Network


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

Hotspot


Dive into the research topics where David Clever is active.

Publication


Featured researches published by David Clever.


Blood | 2011

IPH2101, a novel anti-inhibitory KIR antibody, and lenalidomide combine to enhance the natural killer cell versus multiple myeloma effect.

Don M. Benson; Courtney E. Bakan; Shuhong Zhang; Shauna M. Collins; Jing Liang; Shivani Srivastava; Craig C. Hofmeister; Yvonne A. Efebera; Pascale Andre; Francois Romagne; Mathieu Blery; Cécile Bonnafous; Jianying Zhang; David Clever; Michael A. Caligiuri; Sherif S. Farag

Multiple myeloma (MM) patients who receive killer cell Ig-like receptor (KIR) ligand-mismatched, T cell-depleted, allogeneic transplantation may have a reduced risk of relapse compared with patients who receive KIR ligand-matched grafts, suggesting the importance of this signaling axis in the natural killer (NK) cell-versus-MM effect. Expanding on this concept, IPH2101 (1-7F9), an anti-inhibitory KIR mAb, enhances NK-cell function against autologous MM cells by blocking the engagement of inhibitory KIR with cognate ligands, promoting immune complex formation and NK-cell cytotoxicity specifically against MM cell targets but not normal cells. IPH2101 prevents negative regulatory signals by inhibitory KIR, whereas lenalidomide augments NK-cell function and also appears to up-regulate ligands for activating NK-cell receptors on MM cells. Lenalidomide and a murine anti-inhibitory NK-cell receptor Ab mediate in vivo rejection of a lenalidomide-resistant tumor. These mechanistic, preclinical data support the use of a combination of IPH2101 and lenalidomide in a phase 2 trial for MM.


Cancer Research | 2015

Akt Inhibition Enhances Expansion of Potent Tumor-Specific Lymphocytes with Memory Cell Characteristics

Joseph G. Crompton; Madhusudhanan Sukumar; Rahul Roychoudhuri; David Clever; Alena Gros; Robert L. Eil; Eric Tran; Ken Ichi Hanada; Zhiya Yu; Douglas C. Palmer; Sid P. Kerkar; Ryan D. Michalek; Trevor Upham; Anthony J. Leonardi; Nicolas Acquavella; Ena Wang; Francesco M. Marincola; Luca Gattinoni; Pawel Muranski; Mark S. Sundrud; Christopher A. Klebanoff; Steven A. Rosenberg; Nicholas P. Restifo

Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) results in complete regression of advanced cancer in some patients, but the efficacy of this potentially curative therapy may be limited by poor persistence of TIL after adoptive transfer. Pharmacologic inhibition of the serine/threonine kinase Akt has recently been shown to promote immunologic memory in virus-specific murine models, but whether this approach enhances features of memory (e.g., long-term persistence) in TIL that are characteristically exhausted and senescent is not established. Here, we show that pharmacologic inhibition of Akt enables expansion of TIL with the transcriptional, metabolic, and functional properties characteristic of memory T cells. Consequently, Akt inhibition results in enhanced persistence of TIL after adoptive transfer into an immunodeficient animal model and augments antitumor immunity of CD8 T cells in a mouse model of cell-based immunotherapy. Pharmacologic inhibition of Akt represents a novel immunometabolomic approach to enhance the persistence of antitumor T cells and improve the efficacy of cell-based immunotherapy for metastatic cancer.


Nature | 2016

Ionic immune suppression within the tumour microenvironment limits T cell effector function.

Robert L. Eil; Suman K. Vodnala; David Clever; Christopher A. Klebanoff; Madhusudhanan Sukumar; Jenny H. Pan; Douglas C. Palmer; Alena Gros; Tori N. Yamamoto; Shashank J. Patel; Geoffrey Guittard; Zhiya Yu; Valentina Carbonaro; Klaus Okkenhaug; David S. Schrump; W. Marston Linehan; Rahul Roychoudhuri; Nicholas P. Restifo

Tumours progress despite being infiltrated by tumour-specific effector T cells. Tumours contain areas of cellular necrosis, which are associated with poor survival in a variety of cancers. Here, we show that necrosis releases intracellular potassium ions into the extracellular fluid of mouse and human tumours, causing profound suppression of T cell effector function. Elevation of the extracellular potassium concentration ([K+]e) impairs T cell receptor (TCR)-driven Akt–mTOR phosphorylation and effector programmes. Potassium-mediated suppression of Akt–mTOR signalling and T cell function is dependent upon the activity of the serine/threonine phosphatase PP2A. Although the suppressive effect mediated by elevated [K+]e is independent of changes in plasma membrane potential (Vm), it requires an increase in intracellular potassium ([K+]i). Accordingly, augmenting potassium efflux in tumour-specific T cells by overexpressing the potassium channel Kv1.3 lowers [K+]i and improves effector functions in vitro and in vivo and enhances tumour clearance and survival in melanoma-bearing mice. These results uncover an ionic checkpoint that blocks T cell function in tumours and identify potential new strategies for cancer immunotherapy.


Cell Metabolism | 2016

Mitochondrial Membrane Potential Identifies Cells with Enhanced Stemness for Cellular Therapy.

Madhusudhanan Sukumar; Jie Liu; Gautam U. Mehta; Shashank J. Patel; Rahul Roychoudhuri; Joseph G. Crompton; Christopher A. Klebanoff; Yun Ji; Peng Li; Zhiya Yu; Greg Whitehill; David Clever; Robert L. Eil; Douglas C. Palmer; Suman Mitra; Mahadev Rao; Keyvan Keyvanfar; David S. Schrump; Ena Wang; Francesco M. Marincola; Luca Gattinoni; Warren J. Leonard; Pawel Muranski; Toren Finkel; Nicholas P. Restifo

Long-term survival and antitumor immunity of adoptively transferred CD8(+) T cells is dependent on their metabolic fitness, but approaches to isolate therapeutic T cells based on metabolic features are not well established. Here we utilized a lipophilic cationic dye tetramethylrhodamine methyl ester (TMRM) to identify and isolate metabolically robust T cells based on their mitochondrial membrane potential (ΔΨm). Comprehensive metabolomic and gene expression profiling demonstrated global features of improved metabolic fitness in low-ΔΨm-sorted CD8(+) T cells. Transfer of these low-ΔΨm T cells was associated with superior long-term in vivo persistence and an enhanced capacity to eradicate established tumors compared with high-ΔΨm cells. Use of ΔΨm-based sorting to enrich for cells with superior metabolic features was observed in CD8(+), CD4(+) T cell subsets, and long-term hematopoietic stem cells. This metabolism-based approach to cell selection may be broadly applicable to therapies involving the transfer of HSC or lymphocytes for the treatment of viral-associated illnesses and cancer.


Journal of Clinical Investigation | 2016

Memory T cell–driven differentiation of naive cells impairs adoptive immunotherapy

Christopher A. Klebanoff; Christopher D. Scott; Anthony J. Leonardi; Tori N. Yamamoto; Anthony C. Cruz; Claudia Ouyang; Madhu Ramaswamy; Rahul Roychoudhuri; Yun Ji; Robert L. Eil; Madhusudhanan Sukumar; Joseph G. Crompton; Douglas C. Palmer; Zachary A. Borman; David Clever; Stacy K. Thomas; Shashankkumar Patel; Zhiya Yu; Pawel Muranski; Hui Liu; Ena Wang; Francesco M. Marincola; Alena Gros; Luca Gattinoni; Steven A. Rosenberg; Richard M. Siegel; Nicholas P. Restifo

Adoptive cell transfer (ACT) of purified naive, stem cell memory, and central memory T cell subsets results in superior persistence and antitumor immunity compared with ACT of populations containing more-differentiated effector memory and effector T cells. Despite a clear advantage of the less-differentiated populations, the majority of ACT trials utilize unfractionated T cell subsets. Here, we have challenged the notion that the mere presence of less-differentiated T cells in starting populations used to generate therapeutic T cells is sufficient to convey their desirable attributes. Using both mouse and human cells, we identified a T cell-T cell interaction whereby antigen-experienced subsets directly promote the phenotypic, functional, and metabolic differentiation of naive T cells. This process led to the loss of less-differentiated T cell subsets and resulted in impaired cellular persistence and tumor regression in mouse models following ACT. The T memory-induced conversion of naive T cells was mediated by a nonapoptotic Fas signal, resulting in Akt-driven cellular differentiation. Thus, induction of Fas signaling enhanced T cell differentiation and impaired antitumor immunity, while Fas signaling blockade preserved the antitumor efficacy of naive cells within mixed populations. These findings reveal that T cell subsets can synchronize their differentiation state in a process similar to quorum sensing in unicellular organisms and suggest that disruption of this quorum-like behavior among T cells has potential to enhance T cell-based immunotherapies.


Nature Immunology | 2016

BACH2 regulates CD8+ T cell differentiation by controlling access of AP-1 factors to enhancers

Rahul Roychoudhuri; David Clever; Peng Li; Yoshiyuki Wakabayashi; Kylie M. Quinn; Christopher A. Klebanoff; Yun Ji; Madhusudhanan Sukumar; Robert L. Eil; Zhiya Yu; Rosanne Spolski; Douglas C. Palmer; Jenny H. Pan; Shashank J. Patel; Derek C. Macallan; Giulia Fabozzi; Han Yu Shih; Yuka Kanno; Akihiko Muto; Jun Zhu; Luca Gattinoni; John J. O'Shea; Klaus Okkenhaug; Kazuhiko Igarashi; Warren J. Leonard; Nicholas P. Restifo

T cell antigen receptor (TCR) signaling drives distinct responses depending on the differentiation state and context of CD8+ T cells. We hypothesized that access of signal-dependent transcription factors (TFs) to enhancers is dynamically regulated to shape transcriptional responses to TCR signaling. We found that the TF BACH2 restrains terminal differentiation to enable generation of long-lived memory cells and protective immunity after viral infection. BACH2 was recruited to enhancers, where it limited expression of TCR-driven genes by attenuating the availability of activator protein-1 (AP-1) sites to Jun family signal-dependent TFs. In naive cells, this prevented TCR-driven induction of genes associated with terminal differentiation. Upon effector differentiation, reduced expression of BACH2 and its phosphorylation enabled unrestrained induction of TCR-driven effector programs.


Proceedings of the National Academy of Sciences of the United States of America | 2015

miR-155 augments CD8+ T-cell antitumor activity in lymphoreplete hosts by enhancing responsiveness to homeostatic γc cytokines

Yun Ji; Claudia Wrzesinski; Zhiya Yu; Jinhui Hu; Sanjivan Gautam; Nga V. Hawk; William G. Telford; Douglas C. Palmer; Zulmarie Franco; Madhusudhanan Sukumar; Rahul Roychoudhuri; David Clever; Christopher A. Klebanoff; Charles D. Surh; Thomas A. Waldmann; Nicholas P. Restifo; Luca Gattinoni

Significance We describe here a strategy based on microRNA therapeutics to augment the efficacy of T-cell–based therapies without the requirement of toxic maneuvers such as lymphodepletion preconditioning and the administration of high doses of exogenous cytokines. These findings can lead to the development of safer and more effective T-cell–based therapies for the treatment of patients with advanced cancer. Lymphodepleting regimens are used before adoptive immunotherapy to augment the antitumor efficacy of transferred T cells by removing endogenous homeostatic “cytokine sinks.” These conditioning modalities, however, are often associated with severe toxicities. We found that microRNA-155 (miR-155) enabled tumor-specific CD8+ T cells to mediate profound antitumor responses in lymphoreplete hosts that were not potentiated by immune-ablation. miR-155 enhanced T-cell responsiveness to limited amounts of homeostatic γc cytokines, resulting in delayed cellular contraction and sustained cytokine production. miR-155 restrained the expression of the inositol 5-phosphatase Ship1, an inhibitor of the serine-threonine protein kinase Akt, and multiple negative regulators of signal transducer and activator of transcription 5 (Stat5), including suppressor of cytokine signaling 1 (Socs1) and the protein tyrosine phosphatase Ptpn2. Expression of constitutively active Stat5a recapitulated the survival advantages conferred by miR-155, whereas constitutive Akt activation promoted sustained effector functions. Our results indicate that overexpression of miR-155 in tumor-specific T cells can be used to increase the effectiveness of adoptive immunotherapies in a cell-intrinsic manner without the need for life-threatening, lymphodepleting maneuvers.


Journal of Experimental Medicine | 2015

Cish actively silences TCR signaling in CD8+ T cells to maintain tumor tolerance

Douglas C. Palmer; Geoffrey Guittard; Zulmarie Franco; Joseph G. Crompton; Robert L. Eil; Shashank J. Patel; Yun Ji; Nicholas van Panhuys; Christopher A. Klebanoff; Madhusudhanan Sukumar; David Clever; Anna Chichura; Rahul Roychoudhuri; Rajat Varma; Ena Wang; Luca Gattinoni; Francesco M. Marincola; Lakshmi Balagopalan; Lawrence E. Samelson; Nicholas P. Restifo

Palmer et al. find that Cish, a member of the SOCS family, is induced by TCR stimulation in CD8+ T cells and inhibits their functional avidity against tumor. The authors uncover a novel mechanism of suppression for a SOCS member.


Trends in Immunology | 2014

Reprogramming antitumor immunity

Joseph G. Crompton; David Clever; Raul Vizcardo; Mahendra S. Rao; Nicholas P. Restifo

Regenerative medicine holds great promise in replacing tissues and organs lost to degenerative disease and injury. Application of the principles of cellular reprogramming for the treatment of cancer, however, is not well established. Here, we present an overview of cellular reprogramming techniques used in regenerative medicine, and within this context, envision how the scope of regenerative medicine may be expanded to treat metastatic cancer by revitalizing an exhausted and senescent immune system.


Cancer immunology research | 2015

Type I cytokines synergize with oncogene inhibition to induce tumor growth arrest

Nicolas Acquavella; David Clever; Zhiya Yu; Melody E. Roelke-Parker; Douglas C. Palmer; Liqiang Xi; Holger Pflicke; Yun Ji; Alena Gros; Ken Ichi Hanada; Ian S. Goldlust; Gautam U. Mehta; Christopher A. Klebanoff; Joseph G. Crompton; Madhusudhanan Sukumar; James J. Morrow; Zulmarie Franco; Luca Gattinoni; Hui Liu; Ena Wang; Francesco M. Marincola; David F. Stroncek; Chyi Chia R Lee; Mark Raffeld; Marcus Bosenberg; Rahul Roychoudhuri; Nicholas P. Restifo

Acquavella, Clever, and colleagues show that IFNγ and TNFα synergize with vemurafenib to induce tumor growth arrest, supporting further study of the intersection between immunologic and oncogenic signaling in cancer cells and of treatment strategies combining vemurafenib and T-cell–based immunotherapy. Both targeted inhibition of oncogenic driver mutations and immune-based therapies show efficacy in treatment of patients with metastatic cancer, but responses can be either short lived or incompletely effective. Oncogene inhibition can augment the efficacy of immune-based therapy, but mechanisms by which these two interventions might cooperate are incompletely resolved. Using a novel transplantable BRAFV600E-mutant murine melanoma model (SB-3123), we explored potential mechanisms of synergy between the selective BRAFV600E inhibitor vemurafenib and adoptive cell transfer (ACT)–based immunotherapy. We found that vemurafenib cooperated with ACT to delay melanoma progression without significantly affecting tumor infiltration or effector function of endogenous or adoptively transferred CD8+ T cells, as previously observed. Instead, we found that the T-cell cytokines IFNγ and TNFα synergized with vemurafenib to induce cell-cycle arrest of tumor cells in vitro. This combinatorial effect was recapitulated in human melanoma–derived cell lines and was restricted to cancers bearing a BRAFV600E mutation. Molecular profiling of treated SB-3123 indicated that the provision of vemurafenib promoted the sensitization of SB-3123 to the antiproliferative effects of T-cell effector cytokines. The unexpected finding that immune cytokines synergize with oncogene inhibitors to induce growth arrest has major implications for understanding cancer biology at the intersection of oncogenic and immune signaling and provides a basis for design of combinatorial therapeutic approaches for patients with metastatic cancer. Cancer Immunol Res; 3(1); 37–47. ©2014 AACR. See related commentary by Riddell, p. 23

Collaboration


Dive into the David Clever's collaboration.

Top Co-Authors

Avatar

Nicholas P. Restifo

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
Top Co-Authors

Avatar

Robert L. Eil

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

Madhusudhanan Sukumar

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

Shashank J. Patel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge