Network


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

Hotspot


Dive into the research topics where Steven E. Finkelstein is active.

Publication


Featured researches published by Steven E. Finkelstein.


Journal of Experimental Medicine | 2003

Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells.

Willem W. Overwijk; Marc R. Theoret; Steven E. Finkelstein; Deborah R. Surman; Laurina A. de Jong; Florry A. Vyth-Dreese; Trees A. M. Dellemijn; Paul A. Antony; Paul J. Spiess; Douglas C. Palmer; David M. Heimann; Christopher A. Klebanoff; Zhiya Yu; Leroy N. Hwang; Lionel Feigenbaum; Ada M. Kruisbeek; Steven A. Rosenberg; Nicholas P. Restifo

Many tumor-associated antigens are derived from nonmutated “self” proteins. T cells infiltrating tumor deposits recognize self-antigens presented by tumor cells and can be expanded in vivo with vaccination. These T cells exist in a functionally tolerant state, as they rarely result in tumor eradication. We found that tumor growth and lethality were unchanged in mice even after adoptive transfer of large numbers of T cells specific for an MHC class I–restricted epitope of the self/tumor antigen gp100. We sought to develop new strategies that would reverse the functionally tolerant state of self/tumor antigen-reactive T cells and enable the destruction of large (with products of perpendicular diameters of >50 mm2), subcutaneous, unmanipulated, poorly immunogenic B16 tumors that were established for up to 14 d before the start of treatment. We have defined three elements that are all strictly necessary to induce tumor regression in this model: (a) adoptive transfer of tumor-specific T cells; (b) T cell stimulation through antigen-specific vaccination with an altered peptide ligand, rather than the native self-peptide; and (c) coadministration of a T cell growth and activation factor. Cells, vaccination, or cyto-kine given alone or any two in combination were insufficient to induce tumor destruction. Autoimmune vitiligo was observed in mice cured of their disease. These findings illustrate that adoptive transfer of T cells and IL-2 can augment the function of a cancer vaccine. Furthermore, these data represent the first demonstration of complete cures of large, established, poorly immunogenic, unmanipulated solid tumors using T cells specific for a true self/tumor antigen and form the basis for a new approach to the treatment of patients with cancer.


Journal of Experimental Medicine | 2005

Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells

Luca Gattinoni; Steven E. Finkelstein; Christopher A. Klebanoff; Paul A. Antony; Douglas C. Palmer; Paul J. Spiess; Leroy N. Hwang; Zhiya Yu; Claudia Wrzesinski; David M. Heimann; Charles D. Surh; Steven A. Rosenberg; Nicholas P. Restifo

Depletion of immune elements before adoptive cell transfer (ACT) can dramatically improve the antitumor efficacy of transferred CD8+ T cells, but the specific mechanisms that contribute to this enhanced immunity remain poorly defined. Elimination of CD4+CD25+ regulatory T (T reg) cells has been proposed as a key mechanism by which lymphodepletion augments ACT-based immunotherapy. We found that even in the genetic absence of T reg cells, a nonmyeloablative regimen substantially augmented CD8+ T cell reactivity to self-tissue and tumor. Surprisingly, enhanced antitumor efficacy and autoimmunity was caused by increased function rather than increased numbers of tumor-reactive T cells, as would be expected by homeostatic mechanisms. The γ C cytokines IL-7 and IL-15 were required for augmenting T cell functionality and antitumor activity. Removal of γ C cytokine–responsive endogenous cells using antibody or genetic means resulted in the enhanced antitumor responses similar to those seen after nonmyeloablative conditioning. These data indicate that lymphodepletion removes endogenous cellular elements that act as sinks for cytokines that are capable of augmenting the activity of self/tumor-reactive CD8+ T cells. Thus, the restricted availability of homeostatic cytokines can be a contributing factor to peripheral tolerance, as well as a limiting resource for the effectiveness of tumor-specific T cells.


Journal of Clinical Investigation | 2005

Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8 + T cells

Luca Gattinoni; Christopher A. Klebanoff; Douglas C. Palmer; Claudia Wrzesinski; Keith W. Kerstann; Zhiya Yu; Steven E. Finkelstein; Marc R. Theoret; Steven A. Rosenberg; Nicholas P. Restifo

T cell differentiation is a progressive process characterized by phenotypic and functional changes. By transferring tumor-specific CD8+ T cells into tumor-bearing mice at various stages of differentiation, we evaluated their efficacy for adoptive immunotherapy. We found that administration of naive and early effector T cells, in combination with active immunization and IL-2, resulted in the eradication of large, established tumors. Despite enhanced in vitro antitumor properties, more-differentiated effector T cells were less effective for in vivo tumor treatment. Several events may underlie this paradoxical phenomenon: (a) downregulation of lymphoid-homing and costimulatory molecules; (b) inability to produce IL-2 and access homeostatic cytokines; and (c) entry into a proapoptotic and replicative senescent state. While the progressive acquisition of terminal effector properties is characterized by pronounced in vitro tumor killing, in vivo T cell activation, proliferation, and survival are progressively impaired. These findings suggest that the current methodology for selecting T cells for transfer is inadequate and provide new criteria for the generation and the screening of optimal lymphocyte populations for adoptive immunotherapy.


Journal of Immunology | 2005

CD8+ T Cell Immunity Against a Tumor/Self-Antigen Is Augmented by CD4+ T Helper Cells and Hindered by Naturally Occurring T Regulatory Cells

Paul A. Antony; Ciriaco A. Piccirillo; Akgul Akpinarli; Steven E. Finkelstein; Paul J. Speiss; Deborah R. Surman; Douglas C. Palmer; Chi-Chao Chan; Christopher A. Klebanoff; Willem W. Overwijk; Steven A. Rosenberg; Nicholas P. Restifo

CD4+ T cells control the effector function, memory, and maintenance of CD8+ T cells. Paradoxically, we found that absence of CD4+ T cells enhanced adoptive immunotherapy of cancer when using CD8+ T cells directed against a persisting tumor/self-Ag. However, adoptive transfer of CD4+CD25− Th cells (Th cells) with tumor/self-reactive CD8+ T cells and vaccination into CD4+ T cell-deficient hosts induced autoimmunity and regression of established melanoma. Transfer of CD4+ T cells that contained a mixture of Th and CD4+CD25+ T regulatory cells (Treg cells) or Treg cells alone prevented effective adoptive immunotherapy. Maintenance of CD8+ T cell numbers and function was dependent on Th cells that were capable of IL-2 production because therapy failed when Th cells were derived from IL-2−/− mice. These findings reveal that Th cells can help break tolerance to a persisting self-Ag and treat established tumors through an IL-2-dependent mechanism, but requires simultaneous absence of naturally occurring Treg cells to be effective.


Journal of Experimental Medicine | 2005

Synergy of IL-21 and IL-15 in regulating CD8+ T cell expansion and function

Rong Zeng; Rosanne Spolski; Steven E. Finkelstein; SangKon Oh; Panu E. Kovanen; Christian S. Hinrichs; Cynthia A. Pise-Masison; Michael F. Radonovich; John N. Brady; Nicholas P. Restifo; Jay A. Berzofsky; Warren J. Leonard

Interleukin (IL)-21 is the most recently recognized of the cytokines that share the common cytokine receptor γ chain (γc), which is mutated in humans with X-linked severe combined immunodeficiency. We now report that IL-21 synergistically acts with IL-15 to potently promote the proliferation of both memory (CD44high) and naive (CD44low) phenotype CD8+ T cells and augment interferon-γ production in vitro. IL-21 also cooperated, albeit more weakly, with IL-7, but not with IL-2. Correspondingly, the expansion and cytotoxicity of CD8+ T cells were impaired in IL-21R−/− mice. Moreover, in vivo administration of IL-21 in combination with IL-15 boosted antigen-specific CD8+ T cell numbers and resulted in a cooperative effect on tumor regression, with apparent cures of large, established B16 melanomas. Thus, our studies reveal that IL-21 potently regulates CD8+ T cell expansion and effector function, primarily in a synergistic context with IL-15.


Proceedings of the National Academy of Sciences of the United States of America | 2004

IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells

Christopher A. Klebanoff; Steven E. Finkelstein; Deborah R. Surman; Michael K. Lichtman; Luca Gattinoni; Marc R. Theoret; Navrose Grewal; Paul J. Spiess; Paul A. Antony; Douglas C. Palmer; Yutaka Tagaya; Steven A. Rosenberg; Thomas A. Waldmann; Nicholas P. Restifo

IL-15 and IL-2 possess similar properties, including the ability to induce T cell proliferation. However, whereas IL-2 can promote apoptosis and limit CD8+ memory T cell survival and proliferation, IL-15 helps maintain a memory CD8+ T cell population and can inhibit apoptosis. We sought to determine whether IL-15 could enhance the in vivo function of tumor/self-reactive CD8+ T cells by using a T cell receptor transgenic mouse (pmel-1) whose CD8+ T cells recognize an epitope derived from the self/melanoma antigen gp100. By removing endogenous IL-15 by using tumor-bearing IL-15 knockout hosts or supplementing IL-15 by means of exogenous administration, as a component of culture media or as a transgene expressed by adoptively transferred T cells, we demonstrate that IL-15 can improve the in vivo antitumor activity of adoptively transferred CD8+ T cells. These results provide several avenues for improving adoptive immunotherapy of cancer in patients.


Clinical Cancer Research | 2011

Determinants of Successful CD8+ T-Cell Adoptive Immunotherapy for Large Established Tumors in Mice

Christopher A. Klebanoff; Luca Gattinoni; Douglas C. Palmer; Pawel Muranski; Yun Ji; Christian S. Hinrichs; Zachary A. Borman; Sid P. Kerkar; Christopher D. Scott; Steven E. Finkelstein; Steven A. Rosenberg; Nicholas P. Restifo

Purpose: Adoptive cell transfer (ACT) of tumor infiltrating or genetically engineered T cells can cause durable responses in patients with metastatic cancer. Multiple clinically modifiable parameters can comprise this therapy, including cell dose and phenotype, in vivo antigen restimulation, and common gamma-chain (γc) cytokine support. However, the relative contributions of each these individual components to the magnitude of the antitumor response have yet to be quantified. Experimental Design: To systematically and quantitatively appraise each of these variables, we employed the Pmel-1 mouse model treating large, established B16 melanoma tumors. In addition to cell dose and magnitude of in vivo antigen restimulation, we also evaluated the relative efficacy of central memory (TCM), effector memory (TEM), and stem cell memory (TSCM) subsets on the strength of tumor regression as well as the dose and type of clinically available γc cytokines, including IL-2, IL-7, IL-15, and IL-21. Results: We found that cell dose, T-cell differentiation status, and viral vaccine titer each were correlated strongly and significantly with the magnitude of tumor regression. Surprisingly, although the total number of IL-2 doses was correlated with tumor regression, no significant benefit to prolonged (≥6 doses) administration was observed. Moreover, the specific type and dose of γc cytokine only moderately correlated with response. Conclusion: Collectively, these findings elucidate some of the key determinants of successful ACT immunotherapy for the treatment of cancer in mice and further show that γc cytokines offer a similar ability to effectively drive antitumor T-cell function in vivo. Clin Cancer Res; 17(16); 5343–52. ©2011 AACR.


Journal of Experimental Medicine | 2003

Immune Evasion by Murine Melanoma Mediated through CC Chemokine Receptor-10

Takashi Murakami; Adela R. Cardones; Steven E. Finkelstein; Nicholas P. Restifo; Brenda A. Klaunberg; Frank O. Nestle; S. Sianna Castillo; Phillip A. Dennis; Sam T. Hwang

Human melanoma cells frequently express CC chemokine receptor (CCR)10, a receptor whose ligand (CCL27) is constitutively produced by keratinocytes. Compared with B16 murine melanoma, cells rendered more immunogenic via overexpression of luciferase, B16 cells that overexpressed both luciferase and CCR10 resisted host immune responses and readily formed tumors. In vitro, exposure of tumor cells to CCL27 led to rapid activation of Akt, resistance to cell death induced by melanoma antigen-specific cytotoxic T cells, and phosphatidylinositol-3-kinase (PI3K)–dependent protection from apoptosis induced by Fas cross-linking. In vivo, cutaneous injection of neutralizing antibodies to endogenous CCL27 blocked growth of CCR10-expressing melanoma cells. We propose that CCR10 engagement by locally produced CCL27 allows melanoma cells to escape host immune antitumor killing mechanisms (possibly through activation of PI3K/Akt), thereby providing a means for tumor progression.


Cancer Research | 2004

Dendritic Cells Strongly Boost the Antitumor Activity of Adoptively Transferred T Cells In vivo

Yanyan Lou; Gang Wang; Gregory Lizée; Grace J. Kim; Steven E. Finkelstein; Chiguang Feng; Nicholas P. Restifo; Patrick Hwu

Dendritic cells (DCs) have been well characterized for their ability to initiate cell-mediated immune responses by stimulating naive T cells. However, the use of DCs to stimulate antigen-activated T cells in vivo has not been investigated. In this study, we determined whether DC vaccination could improve the efficacy of activated, adoptively transferred T cells to induce an enhanced antitumor immune response. Mice bearing B16 melanoma tumors expressing the gp100 tumor antigen were treated with cultured, activated T cells transgenic for a T-cell receptor specifically recognizing gp100, with or without concurrent peptide-pulsed DC vaccination. In this model, antigen-specific DC vaccination induced cytokine production, enhanced proliferation, and increased tumor infiltration of adoptively transferred T cells. Furthermore, the combination of DC vaccination and adoptive T-cell transfer led to a more robust antitumor response than the use of each treatment individually. Collectively, these findings illuminate a new potential application for DCs in the in vivo stimulation of adoptively transferred T cells and may be a useful approach for the immunotherapy of cancer.


Cancer Research | 2005

Bcl-2 Overexpression Enhances Tumor-Specific T-Cell Survival

Jehad Charo; Steven E. Finkelstein; Navrose Grewal; Nicholas P. Restifo; Paul F. Robbins; Steven A. Rosenberg

Although immunotherapy based on the adoptive transfer of tumor-specific T lymphocytes has been shown to result in dramatic clinical responses in some patients, the relatively low levels of engraftment and persistence of the adoptively transferred cells may limit these responses in many patients. In an attempt to develop strategies for prolonging the survival of adoptively transferred T cells, we have carried out studies in which T cells obtained from healthy donors as well as tumor-specific T cells were transduced with a retrovirus expressing the human Bcl-2 gene. Our results indicate that these transduced T cells overexpress Bcl-2, are resistant to death, and have a survival advantage following interleukin-2 withdrawal compared with control T cells transduced with a retrovirus expressing green fluorescent protein. Tumor-specific T cells overexpressing Bcl-2 maintained their ability to specifically recognize and respond to target cells. Furthermore, we show that adoptive immunotherapy of an established B16 tumor can be significantly enhanced by overexpressing Bcl-2 in melanoma-specific T-cell receptor transgenic T cells. Our data suggest that adoptive immunotherapy approaches to the treatment of cancer patients may be enhanced using Bcl-2-modified tumor-reactive T cells.

Collaboration


Dive into the Steven E. Finkelstein's collaboration.

Top Co-Authors

Avatar

Nicholas P. Restifo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Steven A. Rosenberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Douglas C. Palmer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luca Gattinoni

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Deborah R. Surman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhiya Yu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Christian S. Hinrichs

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David S. Schrump

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge