scispace - formally typeset
Search or ask a question
Author

Spencer C. Wei

Bio: Spencer C. Wei is an academic researcher from University of Texas MD Anderson Cancer Center. The author has contributed to research in topics: Cytotoxic T cell & CD8. The author has an hindex of 12, co-authored 18 publications receiving 4577 citations. Previous affiliations of Spencer C. Wei include University of California, San Diego & University of California, Berkeley.

Papers
More filters
Journal ArticleDOI
05 Jan 2018-Science
TL;DR: Examination of the oral and gut microbiome of melanoma patients undergoing anti-programmed cell death 1 protein (PD-1) immunotherapy suggested enhanced systemic and antitumor immunity in responding patients with a favorable gut microbiome as well as in germ-free mice receiving fecal transplants from responding patients.
Abstract: Preclinical mouse models suggest that the gut microbiome modulates tumor response to checkpoint blockade immunotherapy; however, this has not been well-characterized in human cancer patients. Here we examined the oral and gut microbiome of melanoma patients undergoing anti-programmed cell death 1 protein (PD-1) immunotherapy (n = 112). Significant differences were observed in the diversity and composition of the patient gut microbiome of responders versus nonresponders. Analysis of patient fecal microbiome samples (n = 43, 30 responders, 13 nonresponders) showed significantly higher alpha diversity (P < 0.01) and relative abundance of bacteria of the Ruminococcaceae family (P < 0.01) in responding patients. Metagenomic studies revealed functional differences in gut bacteria in responders, including enrichment of anabolic pathways. Immune profiling suggested enhanced systemic and antitumor immunity in responding patients with a favorable gut microbiome as well as in germ-free mice receiving fecal transplants from responding patients. Together, these data have important implications for the treatment of melanoma patients with immune checkpoint inhibitors.

2,791 citations

Journal ArticleDOI
TL;DR: The current state of understanding of T-cell costimulatory mechanisms and checkpoint blockade, primarily of CTLA4 and PD-1, is reviewed, and conceptual gaps in knowledge are highlighted.
Abstract: Immune checkpoint blockade is able to induce durable responses across multiple types of cancer, which has enabled the oncology community to begin to envision potentially curative therapeutic approaches. However, the remarkable responses to immunotherapies are currently limited to a minority of patients and indications, highlighting the need for more effective and novel approaches. Indeed, an extraordinary amount of preclinical and clinical investigation is exploring the therapeutic potential of negative and positive costimulatory molecules. Insights into the underlying biological mechanisms and functions of these molecules have, however, lagged significantly behind. Such understanding will be essential for the rational design of next-generation immunotherapies. Here, we review the current state of our understanding of T-cell costimulatory mechanisms and checkpoint blockade, primarily of CTLA4 and PD-1, and highlight conceptual gaps in knowledge. Significance: This review provides an overview of immune checkpoint blockade therapy from a basic biology and immunologic perspective for the cancer research community. Cancer Discov; 8(9); 1069–86. ©2018 AACR.

1,893 citations

Journal ArticleDOI
07 Sep 2017-Cell
TL;DR: These findings indicate that anti-CTLA-4 and anti-PD-1 checkpoint-blockade-induced immune responses are driven by distinct cellular mechanisms, and that checkpoint blockade targets only specific subsets of tumor-infiltrating T cell populations.

856 citations

Journal ArticleDOI
TL;DR: It is reported that TWIST1 is an essential mechanomediator that promotes epithelial–mesenchymal transition (EMT) in response to increasing matrix stiffness that responds to biomechanical signals from the tumour microenvironment to drive EMT, invasion and metastasis.
Abstract: Matrix stiffness potently regulates cellular behaviour in various biological contexts. In breast tumours, the presence of dense clusters of collagen fibrils indicates increased matrix stiffness and correlates with poor survival. It is unclear how mechanical inputs are transduced into transcriptional outputs to drive tumour progression. Here we report that TWIST1 is an essential mechanomediator that promotes epithelial-mesenchymal transition (EMT) in response to increasing matrix stiffness. High matrix stiffness promotes nuclear translocation of TWIST1 by releasing TWIST1 from its cytoplasmic binding partner G3BP2. Loss of G3BP2 leads to constitutive TWIST1 nuclear localization and synergizes with increasing matrix stiffness to induce EMT and promote tumour invasion and metastasis. In human breast tumours, collagen fibre alignment, a marker of increasing matrix stiffness, and reduced expression of G3BP2 together predict poor survival. Our findings reveal a TWIST1-G3BP2 mechanotransduction pathway that responds to biomechanical signals from the tumour microenvironment to drive EMT, invasion and metastasis.

672 citations

Journal ArticleDOI
TL;DR: It is determined that dual blockade of CTLA-4 and PD-1 therapy is sufficient to induce unique cellular responses compared with either monotherapy, which has significant implications for both the cellular mechanisms of action and biomarkers of response to monotherapies and combination therapy.
Abstract: Immune checkpoint blockade therapy targets T cell-negative costimulatory molecules such as cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 (PD-1). Combination anti–CTLA-4 and anti–PD-1 blockade therapy has enhanced efficacy, but it remains unclear through what mechanisms such effects are mediated. A critical question is whether combination therapy targets and modulates the same T cell populations as monotherapies. Using a mass cytometry-based systems approach, we comprehensively profiled the response of T cell populations to monotherapy and combination anti–CTLA-4 plus anti–PD-1 therapy in syngeneic murine tumors and clinical samples. Most effects of monotherapies were additive in the context of combination therapy; however, multiple combination therapy-specific effects were observed. Highly phenotypically exhausted cluster of differentiation 8 (CD8) T cells expand in frequency following anti–PD-1 monotherapy but not combination therapy, while activated terminally differentiated effector CD8 T cells expand only following combination therapy. Combination therapy also led to further increased frequency of T helper type 1 (Th1)-like CD4 effector T cells even though anti–PD-1 monotherapy is not sufficient to do so. Mass cytometry analyses of peripheral blood from melanoma patients treated with immune checkpoint blockade therapies similarly revealed mostly additive effects on the frequencies of T cell subsets along with unique modulation of terminally differentiated effector CD8 T cells by combination ipilimumab plus nivolumab therapy. Together, these findings indicate that dual blockade of CTLA-4 and PD-1 therapy is sufficient to induce unique cellular responses compared with either monotherapy.

203 citations


Cited by
More filters
Journal ArticleDOI
Evan Bolyen1, Jai Ram Rideout1, Matthew R. Dillon1, Nicholas A. Bokulich1, Christian C. Abnet2, Gabriel A. Al-Ghalith3, Harriet Alexander4, Harriet Alexander5, Eric J. Alm6, Manimozhiyan Arumugam7, Francesco Asnicar8, Yang Bai9, Jordan E. Bisanz10, Kyle Bittinger11, Asker Daniel Brejnrod7, Colin J. Brislawn12, C. Titus Brown4, Benjamin J. Callahan13, Andrés Mauricio Caraballo-Rodríguez14, John Chase1, Emily K. Cope1, Ricardo Silva14, Christian Diener15, Pieter C. Dorrestein14, Gavin M. Douglas16, Daniel M. Durall17, Claire Duvallet6, Christian F. Edwardson, Madeleine Ernst18, Madeleine Ernst14, Mehrbod Estaki17, Jennifer Fouquier19, Julia M. Gauglitz14, Sean M. Gibbons20, Sean M. Gibbons15, Deanna L. Gibson17, Antonio Gonzalez14, Kestrel Gorlick1, Jiarong Guo21, Benjamin Hillmann3, Susan Holmes22, Hannes Holste14, Curtis Huttenhower23, Curtis Huttenhower24, Gavin A. Huttley25, Stefan Janssen26, Alan K. Jarmusch14, Lingjing Jiang14, Benjamin D. Kaehler25, Benjamin D. Kaehler27, Kyo Bin Kang28, Kyo Bin Kang14, Christopher R. Keefe1, Paul Keim1, Scott T. Kelley29, Dan Knights3, Irina Koester14, Tomasz Kosciolek14, Jorden Kreps1, Morgan G. I. Langille16, Joslynn S. Lee30, Ruth E. Ley31, Ruth E. Ley32, Yong-Xin Liu, Erikka Loftfield2, Catherine A. Lozupone19, Massoud Maher14, Clarisse Marotz14, Bryan D Martin20, Daniel McDonald14, Lauren J. McIver24, Lauren J. McIver23, Alexey V. Melnik14, Jessica L. Metcalf33, Sydney C. Morgan17, Jamie Morton14, Ahmad Turan Naimey1, Jose A. Navas-Molina34, Jose A. Navas-Molina14, Louis-Félix Nothias14, Stephanie B. Orchanian, Talima Pearson1, Samuel L. Peoples20, Samuel L. Peoples35, Daniel Petras14, Mary L. Preuss36, Elmar Pruesse19, Lasse Buur Rasmussen7, Adam R. Rivers37, Michael S. Robeson38, Patrick Rosenthal36, Nicola Segata8, Michael Shaffer19, Arron Shiffer1, Rashmi Sinha2, Se Jin Song14, John R. Spear39, Austin D. Swafford, Luke R. Thompson40, Luke R. Thompson41, Pedro J. Torres29, Pauline Trinh20, Anupriya Tripathi14, Peter J. Turnbaugh10, Sabah Ul-Hasan42, Justin J. J. van der Hooft43, Fernando Vargas, Yoshiki Vázquez-Baeza14, Emily Vogtmann2, Max von Hippel44, William A. Walters32, Yunhu Wan2, Mingxun Wang14, Jonathan Warren45, Kyle C. Weber46, Kyle C. Weber37, Charles H. D. Williamson1, Amy D. Willis20, Zhenjiang Zech Xu14, Jesse R. Zaneveld20, Yilong Zhang47, Qiyun Zhu14, Rob Knight14, J. Gregory Caporaso1 
TL;DR: QIIME 2 development was primarily funded by NSF Awards 1565100 to J.G.C. and R.K.P. and partial support was also provided by the following: grants NIH U54CA143925 and U54MD012388.
Abstract: QIIME 2 development was primarily funded by NSF Awards 1565100 to J.G.C. and 1565057 to R.K. Partial support was also provided by the following: grants NIH U54CA143925 (J.G.C. and T.P.) and U54MD012388 (J.G.C. and T.P.); grants from the Alfred P. Sloan Foundation (J.G.C. and R.K.); ERCSTG project MetaPG (N.S.); the Strategic Priority Research Program of the Chinese Academy of Sciences QYZDB-SSW-SMC021 (Y.B.); the Australian National Health and Medical Research Council APP1085372 (G.A.H., J.G.C., Von Bing Yap and R.K.); the Natural Sciences and Engineering Research Council (NSERC) to D.L.G.; and the State of Arizona Technology and Research Initiative Fund (TRIF), administered by the Arizona Board of Regents, through Northern Arizona University. All NCI coauthors were supported by the Intramural Research Program of the National Cancer Institute. S.M.G. and C. Diener were supported by the Washington Research Foundation Distinguished Investigator Award.

8,821 citations

Journal ArticleDOI
23 Mar 2018-Science
TL;DR: New-generation combinatorial therapies may overcome resistance mechanisms to immune checkpoint therapy, and evidence points to alterations that converge on the antigen presentation and interferon-γ signaling pathways.
Abstract: The release of negative regulators of immune activation (immune checkpoints) that limit antitumor responses has resulted in unprecedented rates of long-lasting tumor responses in patients with a variety of cancers. This can be achieved by antibodies blocking the cytotoxic T lymphocyte–associated protein 4 (CTLA-4) or the programmed cell death 1 (PD-1) pathway, either alone or in combination. The main premise for inducing an immune response is the preexistence of antitumor T cells that were limited by specific immune checkpoints. Most patients who have tumor responses maintain long-lasting disease control, yet one-third of patients relapse. Mechanisms of acquired resistance are currently poorly understood, but evidence points to alterations that converge on the antigen presentation and interferon-γ signaling pathways. New-generation combinatorial therapies may overcome resistance mechanisms to immune checkpoint therapy.

3,736 citations

Journal ArticleDOI
TL;DR: The current state of understanding of T-cell costimulatory mechanisms and checkpoint blockade, primarily of CTLA4 and PD-1, is reviewed, and conceptual gaps in knowledge are highlighted.
Abstract: Immune checkpoint blockade is able to induce durable responses across multiple types of cancer, which has enabled the oncology community to begin to envision potentially curative therapeutic approaches. However, the remarkable responses to immunotherapies are currently limited to a minority of patients and indications, highlighting the need for more effective and novel approaches. Indeed, an extraordinary amount of preclinical and clinical investigation is exploring the therapeutic potential of negative and positive costimulatory molecules. Insights into the underlying biological mechanisms and functions of these molecules have, however, lagged significantly behind. Such understanding will be essential for the rational design of next-generation immunotherapies. Here, we review the current state of our understanding of T-cell costimulatory mechanisms and checkpoint blockade, primarily of CTLA4 and PD-1, and highlight conceptual gaps in knowledge. Significance: This review provides an overview of immune checkpoint blockade therapy from a basic biology and immunologic perspective for the cancer research community. Cancer Discov; 8(9); 1069–86. ©2018 AACR.

1,893 citations

Journal ArticleDOI
05 Jan 2018-Science
TL;DR: The results suggest that the commensal microbiome may have a mechanistic impact on antitumor immunity in human cancer patients and could lead to improved tumor control, augmented T cell responses, and greater efficacy of anti–PD-L1 therapy.
Abstract: Anti–PD-1–based immunotherapy has had a major impact on cancer treatment but has only benefited a subset of patients. Among the variables that could contribute to interpatient heterogeneity is differential composition of the patients’ microbiome, which has been shown to affect antitumor immunity and immunotherapy efficacy in preclinical mouse models. We analyzed baseline stool samples from metastatic melanoma patients before immunotherapy treatment, through an integration of 16 S ribosomal RNA gene sequencing, metagenomic shotgun sequencing, and quantitative polymerase chain reaction for selected bacteria. A significant association was observed between commensal microbial composition and clinical response. Bacterial species more abundant in responders included Bifidobacterium longum , Collinsella aerofaciens , and Enterococcus faecium. Reconstitution of germ-free mice with fecal material from responding patients could lead to improved tumor control, augmented T cell responses, and greater efficacy of anti–PD-L1 therapy. Our results suggest that the commensal microbiome may have a mechanistic impact on antitumor immunity in human cancer patients.

1,820 citations

Journal ArticleDOI
TL;DR: This guide to cancer immunotherapy provides a comprehensive historical and biological perspective regarding the advent and clinical implementation of cancer immunotherapeutics, with an emphasis on the fundamental importance of T lymphocyte regulation.
Abstract: The T lymphocyte, especially its capacity for antigen-directed cytotoxicity, has become a central focus for engaging the immune system in the fight against cancer. Basic science discoveries elucidating the molecular and cellular biology of the T cell have led to new strategies in this fight, including checkpoint blockade, adoptive cellular therapy and cancer vaccinology. This area of immunological research has been highly active for the past 50 years and is now enjoying unprecedented bench-to-bedside clinical success. Here, we provide a comprehensive historical and biological perspective regarding the advent and clinical implementation of cancer immunotherapeutics, with an emphasis on the fundamental importance of T lymphocyte regulation. We highlight clinical trials that demonstrate therapeutic efficacy and toxicities associated with each class of drug. Finally, we summarize emerging therapies and emphasize the yet to be elucidated questions and future promise within the field of cancer immunotherapy.

1,695 citations