ag presentation by tyrosinase peptide-pulsed T2 cells as well as stimulation with HLA-A2+ melanoma lines compared with T2 cells alone or HLA-A2− melanoma cells. Further analysis of TCR-transduced clones demonstrated a correlation between T cell avidity and cell surface expression of the TCR. Therefore, the TIL 1383I TCR has sufficient affinity to mediate recognition of the physiologic levels of Ag expressed by tumor cells in the absence of CD8 expression.


Hepatology | 2006

Identification of a hepatitis C virus–reactive T cell receptor that does not require CD8 for target cell recognition

Glenda G. Callender; Hugo R. Rosen; Jeffrey J. Roszkowski; Gretchen E. Lyons; Mingli Li; Tamson V. Moore; Natasha Brasic; Mark D. McKee; Michael I. Nishimura

Hepatitis C virus (HCV) has been reported to elicit B and T cell immunity in infected patients. Despite the presence of antiviral immunity, many patients develop chronic infections leading to cirrhosis, hepatocellular carcinoma, and liver failure that can require transplantation. We have previously described the presence of HLA‐A2–restricted, HCV NS3–reactive cytotoxic T lymphocytes (CTL) in the blood of HLA‐A2− liver transplantation patients that received an HLA‐A2+ liver allograft. These T cells are analogous to the “allospecific” T cells that have been described in hematopoietic stem cell transplantation patients. It has been speculated that allospecific T cells express high‐affinity T cell receptors (TCRs). To determine if our HCV‐reactive T cells expressed TCRs with relatively high affinity for antigen, we identified and cloned a TCR from an allospecific HLA‐A2–restricted, HCV:NS3:1406‐1415–reactive CD8+ T cell clone and expressed this HCV TCR in Jurkat cells. Tetramer binding to HCV TCR–transduced Jurkat cells required CD8 expression, whereas antigen recognition did not. In conclusion, based on the reactivity of the TCR‐transduced Jurkat cells, we have identified a TCR that transfers anti‐HCV reactivity to alternate effectors. These data suggest this high affinity HCV‐specific TCR might have potential new immunotherapic implications. (HEPATOLOGY 2006;43:973–981.)


Journal of Translational Medicine | 2005

T cell avidity and tumor recognition: implications and therapeutic strategies

Mark D. McKee; Jeffrey J. Roszkowski; Michael I. Nishimura

In the last two decades, great advances have been made studying the immune response to human tumors. The identification of protein antigens from cancer cells and better techniques for eliciting antigen specific T cell responses in vitro and in vivo have led to improved understanding of tumor recognition by T cells. Yet, much remains to be learned about the intricate details of T cell – tumor cell interactions. Though the strength of interaction between T cell and target is thought to be a key factor influencing the T cell response, investigations of T cell avidity, T cell receptor (TCR) affinity for peptide-MHC complex, and the recognition of peptide on antigen presenting targets or tumor cells reveal complex relationships. Coincident with these investigations, therapeutic strategies have been developed to enhance tumor recognition using antigens with altered peptide structures and T cells modified by the introduction of new antigen binding receptor molecules. The profound effects of these strategies on T cell – tumor interactions and the clinical implications of these effects are of interest to both scientists and clinicians. In recent years, the focus of much of our work has been the avidity and effector characteristics of tumor reactive T cells. Here we review concepts and current results in the field, and the implications of therapeutic strategies using altered antigens and altered effector T cells.


Cancer Immunology, Immunotherapy | 2010

CD34-based enrichment of genetically engineered human T cells for clinical use results in dramatically enhanced tumor targeting

Håkan Norell; Yi Zhang; James McCracken; Telma Martins da Palma; Aaron Lesher; Yueying Liu; Jeffrey J. Roszkowski; Anquanette Temple; Glenda G. Callender; Timothy M. Clay; Rimas J. Orentas; José A. Guevara-Patiño; Michael I. Nishimura

Objective clinical responses can be achieved in melanoma patients by infusion of T cell receptor (TCR) gene transduced T cells. Although promising, the therapy is still largely ineffective, as most patients did not benefit from treatment. That only a minority of the infused T cells were genetically modified and that these were extensively expanded ex vivo may have prevented their efficacy. We developed novel and generally applicable retroviral vectors that allow rapid and efficient selection of T cells transduced with human TCRs. These vectors encode two TCR chains and a truncated CD34 molecule (CD34t) in a single mRNA transcript. Transduced T cells were characterized and the effects of CD34-based enrichment of redirected T cells were evaluated. Both CD8+ and CD4+ T cells could be transduced and efficiently co-expressed all introduced transgenes on their surface. Importantly, more than fivefold enrichment of both the frequency of transduced cells and the specific anti-tumor reactivity of the effector population could be achieved by magnetic beads-based enrichment procedures readily available for clinical grade hematopoietic stem cell isolation. This CD34-based enrichment technology will improve the feasibility of adoptive transfer of clinically relevant effectors. In addition to their enhanced tumor recognition, the enriched redirected T cells may also show superior reactivity and persistence in vivo due to the high purity of transduced cells and the shortened ex vivo culture.


Cancer Immunology, Immunotherapy | 2006

T-cell receptor tetramer binding or the lack there of does not necessitate antigen reactivity in T-cell receptor transduced T cells

Gretchen E. Lyons; Jeffrey J. Roszkowski; Stephen Tzekwung Man; Cassian Yee; W. Martin Kast; Michael I. Nishimura

Genetic transfer of T-cell receptor (TCR) chains provides a means of transferring tumor antigen specificity onto an alternate T-cell population. To determine which tumor reactive TCRs are best suitable for such adoptive transfer, careful evaluation of the resulting TCR modified populations need to be performed. We have previously cloned, and expressed TCRs from melanoma, EBV, HCV, and HPV reactive T-cell clones and found that several routine indicators of T-cell function do not always predict the relative strength of a TCR. Using a combination of tetramer binding assays and antigen recognition assays, we identified TCRs that fall into three classes. One class of TCR did not bind tetramers yet resulted in cells with high avidity for antigen. A second TCR class bound tetramer but did not secrete cytokines in response to antigen. Finally, the third class of TCRs bound tetramer and reacted to antigen as would be expected. We conclude that tetramer binding is not always a good indicator of the function of a cloned TCR or the avidity of a TCR gene modified T cell. Furthermore, our data indicate that the use of tetramer binding alone to identify antigen reactive TCRs may result in the exclusion of TCRs that may be highly reactive or cross reactive to the relevant tumor antigen.


Cancer Research | 2006

Influence of Human CD8 on Antigen Recognition by T-Cell Receptor-Transduced Cells

Gretchen E. Lyons; Tamson V. Moore; Natasha Brasic; Mingli Li; Jeffrey J. Roszkowski; Michael I. Nishimura

The CD8 coreceptor on T cells has two functions. Namely, CD8 acts to stabilize the binding of the T-cell receptor (TCR) to the peptide-MHC complex while localizing p56(lck) (lck) to the TCR/CD3 complex to facilitate early signaling events. Although both functions may be critical for efficient activation of a CTL, little is known about how the structural versus signaling roles of CD8, together with the relative strength of the TCR, influences T-cell function. We have addressed these issues by introducing full-length and truncated versions of the CD8alpha and CD8beta chains into CD8(-) Jurkat cell clones expressing cloned TCRs with known antigen specificity and relative affinities. Using a combination of antigen recognition and tetramer-binding assays, we find that the intracellular lck-binding domain of CD8 is critical for enhanced T-cell activation regardless of the relative strength of the TCR. In contrast, the extracellular domain of CD8 seems to be critical for TCRs with lower affinity but not those with higher affinity. Based on our results, we conclude that there are different requirements for CD8 to enhance T-cell function depending on the strength of its TCR.


Cancer Immunology, Immunotherapy | 2016

TCR gene-modified T cells can efficiently treat established hepatitis C-associated hepatocellular carcinoma tumors

Timothy T. Spear; Glenda G. Callender; Jeffrey J. Roszkowski; Kelly Moxley; Patricia Simms; Kendra C. Foley; David C. Murray; Gina Scurti; Mingli Li; Justin T. Thomas; Alexander Langerman; Elizabeth Garrett-Mayer; Yi Zhang; Michael I. Nishimura

Abstract The success in recent clinical trials using T cell receptor (TCR)-genetically engineered T cells to treat melanoma has encouraged the use of this approach toward other malignancies and viral infections. Although hepatitis C virus (HCV) infection is being treated with a new set of successful direct anti-viral agents, potential for virologic breakthrough or relapse by immune escape variants remains. Additionally, many HCV+ patients have HCV-associated disease, including hepatocellular carcinoma (HCC), which does not respond to these novel drugs. Further exploration of other approaches to address HCV infection and its associated disease are highly warranted. Here, we demonstrate the therapeutic potential of PBL-derived T cells genetically engineered with a high-affinity, HLA-A2-restricted, HCV NS3:1406-1415-reactive TCR. HCV1406 TCR-transduced T cells can recognize naturally processed antigen and elicit CD8-independent recognition of both peptide-loaded targets and HCV+ human HCC cell lines. Furthermore, these cells can mediate regression of established HCV+ HCC in vivo. Our results suggest that HCV TCR-engineered antigen-reactive T cells may be a plausible immunotherapy option to treat HCV-associated malignancies, such as HCC.


Cancer Immunology, Immunotherapy | 2009

Relationship between CD8-dependent antigen recognition, T cell functional avidity, and tumor cell recognition

Tamson V. Moore; Gretchen E. Lyons; Natasha Brasic; Jeffrey J. Roszkowski; Simon Voelkl; Andreas Mackensen; W. Martin Kast; I. Caroline Le Poole; Michael I. Nishimura

Effective immunotherapy using T cell receptor (TCR) gene-modified T cells requires an understanding of the relationship between TCR affinity and functional avidity of T cells. In this study, we evaluate the relative affinity of two TCRs isolated from HLA-A2-restricted, gp100-reactive T cell clones with extremely high functional avidity. Furthermore, one of these T cell clones, was CD4−CD8− indicating that antigen recognition by this clone was CD8 independent. However, when these TCRs were expressed in CD8− Jurkat cells, the resulting Jurkat cells recognized gp100:209–217 peptide loaded T2 cells and had high functional avidity, but could not recognize HLA-A2+ melanoma cells expressing gp100. Tumor cell recognition by Jurkat cells expressing these TCRs could not be induced by exogenously loading the tumor cells with the native gp100:209–217 peptide. These results indicate that functional avidity of a T cell does not necessarily correlate with TCR affinity and CD8-independent antigen recognition by a T cell does not always mean its TCR will transfer CD8-independence to other effector cells. The implications of these findings are that T cells can modulate their functional avidity independent of the affinity of their TCRs.


Journal of Leukocyte Biology | 2016

Hepatitis C virus‐cross‐reactive TCR gene‐modified T cells: a model for immunotherapy against diseases with genomic instability

Timothy T. Spear; Timothy P. Riley; Gretchen E. Lyons; Glenda G. Callender; Jeffrey J. Roszkowski; Yuan Wang; Patricia Simms; Gina Scurti; Kendra C. Foley; David C. Murray; Lance M. Hellman; Rachel H. McMahan; Makio Iwashima; Elizabeth Garrett-Mayer; Hugo R. Rosen; Brian M. Baker; Michael I. Nishimura

A major obstacle hindering the development of effective immunity against viral infections, their associated disease, and certain cancers is their inherent genomic instability. Accumulation of mutations can alter processing and presentation of antigens recognized by antibodies and T cells that can lead to immune escape variants. Use of an agent that can intrinsically combat rapidly mutating viral or cancer‐associated antigens would be quite advantageous in developing effective immunity against such disease. We propose that T cells harboring cross‐reactive TCRs could serve as a therapeutic agent in these instances. With the use of hepatitis C virus, known for its genomic instability as a model for mutated antigen recognition, we demonstrate cross‐reactivity against immunogenic and mutagenic nonstructural protein 3:1406‐1415 and nonstructural protein 3:1073‐1081 epitopes in PBL‐derived, TCR‐gene‐modified T cells. These single TCR‐engineered T cells can CD8‐independently recognize naturally occurring and epidemiologically relevant mutant variants. TCR‐peptide MHC modeling data allow us to rationalize how TCR structural properties accommodate recognition of certain mutated epitopes and how these substitutions impact the requirement of CD8 affinity enhancement for recognition. A better understanding of such TCRs’ promiscuous behavior may allow for exploitation of these properties to develop novel, adoptive T cell‐based therapies for viral infections and cancers exhibiting similar genomic instability.

Collaboration


Dive into the Jeffrey J. Roszkowski's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mingli Li

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

W. Martin Kast

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Cassian Yee

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David C. Murray

Loyola University Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge