Searched for: in-biosketch:true
person:wongk11
Characterization of the Immune Landscape of EGFR-Mutant NSCLC Identifies CD73/Adenosine Pathway as a Potential Therapeutic Target
Le, Xiuning; Negrao, Marcelo V; Reuben, Alexandre; Federico, Lorenzo; Diao, Lixia; McGrail, Daniel; Nilsson, Monique; Robichaux, Jacqulyne; Munoz, Irene Guijarro; Patel, Sonia; Elamin, Yasir; Fan, You-Hong; Lee, Won-Chul; Parra, Edwin; Solis Soto, Luisa Maren; Chen, Runzhe; Li, Jun; Karpinets, Tatiana; Khairullah, Roohussaba; Kadara, Humam; Behrens, Carmen; Sepesi, Boris; Wang, Ruiping; Zhu, Mingrui; Wang, Linghua; Vaporciyan, Ara; Roth, Jack; Swisher, Stephen; Haymaker, Cara; Zhang, Jianhua; Wang, Jing; Wong, Kwok-Kin; Byers, Lauren A; Bernatchez, Chantale; Zhang, Jianjun; Wistuba, Ignacio I; Gibbons, Don L; Akbay, Esra A; Heymach, John V
INTRODUCTION/BACKGROUND:Lung adenocarcinomas harboring EGFR mutations do not respond to immune checkpoint blockade therapy and their EGFR wildtype counterpart. The mechanisms underlying this lack of clinical response have been investigated but remain incompletely understood. METHODS:We analyzed three cohorts of resected lung adenocarcinomas (Profiling of Resistance Patterns of Oncogenic Signaling Pathways in Evaluation of Cancer of Thorax, Immune Genomic Profiling of NSCLC, and The Cancer Genome Atlas) and compared tumor immune microenvironment of EGFR-mutant tumors to EGFR wildtype tumors, to identify actionable regulators to target and potentially enhance the treatment response. RESULTS:EGFR-mutant NSCLC exhibited low programmed death-ligand 1, low tumor mutational burden, decreased number of cytotoxic T cells, and low T cell receptor clonality, consistent with an immune-inert phenotype, though T cell expansion ex vivo was preserved. In an analysis of 75 immune checkpoint genes, the top up-regulated genes in the EGFR-mutant tumors (NT5E and ADORA1) belonged to the CD73/adenosine pathway. Single-cell analysis revealed that the tumor cell population expressed CD73, both in the treatment-naive and resistant tumors. Using coculture systems with EGFR-mutant NSCLC cells, T regulatory cell proportion was decreased with CD73 knockdown. In an immune-competent mouse model of EGFR-mutant lung cancer, the CD73/adenosine pathway was markedly up-regulated and CD73 blockade significantly inhibited tumor growth. CONCLUSIONS:Our work revealed that EGFR-mutant NSCLC has an immune-inert phenotype. We identified the CD73/adenosine pathway as a potential therapeutic target for EGFR-mutant NSCLC.
PMID: 33388477
ISSN: 1556-1380
CID: 4835272
Recent advances in preclinical models for lung squamous cell carcinoma
Pan, Yuanwang; Han, Han; Labbe, Kristen E; Zhang, Hua; Wong, Kwok-Kin
Lung squamous cell carcinoma (LUSC) represents a major subtype of non-small cell lung cancer with limited treatment options. Previous studies have elucidated the complex genetic landscape of LUSC and revealed multiple altered genes and pathways. However, in stark contrast to lung adenocarcinoma, few targetable driver mutations have been established so far and targeted therapies for LUSC remain unsuccessful. Immunotherapy has revolutionized LUSC treatment and is currently approved as the new standard of care. To gain a better understanding of the LUSC biology, improved modeling systems are urgently needed. Preclinical models, particularly those mimicking human disease with an intact tumor immune microenvironment, are an invaluable tool to study cancer development and evaluate new therapeutic targets. Here, we discuss recent advances in LUSC preclinical models, with a focus on genetically engineered mouse models (GEMMs) and organoids, in the context of evolving precision medicine and immunotherapy.
PMID: 33707749
ISSN: 1476-5594
CID: 4819622
Response to Cottu, Bozec, Basse, and Paoletti
Zhang, Hua; Han, Han; He, Tianhui; Labbe, Kristen E; Hernandez, Adrian V; Chen, Haiquan; Velcheti, Vamsidhar; Stebbing, Justin; Wong, Kwok-Kin
PMID: 33404597
ISSN: 1460-2105
CID: 4738932
An Empirical Antigen Selection Method Identifies Neoantigens That Either Elicit Broad Antitumor T-cell Responses or Drive Tumor Growth
Lam, Hubert; McNeil, Lisa K; Starobinets, Hanna; DeVault, Victoria L; Cohen, Roger B; Twardowski, Przemyslaw; Johnson, Melissa L; Gillison, Maura L; Stein, Mark N; Vaishampayan, Ulka N; DeCillis, Arthur P; Foti, James J; Vemulapalli, Vijetha; Tjon, Emily; Ferber, Kyle; DeOliveira, Daniel B; Broom, Wendy; Agnihotri, Parul; Jaffee, Elizabeth M; Wong, Kwok-Kin; Drake, Charles G; Carroll, Pamela M; Davis, Thomas A; Flechtner, Jessica Baker
Neoantigens are critical targets of antitumor T-cell responses. The ATLAS bioassay was developed to identify neoantigens empirically by expressing each unique patient-specific tumor mutation individually in Escherichia coli, pulsing autologous dendritic cells in an ordered array, and testing the patient's T cells for recognition in an overnight assay. Profiling of T cells from patients with lung cancer revealed both stimulatory and inhibitory responses to individual neoantigens. In the murine B16F10 melanoma model, therapeutic immunization with ATLAS-identified stimulatory neoantigens protected animals, whereas immunization with peptides associated with inhibitory ATLAS responses resulted in accelerated tumor growth and abolished efficacy of an otherwise protective vaccine. A planned interim analysis of a clinical study testing a poly-ICLC adjuvanted personalized vaccine containing ATLAS-identified stimulatory neoantigens showed that it is well tolerated. In an adjuvant setting, immunized patients generated both CD4+ and CD8+ T-cell responses, with immune responses to 99% of the vaccinated peptide antigens. SIGNIFICANCE: Predicting neoantigens in silico has progressed, but empirical testing shows that T-cell responses are more nuanced than straightforward MHC antigen recognition. The ATLAS bioassay screens tumor mutations to uncover preexisting, patient-relevant neoantigen T-cell responses and reveals a new class of putatively deleterious responses that could affect cancer immunotherapy design.This article is highlighted in the In This Issue feature, p. 521.
PMID: 33504579
ISSN: 2159-8290
CID: 4836312
Lower airway dysbiosis affects lung cancer progression
Tsay, Jun-Chieh J; Wu, Benjamin G; Sulaiman, Imran; Gershner, Katherine; Schluger, Rosemary; Li, Yonghua; Yie, Ting-An; Meyn, Peter; Olsen, Evan; Perez, Luisannay; Franca, Brendan; Carpenito, Joseph; Iizumi, Tadasu; El-Ashmawy, Mariam; Badri, Michelle; Morton, James T; Shen, Nan; He, Linchen; Michaud, Gaetane; Rafeq, Samaan; Bessich, Jamie L; Smith, Robert L; Sauthoff, Harald; Felner, Kevin; Pillai, Ray; Zavitsanou, Anastasia-Maria; Koralov, Sergei B; Mezzano, Valeria; Loomis, Cynthia A; Moreira, Andre L; Moore, William; Tsirigos, Aristotelis; Heguy, Adriana; Rom, William N; Sterman, Daniel H; Pass, Harvey I; Clemente, Jose C; Li, Huilin; Bonneau, Richard; Wong, Kwok-Kin; Papagiannakopoulos, Thales; Segal, Leopoldo N
In lung cancer, enrichment of the lower airway microbiota with oral commensals commonly occurs and ex vivo models support that some of these bacteria can trigger host transcriptomic signatures associated with carcinogenesis. Here, we show that this lower airway dysbiotic signature was more prevalent in group IIIB-IV TNM stage lung cancer and is associated with poor prognosis, as shown by decreased survival among subjects with early stage disease (I-IIIA) and worse tumor progression as measured by RECIST scores among subjects with IIIB-IV stage disease. In addition, this lower airway microbiota signature was associated with upregulation of IL-17, PI3K, MAPK and ERK pathways in airway transcriptome, and we identified Veillonella parvula as the most abundant taxon driving this association. In a KP lung cancer model, lower airway dysbiosis with V. parvula led to decreased survival, increased tumor burden, IL-17 inflammatory phenotype and activation of checkpoint inhibitor markers.
PMID: 33177060
ISSN: 2159-8290
CID: 4663012
SHP2 inhibition diminishes KRASG12C cycling and promotes tumor microenvironment remodeling
Fedele, Carmine; Li, Shuai; Teng, Kai Wen; Foster, Connor J R; Peng, David; Ran, Hao; Mita, Paolo; Geer, Mitchell J; Hattori, Takamitsu; Koide, Akiko; Wang, Yubao; Tang, Kwan Ho; Leinwand, Joshua; Wang, Wei; Diskin, Brian; Deng, Jiehui; Chen, Ting; Dolgalev, Igor; Ozerdem, Ugur; Miller, George; Koide, Shohei; Wong, Kwok-Kin; Neel, Benjamin G
KRAS is the most frequently mutated human oncogene, and KRAS inhibition has been a longtime goal. Recently, inhibitors were developed that bind KRASG12C-GDP and react with Cys-12 (G12C-Is). Using new affinity reagents to monitor KRASG12C activation and inhibitor engagement, we found that an SHP2 inhibitor (SHP2-I) increases KRAS-GDP occupancy, enhancing G12C-I efficacy. The SHP2-I abrogated RTK feedback signaling and adaptive resistance to G12C-Is in vitro, in xenografts, and in syngeneic KRASG12C-mutant pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer (NSCLC). SHP2-I/G12C-I combination evoked favorable but tumor site-specific changes in the immune microenvironment, decreasing myeloid suppressor cells, increasing CD8+ T cells, and sensitizing tumors to PD-1 blockade. Experiments using cells expressing inhibitor-resistant SHP2 showed that SHP2 inhibition in PDAC cells is required for PDAC regression and remodeling of the immune microenvironment but revealed direct inhibitory effects on tumor angiogenesis and vascularity. Our results demonstrate that SHP2-I/G12C-I combinations confer a substantial survival benefit in PDAC and NSCLC and identify additional potential combination strategies.
PMID: 33045063
ISSN: 1540-9538
CID: 4632492
Serial single-cell profiling analysis of metastatic TNBC during Nab-paclitaxel and pembrolizumab treatment
Deng, Jiehui; Thennavan, Aatish; Shah, Suhagi; Bagdatlioglu, Ece; Klar, Natalie; Heguy, Adriana; Marier, Christian; Meyn, Peter; Zhang, Yutong; Labbe, Kristen; Almonte, Christina; Krogsgaard, Michelle; Perou, Charles M; Wong, Kwok-Kin; Adams, Sylvia
PURPOSE/OBJECTIVE:Immunotherapy has recently been shown to improve outcomes for advanced PD-L1-positive triple-negative breast cancer (TNBC) in the Impassion130 trial, leading to FDA approval of the first immune checkpoint inhibitor in combination with taxane chemotherapy. To further develop predictive biomarkers and improve therapeutic efficacy of the combination, interrogation of the tumor immune microenvironment before therapy as well as during each component of treatment is crucial. Here we use single-cell RNA sequencing (scRNA-seq) on tumor biopsies to assess immune cell changes from two patients with advanced TNBC treated in a prospective trial at predefined serial time points, before treatment, on taxane chemotherapy and on chemo-immunotherapy. METHODS:Both patients (one responder and one progressor) received the trial therapy, in cycle 1 nab-paclitaxel given as single agent, in cycle 2 nab-paclitaxel in combination with pembrolizumab. Tumor core biopsies were obtained at baseline, 3 weeks (after cycle 1, chemotherapy alone) and 6 weeks (after cycle 2, chemo-immunotherapy). Single-cell RNA sequencing (scRNA-seq) of both cancer cells and infiltrating immune cells isolated were performed from fresh tumor core biopsy specimens by 10 × chromium sequencing. RESULTS:). In contrast, tumors from the patient with rapid disease progression showed a prevalent and persistent myeloid compartment. CONCLUSIONS:Our study provides a deep cellular analysis of on-treatment changes during chemo-immunotherapy for advanced TNBC, demonstrating not only feasibility of single-cell analyses on serial tumor biopsies but also the heterogeneity of TNBC and differences in on-treatment changes in responder versus progressor.
PMID: 32949350
ISSN: 1573-7217
CID: 4605282
MUC1-C integrates activation of the IFN-γ pathway with suppression of the tumor immune microenvironment in triple-negative breast cancer
Yamashita, Nami; Long, Mark; Fushimi, Atsushi; Yamamoto, Masaaki; Hata, Tsuyoshi; Hagiwara, Masayuki; Bhattacharya, Atrayee; Hu, Qiang; Wong, Kwok-Kin; Liu, Song; Kufe, Donald
BACKGROUND:Immune checkpoint inhibitors (ICIs) have had a profound impact on the treatment of many tumors; however, their effectiveness against triple-negative breast cancers (TNBCs) has been limited. One factor limiting responsiveness of TNBCs to ICIs is a lack of functional tumor-infiltrating lymphocytes (TILs) in 'non-inflamed' or 'cold' tumor immune microenvironments (TIMEs), although by unknown mechanisms. Targeting MUC1-C in a mouse transgenic TNBC tumor model increases cytotoxic tumor-infiltrating CD8+ T cells (CTLs), supporting a role for MUC1-C in immune evasion. The basis for these findings and whether they extend to human TNBCs are not known. METHODS:Human TNBC cells silenced for MUC1-C using short hairpin RNAs (shRNAs) were analyzed for the effects of MUC1-C on global transcriptional profiles. Differential expression and rank order analysis was used for gene set enrichment analysis (GSEA). Gene expression was confirmed by quantitative reverse-transcription PCR and immunoblotting. The The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets were analyzed for effects of MUC1 on GSEA, cell-type enrichment, and tumor immune dysfunction and exclusion. Single-cell scRNA-seq datasets of TNBC samples were analyzed for normalized expression associations between MUC1 and selected genes within tumor cells. RESULTS:Our results demonstrate that MUC1-C is a master regulator of the TNBC transcriptome and that MUC1-C-induced gene expression is driven by STAT1 and IRF1. We found that MUC1-C activates the inflammatory interferon (IFN)-γ-driven JAK1→STAT1→IRF1 pathway and induces the IDO1 and COX2/PTGS2 effectors, which play key roles in immunosuppression. Involvement of MUC1-C in activating the immunosuppressive IFN-γ pathway was extended by analysis of human bulk and scRNA-seq datasets. We further demonstrate that MUC1 associates with the depletion and dysfunction of CD8+ T cells in the TNBC TIME. CONCLUSIONS:These findings demonstrate that MUC1-C integrates activation of the immunosuppressive IFN-γ pathway with depletion of TILs in the TNBC TIME and provide support for MUC1-C as a potential target for improving TNBC treatment alone and in combination with ICIs. Of translational significance, MUC1-C is a druggable target with chimeric antigen receptor (CAR) T cells, antibody-drug conjugates (ADCs) and a functional inhibitor that are under clinical development.
PMCID:7839859
PMID: 33495298
ISSN: 2051-1426
CID: 4798872
Multiple screening approaches reveal HDAC6 as a novel regulator of glycolytic metabolism in triple-negative breast cancer
Dowling, CatrÃona M; Hollinshead, Kate E R; Di Grande, Alessandra; Pritchard, Justin; Zhang, Hua; Dillon, Eugene T; Haley, Kathryn; Papadopoulos, Eleni; Mehta, Anita K; Bleach, Rachel; Lindner, Andreas U; Mooney, Brian; Düssmann, Heiko; O'Connor, Darran; Prehn, Jochen H M; Wynne, Kieran; Hemann, Michael; Bradner, James E; Kimmelman, Alec C; Guerriero, Jennifer L; Cagney, Gerard; Wong, Kwok-Kin; Letai, Anthony G; Chonghaile, TrÃona NÃ
Triple-negative breast cancer (TNBC) is a subtype of breast cancer without a targeted form of therapy. Unfortunately, up to 70% of patients with TNBC develop resistance to treatment. A known contributor to chemoresistance is dysfunctional mitochondrial apoptosis signaling. We set up a phenotypic small-molecule screen to reveal vulnerabilities in TNBC cells that were independent of mitochondrial apoptosis. Using a functional genetic approach, we identified that a "hit" compound, BAS-2, had a potentially similar mechanism of action to histone deacetylase inhibitors (HDAC). An in vitro HDAC inhibitor assay confirmed that the compound selectively inhibited HDAC6. Using state-of-the-art acetylome mass spectrometry, we identified glycolytic substrates of HDAC6 in TNBC cells. We confirmed that inhibition or knockout of HDAC6 reduced glycolytic metabolism both in vitro and in vivo. Through a series of unbiased screening approaches, we have identified a previously unidentified role for HDAC6 in regulating glycolytic metabolism.
PMCID:7810372
PMID: 33523897
ISSN: 2375-2548
CID: 4775962
Shining a light on metabolic vulnerabilities in non-small cell lung cancer
Dowling, CatrÃona M; Zhang, Hua; Chonghaile, TrÃona NÃ; Wong, Kwok-Kin
Metabolic reprogramming is a hallmark of cancer which contributes to essential processes required for cell survival, growth, and proliferation. Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and its genomic classification has given rise to the design of therapies targeting tumors harboring specific gene alterations that cause aberrant signaling. Lung tumors are characterized with having high glucose and lactate use, and high heterogeneity in their metabolic pathways. Here we review how NSCLC cells with distinct mutations reprogram their metabolic pathways and highlight the potential metabolic vulnerabilities that might lead to the development of novel therapeutic strategies.
PMID: 33130228
ISSN: 1879-2561
CID: 4684092