Try a new search

Format these results:

Searched for:

person:snydea06

in-biosketch:yes

Total Results:

35


Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses

Chiappinelli, Katherine B; Strissel, Pamela L; Desrichard, Alexis; Li, Huili; Henke, Christine; Akman, Benjamin; Hein, Alexander; Rote, Neal S; Cope, Leslie M; Snyder, Alexandra; Makarov, Vladimir; Budhu, Sadna; Slamon, Dennis J; Wolchok, Jedd D; Pardoll, Drew M; Beckmann, Matthias W; Zahnow, Cynthia A; Merghoub, Taha; Chan, Timothy A; Baylin, Stephen B; Strick, Reiner
We show that DNA methyltransferase inhibitors (DNMTis) upregulate immune signaling in cancer through the viral defense pathway. In ovarian cancer (OC), DNMTis trigger cytosolic sensing of double-stranded RNA (dsRNA) causing a type I interferon response and apoptosis. Knocking down dsRNA sensors TLR3 and MAVS reduces this response 2-fold and blocking interferon beta or its receptor abrogates it. Upregulation of hypermethylated endogenous retrovirus (ERV) genes accompanies the response and ERV overexpression activates the response. Basal levels of ERV and viral defense gene expression significantly correlate in primary OC and the latter signature separates primary samples for multiple tumor types from The Cancer Genome Atlas into low versus high expression groups. In melanoma patients treated with an immune checkpoint therapy, high viral defense signature expression in tumors significantly associates with durable clinical response and DNMTi treatment sensitizes to anti-CTLA4 therapy in a pre-clinical melanoma model.
PMCID:4556003
PMID: 26317466
ISSN: 1097-4172
CID: 2200642

Toward understanding and exploiting tumor heterogeneity

Alizadeh, Ash A; Aranda, Victoria; Bardelli, Alberto; Blanpain, Cedric; Bock, Christoph; Borowski, Christine; Caldas, Carlos; Califano, Andrea; Doherty, Michael; Elsner, Markus; Esteller, Manel; Fitzgerald, Rebecca; Korbel, Jan O; Lichter, Peter; Mason, Christopher E; Navin, Nicholas; Pe'er, Dana; Polyak, Kornelia; Roberts, Charles W M; Siu, Lillian; Snyder, Alexandra; Stower, Hannah; Swanton, Charles; Verhaak, Roel G W; Zenklusen, Jean C; Zuber, Johannes; Zucman-Rossi, Jessica
The extent of tumor heterogeneity is an emerging theme that researchers are only beginning to understand. How genetic and epigenetic heterogeneity affects tumor evolution and clinical progression is unknown. The precise nature of the environmental factors that influence this heterogeneity is also yet to be characterized. Nature Medicine, Nature Biotechnology and the Volkswagen Foundation organized a meeting focused on identifying the obstacles that need to be overcome to advance translational research in and tumor heterogeneity. Once these key questions were established, the attendees devised potential solutions. Their ideas are presented here.
PMCID:4785013
PMID: 26248267
ISSN: 1546-170x
CID: 4187242

Liver-directed conversion therapy in metastatic colon cancer

Snyder, Alexandra; Kemeny, Nancy; Shamseddine, Ali; Al-Olayan, Ashwaq; El-Merhi, Fadi; Kelsen, David P; Jamali, Faek; Sidani, Mustafa; Mukherji, Deborah; El-Naghy, Mahmoud; O'Reilly, Eileen M; Saltz, Leonard B; Abou-Alfa, Ghassan K
PMCID:4397241
PMID: 26029460
ISSN: 2078-6891
CID: 4187232

RECK controls breast cancer metastasis by modulating a convergent, STAT3-dependent neoangiogenic switch

Walsh, L A; Roy, D M; Reyngold, M; Giri, D; Snyder, A; Turcan, S; Badwe, C R; Lyman, J; Bromberg, J; King, T A; Chan, T A
Metastasis is the primary cause of cancer-related death in oncology patients. A comprehensive understanding of the molecular mechanisms that cancer cells usurp to promote metastatic dissemination is critical for the development and implementation of novel diagnostic and treatment strategies. Here we show that the membrane protein RECK (Reversion-inducing cysteine-rich protein with kazal motifs) controls breast cancer metastasis by modulating a novel, non-canonical and convergent signal transducer and activator of transcription factor 3 (STAT3)-dependent angiogenic program. Neoangiogenesis and STAT3 hyperactivation are known to be fundamentally important for metastasis, but the root molecular initiators of these phenotypes are poorly understood. Our study identifies loss of RECK as a critical and previously unknown trigger for these hallmarks of metastasis. Using multiple xenograft mouse models, we comprehensively show that RECK inhibits metastasis, concomitant with a suppression of neoangiogenesis at secondary sites, while leaving primary tumor growth unaffected. Further, with functional genomics and biochemical dissection we demonstrate that RECK controls this angiogenic rheostat through a novel complex with cell surface receptors to regulate STAT3 activation, cytokine signaling, and the induction of both vascular endothelial growth factor and urokinase plasminogen activator. In accordance with these findings, inhibition of STAT3 can rescue this phenotype both in vitro and in vivo. Taken together, our study uncovers, for the first time, that RECK is a novel regulator of multiple well-established and robust mediators of metastasis; thus, RECK is a keystone protein that may be exploited in a clinical setting to target metastatic disease from multiple angles.
PMCID:4344427
PMID: 24931164
ISSN: 1476-5594
CID: 4187222

Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer

Rizvi, Naiyer A; Hellmann, Matthew D; Snyder, Alexandra; Kvistborg, Pia; Makarov, Vladimir; Havel, Jonathan J; Lee, William; Yuan, Jianda; Wong, Phillip; Ho, Teresa S; Miller, Martin L; Rekhtman, Natasha; Moreira, Andre L; Ibrahim, Fawzia; Bruggeman, Cameron; Gasmi, Billel; Zappasodi, Roberta; Maeda, Yuka; Sander, Chris; Garon, Edward B; Merghoub, Taha; Wolchok, Jedd D; Schumacher, Ton N; Chan, Timothy A
Immune checkpoint inhibitors, which unleash a patient's own T cells to kill tumors, are revolutionizing cancer treatment. To unravel the genomic determinants of response to this therapy, we used whole-exome sequencing of non-small cell lung cancers treated with pembrolizumab, an antibody targeting programmed cell death-1 (PD-1). In two independent cohorts, higher nonsynonymous mutation burden in tumors was associated with improved objective response, durable clinical benefit, and progression-free survival. Efficacy also correlated with the molecular smoking signature, higher neoantigen burden, and DNA repair pathway mutations; each factor was also associated with mutation burden. In one responder, neoantigen-specific CD8+ T cell responses paralleled tumor regression, suggesting that anti-PD-1 therapy enhances neoantigen-specific T cell reactivity. Our results suggest that the genomic landscape of lung cancers shapes response to anti-PD-1 therapy.
PMCID:4993154
PMID: 25765070
ISSN: 1095-9203
CID: 2200792

Genetic basis for clinical response to CTLA-4 blockade [Letter]

Snyder, Alexandra; Wolchok, Jedd D; Chan, Timothy A
PMID: 25693024
ISSN: 1533-4406
CID: 2200842

Immunogenic peptide discovery in cancer genomes

Snyder, Alexandra; Chan, Timothy A
As immunotherapies to treat malignancy continue to diversify along with the tumor types amenable to treatment, it will become very important to predict which treatment is most likely to benefit a given patient. Tumor neoantigens, novel peptides resulting from somatic tumor mutations and recognized by the immune system as foreign, are likely to contribute significantly to the efficacy of immunotherapy. Multiple in silico methods have been developed to predict whether peptides, including tumor neoantigens, will be presented by the major histocompatibility complex (MHC) Class I or Class II, and interact with the T cell receptor (TCR). The methods for neoantigen prediction will be reviewed here, along with the most important examples of their use in the field of oncology.
PMCID:6657809
PMID: 25588790
ISSN: 1879-0380
CID: 4187252

Immunotherapy of Melanoma

Snyder, Alexandra; Zamarin, Dmitriy; Wolchok, Jedd D
The history of immunotherapy is rooted in the treatment of melanoma and therapy with immune checkpoint-blocking agents is now a cornerstone for the treatment of metastatic melanoma. The first effective immunotherapies approved by the US Food and Drug Administration in melanoma included interleukin-2 for metastatic disease and interferon alpha in the adjuvant setting. These were followed by a group of new therapies, including checkpoint-blocking antibodies targeting cytotoxic T lymphocyte-associated protein 4 and programmed cell death protein 1. Therapies intended to 'reeducate' T cells, such as tumor-infiltrating lymphocyte therapy, oncolytic viruses and tumor vaccines, have yielded promising results and are under development. Finally, the integration of the above therapies as well as development of new coinhibitory and costimulatory agents, though in early stages, appear very promising and likely represent the next phase in drug development for the treatment of metastatic melanoma.
PMID: 26376963
ISSN: 2296-1887
CID: 2200612

Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma

Snyder, Alexandra; Makarov, Vladimir; Merghoub, Taha; Yuan, Jianda; Zaretsky, Jesse M; Desrichard, Alexis; Walsh, Logan A; Postow, Michael A; Wong, Phillip; Ho, Teresa S; Hollmann, Travis J; Bruggeman, Cameron; Kannan, Kasthuri; Li, Yanyun; Elipenahli, Ceyhan; Liu, Cailian; Harbison, Christopher T; Wang, Lisu; Ribas, Antoni; Wolchok, Jedd D; Chan, Timothy A
Background Immune checkpoint inhibitors are effective cancer treatments, but molecular determinants of clinical benefit are unknown. Ipilimumab and tremelimumab are antibodies against cytotoxic T-lymphocyte antigen 4 (CTLA-4). Anti-CTLA-4 treatment prolongs overall survival in patients with melanoma. CTLA-4 blockade activates T cells and enables them to destroy tumor cells. Methods We obtained tumor tissue from patients with melanoma who were treated with ipilimumab or tremelimumab. Whole-exome sequencing was performed on tumors and matched blood samples. Somatic mutations and candidate neoantigens generated from these mutations were characterized. Neoantigen peptides were tested for the ability to activate lymphocytes from ipilimumab-treated patients. Results Malignant melanoma exomes from 64 patients treated with CTLA-4 blockade were characterized with the use of massively parallel sequencing. A discovery set consisted of 11 patients who derived a long-term clinical benefit and 14 patients who derived a minimal benefit or no benefit. Mutational load was associated with the degree of clinical benefit (P=0.01) but alone was not sufficient to predict benefit. Using genomewide somatic neoepitope analysis and patient-specific HLA typing, we identified candidate tumor neoantigens for each patient. We elucidated a neoantigen landscape that is specifically present in tumors with a strong response to CTLA-4 blockade. We validated this signature in a second set of 39 patients with melanoma who were treated with anti-CTLA-4 antibodies. Predicted neoantigens activated T cells from the patients treated with ipilimumab. Conclusions These findings define a genetic basis for benefit from CTLA-4 blockade in melanoma and provide a rationale for examining exomes of patients for whom anti-CTLA-4 agents are being considered. (Funded by the Frederick Adler Fund and others.).
PMCID:4315319
PMID: 25409260
ISSN: 0028-4793
CID: 1355992

A virtual consult service to optimize clinical trial participation in patients with metastatic breast cancer. [Meeting Abstract]

Cadoo, Karen Anne; Shah, Payal Deepak; Charen, Alexandra Snyder; Bromberg, Jacqueline; Comen, Elizabeth Anne; D\Andrea, Gabriella; Dang, Chau T.; Gilewski, Teresa; Gucalp, Ayca; Lake, Diana; Seidman, Andrew David; Theodoulou, Maria; Traina, Tiffany A.; Hudis, Clifford A.; Norton, Larry; Moynahan, Mary Ellen; Modi, Shanu; Dicker, Maura N.; Baselga, Jose; Chandarlapaty, Sarat
ISI:000358613203786
ISSN: 0732-183x
CID: 4187302