Network


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

Hotspot


Dive into the research topics where Marissa Vignali is active.

Publication


Featured researches published by Marissa Vignali.


Science Translational Medicine | 2015

High-throughput pairing of T cell receptor α and β sequences

Bryan Howie; Anna Sherwood; Ashley D. Berkebile; Jan Berka; Ryan Emerson; David Williamson; Ilan Kirsch; Marissa Vignali; Mark J. Rieder; Christopher S. Carlson; Harlan Robins

T cell receptor α and β sequences can be accurately paired from hundreds of thousands of T cell clones in parallel. T cell receptor chains pair off High-throughput immunosequencing can take a snapshot of the repertoire of immune cells, providing a broad picture of the immune response at any given time and tracking how the immune response changes as a result of perturbations such as vaccines, infection, or cancer. However, this approach has been limited by the inability to determine which TCR α and TCR β chains combine to form specific T cell receptors in a given cell. Now, Howie et al. report and validate a high-throughput method to pair TCR α and β segments without the need for single-cell technologies. They confirm that their method can be used for T cells from both blood and solid tissues. The T cell receptor (TCR) protein is a heterodimer composed of an α chain and a β chain. TCR genes undergo somatic DNA rearrangements to generate the diversity of T cell binding specificities needed for effective immunity. Recently, high-throughput immunosequencing methods have been developed to profile the TCR α (TCRA) and TCR β (TCRB) repertoires. However, these methods cannot determine which TCRA and TCRB chains combine to form a specific TCR, which is essential for many functional and therapeutic applications. We describe and validate a method called pairSEQ, which can leverage the diversity of TCR sequences to accurately pair hundreds of thousands of TCRA and TCRB sequences in a single experiment. Our TCR pairing method uses standard laboratory consumables and equipment without the need for single-cell technologies. We show that pairSEQ can be applied to T cells from both blood and solid tissues, such as tumors.


Molecular Oncology | 2015

T-cell receptor profiling in cancer

Ilan Kirsch; Marissa Vignali; Harlan Robins

Immunosequencing is a platform technology that allows the enumeration, specification and quantification of each and every B‐ and/or T‐cell in any biologic sample of interest. Thus, it provides an assessment of the level and distribution of all the clonal lymphocytes in any sample, and allows “tracking” of a single clone or multiple clones of interest over time or from tissue to tissue within a given patient. It is based on bias‐controlled multiplex PCR and high‐throughput sequencing, and it is highly accurate, standardized, and sensitive. In this review, we provide evidence that immunosequencing is becoming an important analytic tool for the emerging field of immune‐oncology, and describe several applications of this approach, including the assessment of residual disease post therapy in lymphoid malignancies, the prediction of response to immunotherapeutics of solid tumors containing tumor infiltrating lymphocytes, the identification of clonal responses in vaccination, infectious disease, bone marrow reconstitution, and autoimmunity, and the exploration of whether there are population‐based stereotyped responses to certain exposures or interventions.


Nature Genetics | 2017

Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T cell repertoire

Ryan Emerson; William DeWitt; Marissa Vignali; Jenna Gravley; Joyce K Hu; Edward J Osborne; Cindy Desmarais; Mark Klinger; Christopher S. Carlson; John A. Hansen; Mark J. Rieder; Harlan Robins

An individuals T cell repertoire dynamically encodes their pathogen exposure history. To determine whether pathogen exposure signatures can be identified by documenting public T cell receptors (TCRs), we profiled the T cell repertoire of 666 subjects with known cytomegalovirus (CMV) serostatus by immunosequencing. We developed a statistical classification framework that could diagnose CMV status from the resulting catalog of TCRβ sequences with high specificity and sensitivity in both the original cohort and a validation cohort of 120 different subjects. We also confirmed that three of the identified CMV-associated TCRβ molecules bind CMV in vitro, and, moreover, we used this approach to accurately predict the HLA-A and HLA-B alleles of most subjects in the first cohort. As all memory T cell responses are encoded in the common format of somatic TCR recombination, our approach could potentially be generalized to a wide variety of disease states, as well as other immunological phenotypes, as a highly parallelizable diagnostic strategy.


Journal of Virology | 2015

Dynamics of the Cytotoxic T Cell Response to a Model of Acute Viral Infection

William DeWitt; Ryan Emerson; Paul Lindau; Marissa Vignali; Thomas M. Snyder; Cindy Desmarais; Catherine Sanders; Heidi Utsugi; Edus H. Warren; Juliana McElrath; Karen W. Makar; Anna Wald; Harlan Robins

ABSTRACT A detailed characterization of the dynamics and breadth of the immune response to an acute viral infection, as well as the determinants of recruitment to immunological memory, can greatly contribute to our basic understanding of the mechanics of the human immune system and can ultimately guide the design of effective vaccines. In addition to neutralizing antibodies, T cells have been shown to be critical for the effective resolution of acute viral infections. We report the first in-depth analysis of the dynamics of the CD8+ T cell repertoire at the level of individual T cell clonal lineages upon vaccination of human volunteers with a single dose of YF-17D. This live attenuated yellow fever virus vaccine yields sterile, long-term immunity and has been previously used as a model to understand the immune response to a controlled acute viral infection. We identified and enumerated unique CD8+ T cell clones specifically induced by this vaccine through a combined experimental and statistical approach that included high-throughput sequencing of the CDR3 variable region of the T cell receptor β-chain and an algorithm that detected significantly expanded T cell clones. This allowed us to establish that (i) on average, ∼2,000 CD8+ T cell clones were induced by YF-17D, (ii) 5 to 6% of the responding clones were recruited to long-term memory 3 months postvaccination, (iii) the most highly expanded effector clones were preferentially recruited to the memory compartment, and (iv) a fraction of the YF-17D-induced clones could be identified from peripheral blood lymphocytes solely by measuring clonal expansion. IMPORTANCE The exhaustive investigation of pathogen-induced effector T cells is essential to accurately quantify the dynamics of the human immune response. The yellow fever vaccine (YFV) has been broadly used as a model to understand how a controlled, self-resolving acute viral infection induces an effective and long-term protective immune response. Here, we extend this previous work by reporting the identity of activated effector T cell clones that expand in response to the YFV 2 weeks postvaccination (as defined by their unique T cell receptor gene sequence) and by tracking clones that enter the memory compartment 3 months postvaccination. This is the first study to use high-throughput sequencing of immune cells to characterize the breadth of the antiviral effector cell response and to determine the contribution of unique virus-induced clones to the long-lived memory T cell repertoire. Thus, this study establishes a benchmark against which future vaccines can be compared to predict their efficacy.


PLOS Medicine | 2017

Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: An exploratory multi-omic analysis

Alexandra Snyder; Tavi Nathanson; Samuel Funt; Arun Ahuja; Jacqueline Buros Novik; Matthew D. Hellmann; Eliza Chang; Bülent Arman Aksoy; Hikmat Al-Ahmadie; Erik Yusko; Marissa Vignali; Sharon Benzeno; Mariel Elena Boyd; Meredith Maisie Moran; Gopa Iyer; Harlan Robins; Elaine R. Mardis; Taha Merghoub; Jeff Hammerbacher; Jonathan E. Rosenberg; Dean F. Bajorin

Background Inhibition of programmed death-ligand 1 (PD-L1) with atezolizumab can induce durable clinical benefit (DCB) in patients with metastatic urothelial cancers, including complete remissions in patients with chemotherapy refractory disease. Although mutation load and PD-L1 immune cell (IC) staining have been associated with response, they lack sufficient sensitivity and specificity for clinical use. Thus, there is a need to evaluate the peripheral blood immune environment and to conduct detailed analyses of mutation load, predicted neoantigens, and immune cellular infiltration in tumors to enhance our understanding of the biologic underpinnings of response and resistance. Methods and findings The goals of this study were to (1) evaluate the association of mutation load and predicted neoantigen load with therapeutic benefit and (2) determine whether intratumoral and peripheral blood T cell receptor (TCR) clonality inform clinical outcomes in urothelial carcinoma treated with atezolizumab. We hypothesized that an elevated mutation load in combination with T cell clonal dominance among intratumoral lymphocytes prior to treatment or among peripheral T cells after treatment would be associated with effective tumor control upon treatment with anti-PD-L1 therapy. We performed whole exome sequencing (WES), RNA sequencing (RNA-seq), and T cell receptor sequencing (TCR-seq) of pretreatment tumor samples as well as TCR-seq of matched, serially collected peripheral blood, collected before and after treatment with atezolizumab. These parameters were assessed for correlation with DCB (defined as progression-free survival [PFS] >6 months), PFS, and overall survival (OS), both alone and in the context of clinical and intratumoral parameters known to be predictive of survival in this disease state. Patients with DCB displayed a higher proportion of tumor-infiltrating T lymphocytes (TIL) (n = 24, Mann-Whitney p = 0.047). Pretreatment peripheral blood TCR clonality below the median was associated with improved PFS (n = 29, log-rank p = 0.048) and OS (n = 29, log-rank p = 0.011). Patients with DCB also demonstrated more substantial expansion of tumor-associated TCR clones in the peripheral blood 3 weeks after starting treatment (n = 22, Mann-Whitney p = 0.022). The combination of high pretreatment peripheral blood TCR clonality with elevated PD-L1 IC staining in tumor tissue was strongly associated with poor clinical outcomes (n = 10, hazard ratio (HR) (mean) = 89.88, HR (median) = 23.41, 95% CI [2.43, 506.94], p(HR > 1) = 0.0014). Marked variations in mutation loads were seen with different somatic variant calling methodologies, which, in turn, impacted associations with clinical outcomes. Missense mutation load, predicted neoantigen load, and expressed neoantigen load did not demonstrate significant association with DCB (n = 25, Mann-Whitney p = 0.22, n = 25, Mann-Whitney p = 0.55, and n = 25, Mann-Whitney p = 0.29, respectively). Instead, we found evidence of time-varying effects of somatic mutation load on PFS in this cohort (n = 25, p = 0.044). A limitation of our study is its small sample size (n = 29), a subset of the patients treated on IMvigor 210 (NCT02108652). Given the number of exploratory analyses performed, we intend for these results to be hypothesis-generating. Conclusions These results demonstrate the complex nature of immune response to checkpoint blockade and the compelling need for greater interrogation and data integration of both host and tumor factors. Incorporating these variables in prospective studies will facilitate identification and treatment of resistant patients.


Clinical Cancer Research | 2017

Fractionated radiation therapy stimulates anti-tumor immunity mediated by both resident and infiltrating polyclonal T-cell populations when combined with PD1 blockade

Simon J. Dovedi; Eleanor J. Cheadle; Amy Popple; Edmund Poon; Michelle Morrow; Ross Stewart; Erik Yusko; Catherine Sanders; Marissa Vignali; Ryan Emerson; Harlan Robins; Robert W. Wilkinson; Jamie Honeychurch; Tim Illidge

Purpose: Radiotherapy is a highly effective anticancer treatment forming part of the standard of care for the majority of patients, but local and distal disease recurrence remains a major cause of mortality. Radiotherapy is known to enhance tumor immunogenicity; however, the contribution and mechanisms of radiotherapy-induced immune responses are unknown. Experimental Design: The impact of low-dose fractionated radiotherapy (5 × 2 Gy) alone and in combination with αPD-1 mAb on the tumor microenvironment was evaluated by flow cytometry and next-generation sequencing of the T-cell receptor (TCR) repertoire. A dual-tumor model was used, with fractionated radiotherapy delivered to a single tumor site to enable evaluation of the local and systemic response to treatment and ability to induce abscopal responses outside the radiation field. Results: We show that fractionated radiotherapy leads to T-cell infiltration at the irradiated site; however, the TCR landscape remains dominated by polyclonal expansion of preexisting T-cell clones. Adaptive resistance via the PD-1/PD-L1 pathway restricts the generation of systemic anticancer immunity following radiotherapy, which can be overcome through combination with αPD-1 mAb leading to improved local and distal tumor control. Moreover, we show that effective clearance of tumor following combination therapy is dependent on both T cells resident in the tumor at the time of radiotherapy and infiltrating T cells. Conclusions: These data provide evidence that radiotherapy can enhance T-cell trafficking to locally treated tumor sites and augment preexisting anticancer T-cell responses with the capacity to mediate regression of out-of-field tumor lesions when delivered in combination with αPD-1 mAb therapy. Clin Cancer Res; 23(18); 5514–26. ©2017 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities

Sumit K. Subudhi; Ana Aparicio; Jianjun Gao; Amado J. Zurita; John C. Araujo; Christopher J. Logothetis; Salahaldin A. Tahir; Brinda R. Korivi; Rebecca S. Slack; Luis Vence; Ryan Emerson; Erik Yusko; Marissa Vignali; Harlan Robins; Jingjing Sun; James P. Allison; Padmanee Sharma

Significance Immune checkpoint therapies have garnered significant attention due to their ability to induce dramatic clinical responses in patients with various solid tumor malignancies, including prostate cancer. However, these therapeutic agents often elicit immune-related adverse events (irAEs) that may result in substantial morbidity. Early intervention can markedly reduce the severity of the irAEs, but biomarkers that allow for their early detection and guide their management are lacking. Based on peripheral blood samples collected longitudinally in two prostate cancer clinical trials, we propose CD8 T-cell clonal expansion within the systemic circulation as a potential correlative biomarker of immune-related adverse events that occur with ipilimumab therapy. Immune checkpoint therapies, such as ipilimumab, induce dramatic antitumor responses in a subset of patients with advanced malignancies, but they may also induce inflammatory responses and toxicities termed immune-related adverse events (irAEs). These irAEs are often low grade and manageable, but severe irAEs may lead to prolonged hospitalizations or fatalities. Early intervention is necessary to minimize morbidities that occur with severe irAEs. However, correlative biomarkers are currently lacking. In a phase II clinical trial that treated 27 patients with metastatic prostate cancer, we aimed to test the safety and efficacy of androgen deprivation therapy plus ipilimumab. In this study, we observed grade 3 toxicities in >40% of treated patients, which led to early closure of the study. Because ipilimumab enhances T-cell responses, we hypothesized that increased clonal T-cell responses in the systemic circulation may contribute to irAEs. Sequencing of the T-cell receptor β-chains in purified T cells revealed clonal expansion of CD8 T cells, which occurred in blood samples collected before the onset of grade 2–3 irAEs. These initial results suggested that expansion of ≥55 CD8 T-cell clones preceded the development of severe irAEs. We further evaluated available blood samples from a second trial and determined that patients who experienced grade 2–3 irAEs also had expansion of ≥55 CD8 T-cell clones in blood samples collected before the onset of irAEs. We propose that CD8 T-cell clonal expansion may be a correlative biomarker to enable close monitoring and early intervention for patients receiving ipilimumab.


PLOS ONE | 2016

A Public Database of Memory and Naive B-Cell Receptor Sequences.

William DeWitt; Paul Lindau; Thomas M. Snyder; Anna Sherwood; Marissa Vignali; Christopher S. Carlson; Philip D. Greenberg; Natalie Duerkopp; Ryan Emerson; Harlan Robins

The vast diversity of B-cell receptors (BCR) and secreted antibodies enables the recognition of, and response to, a wide range of epitopes, but this diversity has also limited our understanding of humoral immunity. We present a public database of more than 37 million unique BCR sequences from three healthy adult donors that is many fold deeper than any existing resource, together with a set of online tools designed to facilitate the visualization and analysis of the annotated data. We estimate the clonal diversity of the naive and memory B-cell repertoires of healthy individuals, and provide a set of examples that illustrate the utility of the database, including several views of the basic properties of immunoglobulin heavy chain sequences, such as rearrangement length, subunit usage, and somatic hypermutation positions and dynamics.


Cancer | 2017

T-cell infiltration and clonality correlate with programmed cell death protein 1 and programmed death-ligand 1 expression in patients with soft tissue sarcomas

Seth M. Pollack; Qianchuan He; Jennifer H. Yearley; Ryan Emerson; Marissa Vignali; Yuzheng Zhang; Mary W. Redman; Kelsey K. Baker; Sara Cooper; Bailey Donahue; Elizabeth T. Loggers; Lee D. Cranmer; M.B. Spraker; Y. David Seo; Venu G. Pillarisetty; Robert W. Ricciotti; Benjamin Hoch; Terrill K. McClanahan; Erin Murphy; Wendy M. Blumenschein; Steven M. Townson; Sharon Benzeno; Stanley R. Riddell; Robin L. Jones

Patients with metastatic sarcomas have poor outcomes and although the disease may be amenable to immunotherapies, information regarding the immunologic profiles of soft tissue sarcoma (STS) subtypes is limited.


Cancer Discovery | 2017

TCR Repertoire Intratumor Heterogeneity in Localized Lung Adenocarcinomas: An Association with Predicted Neoantigen Heterogeneity and Postsurgical Recurrence

Alexandre Reuben; Rachel Gittelman; Jianjun Gao; Jiexin Zhang; Erik Yusko; Chang Jiun Wu; Ryan Emerson; Jianhua Zhang; Christopher Tipton; Jun Li; Kelly Quek; Vancheswaran Gopalakrishnan; Runzhe Chen; Luis Vence; Tina Cascone; Marissa Vignali; Junya Fujimoto; Jaime Rodriguez-Canales; Edwin R. Parra; Latasha Little; Curtis Gumbs; Marie Andrée Forget; Lorenzo Federico; Cara Haymaker; Carmen Behrens; Sharon Benzeno; Chantale Bernatchez; Boris Sepesi; Don L. Gibbons; Jennifer A. Wargo

Genomic intratumor heterogeneity (ITH) may be associated with postsurgical relapse of localized lung adenocarcinomas. Recently, mutations, through generation of neoantigens, were shown to alter tumor immunogenicity through T-cell responses. Here, we performed sequencing of the T-cell receptor (TCR) in 45 tumor regions from 11 localized lung adenocarcinomas and observed substantial intratumor differences in T-cell density and clonality with the majority of T-cell clones restricted to individual tumor regions. TCR ITH positively correlated with predicted neoantigen ITH, suggesting that spatial differences in the T-cell repertoire may be driven by distinct neoantigens in different tumor regions. Finally, a higher degree of TCR ITH was associated with an increased risk of postsurgical relapse and shorter disease-free survival, suggesting a potential clinical significance of T-cell repertoire heterogeneity.Significance: The present study provides insights into the ITH of the T-cell repertoire in localized lung adenocarcinomas and its potential biological and clinical impact. The results suggest that T-cell repertoire ITH may be tightly associated to genomic ITH and disease relapse. Cancer Discov; 7(10); 1088-97. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1047.

Collaboration


Dive into the Marissa Vignali's collaboration.

Top Co-Authors

Avatar

Harlan Robins

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Ryan Emerson

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

William DeWitt

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Anna Sherwood

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Christopher S. Carlson

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jianjun Gao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Luis Vence

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Mark J. Rieder

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Padmanee Sharma

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge