Network


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

Hotspot


Dive into the research topics where Christopher A. Klebanoff is active.

Publication


Featured researches published by Christopher A. Klebanoff.


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.


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.


Immunological Reviews | 2006

CD8+ T-cell memory in tumor immunology and immunotherapy

Christopher A. Klebanoff; Luca Gattinoni; Nicholas P. Restifo

Summary:  The cellular and molecular mechanisms underlying the formation of distinct central, effector, and exhausted CD8+ T‐cell memory subsets were first described in the setting of acute and chronic viral diseases. The role of these T‐cell memory subsets are now being illuminated as relevant to the tumor‐bearing state. The generation and persistence of productive CD8+ T‐cell memory subsets is determined, in part, by antigen clearance, costimulation, responsiveness to homeostatic cytokines, and CD4+ T‐helper cells. By contrast, chronic exposure to antigen, negative costimulation, and immunomodulation by CD4+ T regulatory cells corrupt productive CD8+ T memory formation. It has become clear from human and mouse studies that the mere generation of CD8+ T‐cell memory is not a ‘surrogate marker’ for cancer vaccine efficacy. Some current cancer vaccine strategies may fail because they amplify, rather than correct or reset, the corrupted CD8+ memory population. Thus, much of the present effort in the development of vaccines for cancer and chronic infectious diseases is aimed at creating effective memory responses. Therapeutic vaccines for cancer and chronic infectious diseases may achieve consistent efficacy by ablation of the dysfunctional immune state and the provision of newly generated, non‐corrupted memory cells by adoptive cell transfer.


Blood | 2008

IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy

Christian S. Hinrichs; Rosanne Spolski; Chrystal M. Paulos; Luca Gattinoni; Keith W. Kerstann; Douglas C. Palmer; Christopher A. Klebanoff; Steven A. Rosenberg; Warren J. Leonard; Nicholas P. Restifo

IL-2 and IL-21 are closely related cytokines that might have arisen by gene duplication. Both cytokines promote the function of effector CD8(+) T cells, but their distinct effects on antigen-driven differentiation of naive CD8(+) T cells into effector CD8(+) T cells are not clearly understood. We found that antigen-induced expression of Eomesodermin (Eomes) and maturation of naive CD8(+) T cells into granzyme B- and CD44-expressing effector CD8(+) T cells was enhanced by IL-2, but, unexpectedly, suppressed by IL-21. Furthermore, IL-21 repressed expression of IL-2Ra and inhibited IL-2-mediated acquisition of a cytolytic CD8(+) T-cell phenotype. Despite its inhibitory effects, IL-21 did not induce anergy, but instead potently enhanced the capacity of cells to mediate tumor regression upon adoptive transfer. In contrast, IL-2 impaired the subsequent antitumor function of transferred cells. Gene expression studies revealed a distinct IL-21 program that was characterized phenotypically by increased expression of L-selectin and functionally by enhanced antitumor immunity that was not reversed by secondary in vitro stimulation with antigen and IL-2. Thus, the efficacy of CD8(+) T cells for adoptive immunotherapy can be influenced by opposing differentiation programs conferred by IL-2 and IL-21, a finding with important implications for the development of cellular cancer therapies.


Journal of Clinical Investigation | 2013

Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function

Madhusudhanan Sukumar; Jie Liu; Yun Ji; Murugan Subramanian; Joseph G. Crompton; Zhiya Yu; Rahul Roychoudhuri; Douglas C. Palmer; Pawel Muranski; Edward D. Karoly; Robert P. Mohney; Christopher A. Klebanoff; Ashish Lal; Toren Finkel; Nicholas P. Restifo; Luca Gattinoni

Naive CD8+ T cells rely upon oxidation of fatty acids as a primary source of energy. After antigen encounter, T cells shift to a glycolytic metabolism to sustain effector function. It is unclear, however, whether changes in glucose metabolism ultimately influence the ability of activated T cells to become long-lived memory cells. We used a fluorescent glucose analog, 2-NBDG, to quantify glucose uptake in activated CD8+ T cells. We found that cells exhibiting limited glucose incorporation had a molecular profile characteristic of memory precursor cells and an increased capacity to enter the memory pool compared with cells taking up high amounts of glucose. Accordingly, enforcing glycolytic metabolism by overexpressing the glycolytic enzyme phosphoglycerate mutase-1 severely impaired the ability of CD8+ T cells to form long-term memory. Conversely, activation of CD8+ T cells in the presence of an inhibitor of glycolysis, 2-deoxyglucose, enhanced the generation of memory cells and antitumor functionality. Our data indicate that augmenting glycolytic flux drives CD8+ T cells toward a terminally differentiated state, while its inhibition preserves the formation of long-lived memory CD8+ T cells. These results have important implications for improving the efficacy of T cell-based therapies against chronic infectious diseases and cancer.


Nature Reviews Cancer | 2012

Paths to stemness: building the ultimate antitumour T cell

Luca Gattinoni; Christopher A. Klebanoff; Nicholas P. Restifo

Stem cells are defined by the ability to self-renew and to generate differentiated progeny, qualities that are maintained by evolutionarily conserved pathways that can lead to cancer when deregulated. There is now evidence that these stem cell-like attributes and signalling pathways are also shared among subsets of mature memory T lymphocytes. We discuss how using stem cell-like T cells can overcome the limitations of current adoptive T cell therapies, including inefficient T cell engraftment, persistence and ability to mediate prolonged immune attack. Conferring stemness to antitumour T cells might unleash the full potential of cellular therapies.


Immunity | 2011

Th17 Cells Are Long Lived and Retain a Stem Cell-like Molecular Signature

Pawel Muranski; Zachary A. Borman; Sid P. Kerkar; Christopher A. Klebanoff; Yun Ji; Luis Sanchez-Perez; Madhusudhanan Sukumar; Robert N. Reger; Zhiya Yu; Steven J. Kern; Rahul Roychoudhuri; Gabriela A. Ferreyra; Wei Shen; Scott K. Durum; Lionel Feigenbaum; Douglas C. Palmer; Paul A. Antony; Chi-Chao Chan; Arian Laurence; Robert L. Danner; Luca Gattinoni; Nicholas P. Restifo

Th17 cells have been described as short lived, but this view is at odds with their capacity to trigger protracted damage to normal and transformed tissues. We report that Th17 cells, despite displaying low expression of CD27 and other phenotypic markers of terminal differentiation, efficiently eradicated tumors and caused autoimmunity, were long lived, and maintained a core molecular signature resembling early memory CD8(+) cells with stem cell-like properties. In addition, we found that Th17 cells had high expression of Tcf7, a direct target of the Wnt and β-catenin signaling axis, and accumulated β-catenin, a feature observed in stem cells. In vivo, Th17 cells gave rise to Th1-like effector cell progeny and also self-renewed and persisted as IL-17A-secreting cells. Multipotency was required for Th17 cell-mediated tumor eradication because effector cells deficient in IFN-γ or IL-17A had impaired activity. Thus, Th17 cells are not always short lived and are a less-differentiated subset capable of superior persistence and functionality.

Collaboration


Dive into the Christopher A. Klebanoff's collaboration.

Top Co-Authors

Avatar

Nicholas P. Restifo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Luca Gattinoni

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

Zhiya Yu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Rahul Roychoudhuri

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
Top Co-Authors

Avatar

Madhusudhanan Sukumar

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yun Ji

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert L. Eil

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge