Network


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

Hotspot


Dive into the research topics where Todd D. Prickett is active.

Publication


Featured researches published by Todd D. Prickett.


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 Genetics | 2009

Analysis of the tyrosine kinome in melanoma reveals recurrent mutations in ERBB4

Todd D. Prickett; Neena S Agrawal; Xiaomu Wei; Kristin E Yates; Jimmy Lin; John R. Wunderlich; Julia C. Cronin; Pedro Cruz; Nisc Comparative Sequencing Program; Steven A. Rosenberg; Yardena Samuels

Tyrosine phosphorylation is important in signaling pathways underlying tumorigenesis. We performed a mutational analysis of the protein tyrosine kinase (PTK) gene family in cutaneous metastatic melanoma. We identified 30 somatic mutations affecting the kinase domains of 19 PTKs and subsequently evaluated the entire coding regions of the genes encoding these 19 PTKs for somatic mutations in 79 melanoma samples. We found ERBB4 mutations in 19% of individuals with melanoma and found mutations in two other kinases (FLT1 and PTK2B) in 10% of individuals with melanomas. We examined seven missense mutations in the most commonly altered PTK gene, ERBB4, and found that they resulted in increased kinase activity and transformation ability. Melanoma cells expressing mutant ERBB4 had reduced cell growth after shRNA-mediated knockdown of ERBB4 or treatment with the ERBB inhibitor lapatinib. These studies could lead to personalized therapeutics specifically targeting the kinases that are mutationally altered in individual melanomas.


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.


Nature Genetics | 2009

Analysis of the matrix metalloproteinase family reveals that MMP8 is often mutated in melanoma

Lavanya H. Palavalli; Todd D. Prickett; John R. Wunderlich; Xiaomu Wei; Allison S. Burrell; Patricia Porter-Gill; Sean Davis; Chenwei Wang; Julia C. Cronin; Neena S Agrawal; Jimmy Lin; Wendy Westbroek; Shelley Hoogstraten-Miller; Alfredo A. Molinolo; Patricia Fetsch; Armando C. Filie; Michael P. O'Connell; Carolyn E. Banister; Jason Howard; Phillip Buckhaults; Ashani T. Weeraratna; Lawrence C. Brody; Steven A. Rosenberg; Yardena Samuels

A mutational analysis of the matrix metalloproteinase (MMP) gene family in human melanoma identified somatic mutations in 23% of melanomas. Five mutations in one of the most commonly mutated genes, MMP8, reduced MMP enzyme activity. Expression of wild-type but not mutant MMP8 in human melanoma cells inhibited growth on soft agar in vitro and tumor formation in vivo, suggesting that wild-type MMP-8 has the ability to inhibit melanoma progression.


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.


Pigment Cell & Melanoma Research | 2009

Frequent Mutations in the MITF Pathway in Melanoma

Julia C. Cronin; John R. Wunderlich; Stacie K. Loftus; Todd D. Prickett; Xiaomu Wei; Katie Ridd; Swapna Vemula; Allison S. Burrell; Neena S Agrawal; Jimmy Lin; Carolyn E. Banister; Phillip Buckhaults; Steven A. Rosenberg; Boris C. Bastian; William J. Pavan; Yardena Samuels

Microphthalmia‐associated transcription factor (MITF) is involved in melanocyte cell development, pigmentation and neoplasia. To determine whether MITF is somatically mutated in melanoma, we compared the sequence of MITF from primary and metastatic lesions to patient‐matched normal DNA. In the 50 metastatic melanoma tumor lines analysed, we discovered four samples that had genomic amplifications of MITF and four that had MITF mutations in the regions encoding the transactivation, DNA binding or basic, helix‐loop‐helix domains. Sequence analysis for SOX10, a transcription factor, which both acts upstream of MITF and synergizes with MITF, identified an additional three samples with frameshift or nonsense mutations. Microphthalmia‐associated transcription factor and SOX10 were found to be mutated in a mutually exclusive fashion, possibly suggesting disruption in a common genetic pathway. Taken together we found that over 20% of the metastatic melanoma cases had alterations in the MITF pathway. We show that the MITF pathway is also altered in primary melanomas: 2/26 demonstrated mutations in MITF and 6/55 demonstrated mutations in SOX10. Our findings suggest that altered MITF function during melanomagenesis can be achieved by MITF amplification, MITF single base substitutions or by mutation of its regulator SOX10.


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.


Clinical Cancer Research | 2012

Molecular Pathways: Dysregulated Glutamatergic Signaling Pathways in Cancer

Todd D. Prickett; Yardena Samuels

The neurotransmitter glutamate interacts with glutamate receptor proteins, leading to the activation of multiple signaling pathways. Dysfunction in the glutamatergic signaling pathway is well established as a frequent player in diseases such as schizophrenia, Alzheimer disease, and brain tumors (gliomas). Recently, aberrant functioning of this pathway has also been shown in melanoma. In both glioma and melanoma, glutamate secretion stimulates tumor growth, proliferation, and survival through activation of the mitogen-activated protein kinase and phosphoinositide 3-kinase/Akt pathways. In the future, extracellular glutamate levels and glutamatergic signaling may serve as biological markers for tumorigenicity and facilitate targeted therapy for melanoma. Clin Cancer Res; 18(16); 4240–6. ©2012 AACR.


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 Todd D. Prickett's collaboration.

Top Co-Authors

Avatar

Steven A. Rosenberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jared J. Gartner

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

Maria R. Parkhurst

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xiaomu Wei

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John R. Wunderlich

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yong-Chen Lu

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge