Network


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

Hotspot


Dive into the research topics where Robert L. Eil is active.

Publication


Featured researches published by Robert L. Eil.


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 | 2017

Identification of essential genes for cancer immunotherapy.

Shashank J. Patel; Neville E. Sanjana; Rigel J. Kishton; Arash Eidizadeh; Suman K. Vodnala; Maggie Cam; Jared J. Gartner; Li Jia; Seth M. Steinberg; Tori N. Yamamoto; Anand Merchant; Gautam U. Mehta; Anna Chichura; Ophir Shalem; Eric Tran; Robert L. Eil; Madhusudhanan Sukumar; Eva Perez Guijarro; Chi-Ping Day; Paul D. Robbins; Steve Feldman; Glenn Merlino; Feng Zhang; Nicholas P. Restifo

Somatic gene mutations can alter the vulnerability of cancer cells to T-cell-based immunotherapies. Here we perturbed genes in human melanoma cells to mimic loss-of-function mutations involved in resistance to these therapies, by using a genome-scale CRISPR–Cas9 library that consisted of around 123,000 single-guide RNAs, and profiled genes whose loss in tumour cells impaired the effector function of CD8+ T cells. The genes that were most enriched in the screen have key roles in antigen presentation and interferon-γ signalling, and correlate with cytolytic activity in patient tumours from The Cancer Genome Atlas. Among the genes validated using different cancer cell lines and antigens, we identified multiple loss-of-function mutations in APLNR, encoding the apelin receptor, in patient tumours that were refractory to immunotherapy. We show that APLNR interacts with JAK1, modulating interferon-γ responses in tumours, and that its functional loss reduces the efficacy of adoptive cell transfer and checkpoint blockade immunotherapies in mouse models. Our results link the loss of essential genes for the effector function of CD8+ T cells with the resistance or non-responsiveness of cancer to immunotherapies.


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.


Current Opinion in Immunology | 2015

The interplay of effector and regulatory T cells in cancer

Rahul Roychoudhuri; Robert L. Eil; Nicholas P. Restifo

Regulatory T (Treg) cells suppress effector T (Teff) cells and prevent immune-mediated rejection of cancer. Much less appreciated are mechanisms by which Teff cells antagonize Treg cells. Herein, we consider how complex reciprocal interactions between Teff and Treg cells shape their population dynamics within tumors. Under states of tolerance, including during tumor escape, suppressed Teff cells support Treg cell populations through antigen-dependent provision of interleukin (IL)-2. During immune activation, Teff cells can lose this supportive capacity and directly antagonize Treg cell populations to neutralize their immunosuppressive function. While this latter state is rarely achieved spontaneously within tumors, we propose that therapeutic induction of immune activation has the potential to stably disrupt immunosuppressive population states resulting in durable cancer regression.


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.


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.


Cancer | 2014

Nomogram for predicting the benefit of neoadjuvant chemoradiotherapy for patients with esophageal cancer: A SEER‐Medicare analysis

Robert L. Eil; Brian S. Diggs; Samuel J. Wang; James P. Dolan; John G. Hunter; Charles R. Thomas

The survival impact of neoadjuvant chemoradiotherapy (CRT) on esophageal cancer remains difficult to establish for specific patients. The aim of the current study was to create a Web‐based prediction tool providing individualized survival projections based on tumor and treatment data.


Journal of Clinical Investigation | 2016

The transcription factor BACH2 promotes tumor immunosuppression

Rahul Roychoudhuri; Robert L. Eil; David Clever; Christopher A. Klebanoff; Madhusudhanan Sukumar; Francis M. Grant; Zhiya Yu; Gautam U. Mehta; Hui Liu; Ping Jin; Yun Ji; Douglas C. Palmer; Jenny H. Pan; Anna Chichura; Joseph G. Crompton; Shashank J. Patel; David F. Stroncek; Ena Wang; Francesco M. Marincola; Klaus Okkenhaug; Luca Gattinoni; Nicholas P. Restifo

The immune system has a powerful ability to recognize and kill cancer cells, but its function is often suppressed within tumors, preventing clearance of disease. Functionally diverse innate and adaptive cellular lineages either drive or constrain immune reactions within tumors. The transcription factor (TF) BACH2 regulates the differentiation of multiple innate and adaptive cellular lineages, but its role in controlling tumor immunity has not been elucidated. Here, we demonstrate that BACH2 is required to establish immunosuppression within tumors. Tumor growth was markedly impaired in Bach2-deficient mice and coincided with intratumoral activation of both innate and adaptive immunity. However, augmented tumor clearance in the absence of Bach2 was dependent upon the adaptive immune system. Analysis of tumor-infiltrating lymphocytes from Bach2-deficient mice revealed high frequencies of rapidly proliferating effector CD4+ and CD8+ T cells that expressed the inflammatory cytokine IFN-γ. Effector T cell activation coincided with a reduction in the frequency of intratumoral Foxp3+ Tregs. Mechanistically, BACH2 promoted tumor immunosuppression through Treg-mediated inhibition of intratumoral CD8+ T cells and IFN-γ. These findings demonstrate that BACH2 is a key component of the molecular program of tumor immunosuppression and identify therapeutic targets for the reversal of immunosuppression in cancer.

Collaboration


Dive into the Robert L. Eil's collaboration.

Top Co-Authors

Avatar

Nicholas P. Restifo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rahul Roychoudhuri

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

Shashank J. Patel

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

Jenny H. Pan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Luca Gattinoni

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge