Network


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

Hotspot


Dive into the research topics where Jared J. Gartner is active.

Publication


Featured researches published by Jared J. Gartner.


Nature Medicine | 2013

Mining Exomic Sequencing Data to Identify Mutated Antigens Recognized by Adoptively Transferred Tumor-reactive T cells

Paul F. Robbins; Yong-Chen Lu; Mona El-Gamil; Yong F. Li; Colin Gross; Jared J. Gartner; Jimmy C. Lin; Jamie K. Teer; Paul F. Cliften; Eric Tycksen; Yardena Samuels; Steven A. Rosenberg

Substantial regressions of metastatic lesions have been observed in up to 70% of patients with melanoma who received adoptively transferred autologous tumor-infiltrating lymphocytes (TILs) in phase 2 clinical trials. In addition, 40% of patients treated in a recent trial experienced complete regressions of all measurable lesions for at least 5 years following TIL treatment. To evaluate the potential association between the ability of TILs to mediate durable regressions and their ability to recognize potent antigens that presumably include mutated gene products, we developed a new screening approach involving mining whole-exome sequence data to identify mutated proteins expressed in patient tumors. We then synthesized and evaluated candidate mutated T cell epitopes that were identified using a major histocompatibility complex–binding algorithm for recognition by TILs. Using this approach, we identified mutated antigens expressed on autologous tumor cells that were recognized by three bulk TIL lines from three individuals with melanoma that were associated with objective tumor regressions following adoptive transfer. This simplified approach for identifying mutated antigens recognized by T cells avoids the need to generate and laboriously screen cDNA libraries from tumors and may represent a generally applicable method for identifying mutated antigens expressed in a variety of tumor types.


Nature Genetics | 2011

Exome sequencing identifies GRIN2A as frequently mutated in melanoma

Xiaomu Wei; Vijay Walia; Jimmy Lin; Jamie K. Teer; Todd D. Prickett; Jared J. Gartner; Sean Davis; Katherine Stemke-Hale; Michael A. Davies; Jeffrey E. Gershenwald; William H. Robinson; Steven E. Robinson; Steven A. Rosenberg; Yardena Samuels

The incidence of melanoma is increasing more than any other cancer, and knowledge of its genetic alterations is limited. To systematically analyze such alterations, we performed whole-exome sequencing of 14 matched normal and metastatic tumor DNAs. Using stringent criteria, we identified 68 genes that appeared to be somatically mutated at elevated frequency, many of which are not known to be genetically altered in tumors. Most importantly, we discovered that TRRAP harbored a recurrent mutation that clustered in one position (p. Ser722Phe) in 6 out of 167 affected individuals (∼4%), as well as a previously unidentified gene, GRIN2A, which was mutated in 33% of melanoma samples. The nature, pattern and functional evaluation of the TRRAP recurrent mutation suggest that TRRAP functions as an oncogene. Our study provides, to our knowledge, the most comprehensive map of genetic alterations in melanoma to date and suggests that the glutamate signaling pathway is involved in this disease.


Nature Medicine | 2016

Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients

Alena Gros; Maria R. Parkhurst; Eric Tran; Anna Pasetto; Paul F. Robbins; Sadia Ilyas; Todd D. Prickett; Jared J. Gartner; Jessica S Crystal; Ilana M. Roberts; Kasia Trebska-Mcgowan; John R. Wunderlich; James Chih-Hsin Yang; Steven A. Rosenberg

Detection of lymphocytes that target tumor-specific mutant neoantigens—derived from products encoded by mutated genes in the tumor—is mostly limited to tumor-resident lymphocytes, but whether these lymphocytes often occur in the circulation is unclear. We recently reported that intratumoral expression of the programmed cell death 1 (PD-1) receptor can guide the identification of the patient-specific repertoire of tumor-reactive CD8+ lymphocytes that reside in the tumor. In view of these findings, we investigated whether PD-1 expression on peripheral blood lymphocytes could be used as a biomarker to detect T cells that target neoantigens. By using a high-throughput personalized screening approach, we identified neoantigen-specific lymphocytes in the peripheral blood of three of four melanoma patients. Despite their low frequency in the circulation, we found that CD8+PD-1+, but not CD8+PD-1−, cell populations had lymphocytes that targeted 3, 3 and 1 unique, patient-specific neoantigens, respectively. We show that neoantigen-specific T cells and gene-engineered lymphocytes expressing neoantigen-specific T cell receptors (TCRs) isolated from peripheral blood recognized autologous tumors. Notably, the tumor-antigen specificities and TCR repertoires of the circulating and tumor-infiltrating CD8+PD-1+ cells appeared similar, implying that the circulating CD8+PD-1+ lymphocytes could provide a window into the tumor-resident antitumor lymphocytes. Thus, expression of PD-1 identifies a diverse and patient-specific antitumor T cell response in peripheral blood, providing a novel noninvasive strategy to develop personalized therapies using neoantigen-reactive lymphocytes or TCRs to treat cancer.


Science | 2015

Immunogenicity of somatic mutations in human gastrointestinal cancers

Eric Tran; Mojgan Ahmadzadeh; Yong-Chen Lu; Alena Gros; Paul F. Robbins; Jared J. Gartner; Zhili Zheng; Yong F. Li; Satyajit Ray; John R. Wunderlich; Robert Somerville; Steven A. Rosenberg

Low mutation rate okay for T cells Cancers that tend to have high numbers of mutations, such as melanoma and smoking-induced lung cancer, respond well to immunotherapies, whereas those with fewer mutations, such as pancreatic cancer, do not. Tran et al. searched for tumor mutation–reactive T cells in 10 patients with metastatic gastrointestinal cancers, which have relatively low mutation burdens, and discovered that 9 out of 10 harbored such cells. T cells from one patient recognized a mutation common to many types of cancers. Engineering T cells to express this particular mutation-reactive T cell receptor may extend adoptive cell immunotherapy to a larger pool of patients than previously anticipated. Science, this issue p. 1387 Individuals with cancers that have low mutation frequencies often harbor mutation-reactive T cells. It is unknown whether the human immune system frequently mounts a T cell response against mutations expressed by common epithelial cancers. Using a next-generation sequencing approach combined with high-throughput immunologic screening, we demonstrated that tumor-infiltrating lymphocytes (TILs) from 9 out of 10 patients with metastatic gastrointestinal cancers contained CD4+ and/or CD8+ T cells that recognized one to three neo-epitopes derived from somatic mutations expressed by the patient’s own tumor. There were no immunogenic epitopes shared between these patients. However, we identified in one patient a human leukocyte antigen–C*08:02–restricted T cell receptor from CD8+ TILs that targeted the KRASG12D hotspot driver mutation found in many human cancers. Thus, a high frequency of patients with common gastrointestinal cancers harbor immunogenic mutations that can potentially be exploited for the development of highly personalized immunotherapies.


Nature Genetics | 2011

Exon capture analysis of G protein-coupled receptors identifies activating mutations in GRM3 in melanoma.

Todd D. Prickett; Xiaomu Wei; Isabel Cardenas-Navia; Jamie K. Teer; Jimmy Lin; Vijay Walia; Jared J. Gartner; Jiji Jiang; Praveen F. Cherukuri; Alfredo A. Molinolo; Michael A. Davies; Jeffrey E. Gershenwald; Katherine Stemke-Hale; Steven A. Rosenberg; Elliott H. Margulies; Yardena Samuels

G protein-coupled receptors (GPCRs), the largest human gene family, are important regulators of signaling pathways. However, knowledge of their genetic alterations is limited. In this study, we used exon capture and massively parallel sequencing methods to analyze the mutational status of 734 GPCRs in melanoma. This investigation revealed that one family member, GRM3, was frequently mutated and that one of its mutations clustered within one position. Biochemical analysis of GRM3 alterations revealed that mutant GRM3 selectively regulated the phosphorylation of MEK, leading to increased anchorage-independent growth and migration. Melanoma cells expressing mutant GRM3 had reduced cell growth and cellular migration after short hairpin RNA–mediated knockdown of GRM3 or treatment with a selective MEK inhibitor, AZD-6244, which is currently being used in phase 2 clinical trials. Our study yields the most comprehensive map of genetic alterations in the GPCR gene family.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Whole-genome sequencing identifies a recurrent functional synonymous mutation in melanoma

Jared J. Gartner; Stephen C. J. Parker; Todd D. Prickett; Ken Dutton-Regester; Michael L. Stitzel; Jimmy C. Lin; Sean Davis; Vijaya L. Simhadri; Sujata Jha; Nobuko Katagiri; Valer Gotea; Jamie K. Teer; Xiaomu Wei; Mario A. Morken; Umesh Bhanot; Guo Chen; Laura Elnitski; Michael A. Davies; Jeffrey E. Gershenwald; Hannah Carter; Rachel Karchin; William H. Robinson; Steven E. Robinson; Steven A. Rosenberg; Francis S. Collins; Giovanni Parmigiani; Anton A. Komar; Chava Kimchi-Sarfaty; Nicholas K. Hayward; Elliott H. Margulies

Synonymous mutations, which do not alter the protein sequence, have been shown to affect protein function [Sauna ZE, Kimchi-Sarfaty C (2011) Nat Rev Genet 12(10):683–691]. However, synonymous mutations are rarely investigated in the cancer genomics field. We used whole-genome and -exome sequencing to identify somatic mutations in 29 melanoma samples. Validation of one synonymous somatic mutation in BCL2L12 in 285 samples identified 12 cases that harbored the recurrent F17F mutation. This mutation led to increased BCL2L12 mRNA and protein levels because of differential targeting of WT and mutant BCL2L12 by hsa-miR-671–5p. Protein made from mutant BCL2L12 transcript bound p53, inhibited UV-induced apoptosis more efficiently than WT BCL2L12, and reduced endogenous p53 target gene transcription. This report shows selection of a recurrent somatic synonymous mutation in cancer. Our data indicate that silent alterations have a role to play in human cancer, emphasizing the importance of their investigation in future cancer genome studies.


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.


Annual Review of Genomics and Human Genetics | 2013

The Genetics of Melanoma: Recent Advances ∗

Victoria Hill; Jared J. Gartner; Yardena Samuels; Alisa M. Goldstein

Cutaneous malignant melanoma results from the interplay of genetic, host, and environmental factors. Genetic factors implicated in melanoma etiology include inherited high-, intermediate-, and low-risk susceptibility genes as well as numerous somatic mutations in melanoma tumors. CDKN2A is the major high-risk melanoma susceptibility gene identified to date. Recent identification of low-risk loci has been accomplished predominantly through genome-wide association studies. Whole-exome and whole-genome studies have identified numerous genes somatically altered in melanoma tumors and highlighted a higher mutation load in melanoma tumors compared with those in other cancers. This higher load is believed to be attributable to the preponderance of cytosine-to-thymine nucleotide substitutions as a result of UV radiation exposure. Technological advances, particularly next-generation sequencing, have increased the opportunities for germline and somatic gene discovery in melanoma and are opening up new avenues for understanding melanoma pathogenesis as well as leading to new opportunities for treatment.


Science | 2017

Landscape of immunogenic tumor antigens in successful immunotherapy of virally induced epithelial cancer

Sanja Stevanovic; Anna Pasetto; Sarah R. Helman; Jared J. Gartner; Todd D. Prickett; Bryan Howie; Harlan Robins; Paul F. Robbins; Christopher A. Klebanoff; Steven A. Rosenberg; Christian S. Hinrichs

Targeting nonviral antigens in viral-driven cancer Adoptive cell transfer harnesses a patients own T cells to destroy cancer. The strategy can successfully treat epithelial tumors driven by human papillomavirus (HPV), but it remains unclear why only some patients respond. Stevanović et al. examined the antitumor T cell response associated with HPV+ cervical cancers that underwent complete regression. Unexpectedly, reactive T cells were not directed against virally associated antigens, but rather against cancer germline antigens or neoantigens not previously recognized by the immune system. These findings counter the widely held belief that T cell responses against viral antigens are responsible for therapeutic effects in HPV-driven cancers. Science, this issue p. 200 Reactive T cells directed against nonviral antigens lead to regression of human papilloma virus–positive cervical cancer. Immunotherapy has clinical activity in certain virally associated cancers. However, the tumor antigens targeted in successful treatments remain poorly defined. We used a personalized immunogenomic approach to elucidate the global landscape of antitumor T cell responses in complete regression of human papillomavirus–associated metastatic cervical cancer after tumor-infiltrating adoptive T cell therapy. Remarkably, immunodominant T cell reactivities were directed against mutated neoantigens or a cancer germline antigen, rather than canonical viral antigens. T cells targeting viral tumor antigens did not display preferential in vivo expansion. Both viral and nonviral tumor antigen–specific T cells resided predominantly in the programmed cell death 1 (PD-1)–expressing T cell compartment, which suggests that PD-1 blockade may unleash diverse antitumor T cell reactivities. These findings suggest a new paradigm of targeting nonviral antigens in immunotherapy of virally associated cancers.


Cancer immunology research | 2016

Tumor- and neoantigen-reactive T-cell receptors can be identified based on their frequency in fresh tumor

Anna Pasetto; Alena Gros; Paul F. Robbins; Drew C. Deniger; Todd D. Prickett; Rodrigo Matus-Nicodemos; Bryan Howie; Harlan Robins; Maria R. Parkhurst; Jared J. Gartner; Katarzyna Trebska-McGowan; Jessica S. Crystal; Steven A. Rosenberg

Effective adoptive T-cell therapy requires multiple tumor-epitope reactive T-cell clones. Fresh TILs were found to frequently contain such cells. Their TCRs were rapidly isolated based only on their frequency and could be used for personalized TCR-gene therapy. Adoptive transfer of T cells with engineered T-cell receptor (TCR) genes that target tumor-specific antigens can mediate cancer regression. Accumulating evidence suggests that the clinical success of many immunotherapies is mediated by T cells targeting mutated neoantigens unique to the patient. We hypothesized that the most frequent TCR clonotypes infiltrating the tumor were reactive against tumor antigens. To test this hypothesis, we developed a multistep strategy that involved TCRB deep sequencing of the CD8+PD-1+ T-cell subset, matching of TCRA–TCRB pairs by pairSEQ and single-cell RT-PCR, followed by testing of the TCRs for tumor-antigen specificity. Analysis of 12 fresh metastatic melanomas revealed that in 11 samples, up to 5 tumor-reactive TCRs were present in the 5 most frequently occurring clonotypes, which included reactivity against neoantigens. These data show the feasibility of developing a rapid, personalized TCR-gene therapy approach that targets the unique set of antigens presented by the autologous tumor without the need to identify their immunologic reactivity. Cancer Immunol Res; 4(9); 734–43. ©2016 AACR.

Collaboration


Dive into the Jared J. Gartner's collaboration.

Top Co-Authors

Avatar

Steven A. Rosenberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Todd D. Prickett

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Paul F. Robbins

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yardena Samuels

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Alena Gros

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yong-Chen Lu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Maria R. Parkhurst

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Eric Tran

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John R. Wunderlich

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert Somerville

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge