168 results on '"Conejo-Garcia JR"'
Search Results
2. Tumour-intrinsic PDL1 signals regulate the Chk2 DNA damage response in cancer cells and mediate resistance to Chk1 inhibitors.
- Author
-
Murray CE, Kornepati AVR, Ontiveros C, Liao Y, de la Peña Avalos B, Rogers CM, Liu Z, Deng Y, Bai H, Kari S, Padron AS, Boyd JT, Reyes R, Clark CA, Svatek RS, Li R, Hu Y, Wang M, Conejo-Garcia JR, Byers LA, Ramkumar K, Sood AK, Lee JM, Burd CE, Vadlamudi RK, Gupta HB, Zhao W, Dray E, Sung P, and Curiel TJ
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Neoplasms metabolism, Neoplasms genetics, Neoplasms drug therapy, Neoplasms pathology, Signal Transduction drug effects, Checkpoint Kinase 1 metabolism, DNA Damage, Checkpoint Kinase 2 metabolism, Checkpoint Kinase 2 genetics, B7-H1 Antigen metabolism, B7-H1 Antigen genetics, Drug Resistance, Neoplasm genetics, Protein Kinase Inhibitors pharmacology
- Abstract
Background: Aside from the canonical role of PDL1 as a tumour surface-expressed immune checkpoint molecule, tumour-intrinsic PDL1 signals regulate non-canonical immunopathological pathways mediating treatment resistance whose significance, mechanisms, and therapeutic targeting remain incompletely understood. Recent reports implicate tumour-intrinsic PDL1 signals in the DNA damage response (DDR), including promoting homologous recombination DNA damage repair and mRNA stability of DDR proteins, but many mechanistic details remain undefined., Methods: We genetically depleted PDL1 from transplantable mouse and human cancer cell lines to understand consequences of tumour-intrinsic PDL1 signals in the DNA damage response. We complemented this work with studies of primary human tumours and inducible mouse tumours. We developed novel approaches to show tumour-intrinsic PDL1 signals in specific subcellular locations. We pharmacologically depleted tumour PDL1 in vivo in mouse models with repurposed FDA-approved drugs for proof-of-concept clinical translation studies., Results: We show that tumour-intrinsic PDL1 promotes the checkpoint kinase-2 (Chk2)-mediated DNA damage response. Intracellular but not surface-expressed PDL1 controlled Chk2 protein content post-translationally and independently of PD1 by antagonising PIRH2 E3 ligase-mediated Chk2 polyubiquitination and protein degradation. Genetic tumour PDL1 depletion specifically reduced tumour Chk2 content but not ATM, ATR, or Chk1 DDR proteins, enhanced Chk1 inhibitor (Chk1i) synthetic lethality in vitro in diverse human and murine tumour models, and improved Chk1i efficacy in vivo. Pharmacologic tumour PDL1 depletion with cefepime or ceftazidime replicated genetic tumour PDL1 depletion by reducing tumour Chk2, inducing Chk1i synthetic lethality in a tumour PDL1-dependent manner, and reducing in vivo tumour growth when combined with Chk1i., Conclusions: Our data challenge the prevailing surface PDL1 paradigm, elucidate important and previously unappreciated roles for tumour-intrinsic PDL1 in regulating the ATM/Chk2 DNA damage response axis and E3 ligase-mediated protein degradation, suggest tumour PDL1 as a biomarker for Chk1i efficacy, and support the rapid clinical potential of pharmacologic tumour PDL1 depletion to treat selected cancers., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
3. Transgelin 2 guards T cell lipid metabolism and antitumour function.
- Author
-
Hwang SM, Awasthi D, Jeong J, Sandoval TA, Chae CS, Ramos Y, Tan C, Marin Falco M, Salvagno C, Emmanuelli A, McBain IT, Mishra B, Ivashkiv LB, Zamarin D, Cantillo E, Chapman-Davis E, Holcomb K, Morales DK, Yu X, Rodriguez PC, Conejo-Garcia JR, Kaczocha M, Vähärautio A, Song M, and Cubillos-Ruiz JR
- Abstract
Mounting effective immunity against pathogens and tumours relies on the successful metabolic programming of T cells by extracellular fatty acids
1-3 . Fatty-acid-binding protein 5 (FABP5) has a key role in this process by coordinating the efficient import and trafficking of lipids that fuel mitochondrial respiration to sustain the bioenergetic requirements of protective CD8+ T cells4,5 . However, the mechanisms that govern this immunometabolic axis remain unexplored. Here we report that the cytoskeletal organizer transgelin 2 (TAGLN2) is necessary for optimal fatty acid uptake, mitochondrial respiration and anticancer function in CD8+ T cells. TAGLN2 interacts with FABP5 to facilitate its cell surface localization and function in activated CD8+ T cells. Analyses of ovarian cancer specimens revealed that endoplasmic reticulum (ER) stress responses induced by the tumour microenvironment repress TAGLN2 in infiltrating CD8+ T cells, thereby enforcing their dysfunctional state. Restoring TAGLN2 expression in ER-stressed CD8+ T cells increased their lipid uptake, mitochondrial respiration and cytotoxic capacity. Accordingly, chimeric antigen receptor T cells overexpressing TAGLN2 bypassed the detrimental effects of tumour-induced ER stress and demonstrated therapeutic efficacy in mice with metastatic ovarian cancer. Our study establishes the role of cytoskeletal TAGLN2 in T cell lipid metabolism and highlights the potential to enhance cellular immunotherapy in solid malignancies by preserving the TAGLN2-FABP5 axis., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
4. Pan-cancer γδ TCR analysis uncovers clonotype diversity and prognostic potential.
- Author
-
Yu X, Song L, Cen L, Cao B, Tao R, Shen Y, Abate-Daga D, Rodriguez PC, Conejo-Garcia JR, and Wang X
- Subjects
- Humans, Prognosis, Clone Cells, Intraepithelial Lymphocytes immunology, Intraepithelial Lymphocytes metabolism, Gene Expression Regulation, Neoplastic, T-Lymphocytes immunology, T-Lymphocytes metabolism, Receptors, Antigen, T-Cell, gamma-delta genetics, Receptors, Antigen, T-Cell, gamma-delta immunology, Receptors, Antigen, T-Cell, gamma-delta metabolism, Neoplasms immunology, Neoplasms genetics
- Abstract
Gamma-delta T cells (γδ T cells) play a crucial role in both innate and adaptive immunity within tumors, yet their presence and prognostic value in cancer remain underexplored. This study presents a large-scale analysis of γδ T cell receptor (γδ TCR) reads from 11,000 tumor samples spanning 33 cancer types, utilizing the TRUST4 algorithm. Our findings reveal extensive diversity in γδ TCR clonality and gene expression, underscoring the potential of γδ T cells as prognostic biomarkers in various cancers. We further demonstrate the utility of TCR gamma (TRG) and delta (TRD) gene expression from standard RNA-sequencing (RNA-seq) data. This comprehensive dataset offers a valuable resource for advancing γδ T cell research, with implications for enhanced immunotherapy approaches or alternative therapeutic strategies. Additionally, our centralized database supports translational research into the therapeutic significance of γδ T cells., Competing Interests: Declaration of interests J.R.C.-G. is a consultant for Anixa Biosciences and Alloy Therapeutics; holds stock options in Compass Therapeutics, Anixa Biosciences, and Alloy Therapeutics; has a patent licensed by Anixa Biosciences; owns intellectual property filed with Compass Therapeutics; and is a co-founder of Cellepus Therapeutics, a CAR T cell company., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Unraveling spontaneous humoral immune responses against human cancer: A road to novel immunotherapies.
- Author
-
Conejo-Garcia JR, Lopez-Bailon LU, and Anadon CM
- Abstract
In immuno-oncology, the focus has traditionally been on αβ T cells, and immune checkpoint inhibitors that primarily target PD-1 or CTLA4 in these lymphocytes have revolutionized the management of multiple human malignancies. However, recent research highlights the crucial role of B cells and the antibodies they produce in antagonizing malignant progression, offering new avenues for immunotherapy. Our group has demonstrated that dimeric IgA can penetrate tumor cells, neutralize oncogenic drivers in endosomes, and expel them from the cytosol. This mechanistic insight suggests that engineered antibodies targeting this pathway may effectively reach previously inaccessible targets. Investigating antibody production within intratumoral germinal centers and understanding the impact of different immunoglobulins on malignant progression could furnish new tools for the therapeutic arsenal, including the development of tumor-penetrating antibodies. This review aims to elucidate the nature of humoral adaptive immune responses in human cancer and explore how they could herald a new era of immunotherapeutic modalities. By expanding the scope of anti-tumor immunotherapies, these approaches have the potential to benefit a broader range of cancer patients, particularly through the utilization of tumor cell-penetrating antibodies., (© The Author(s) 2024. Published by Oxford University Press on behalf of Society for Leukocyte Biology. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
6. CGPA: multi-context insights from the cancer gene prognosis atlas.
- Author
-
Cao B, Yu X, Gonzalez G, Murthy AR, Li T, Shen Y, Yao S, Conejo-Garcia JR, Jiang P, and Wang X
- Abstract
Cancer transcriptomic data are used extensively to interrogate the prognostic value of targeted genes, yet basic scientists and clinicians have predominantly relied on univariable survival analysis for this purpose. This method often fails to capture the full prognostic potential and contextual relevance of the genes under study, inadvertently omitting a group of genes we term univariable missed-opportunity prognostic (UMOP) genes. Recognizing the complexity of revealing multifaceted prognostic implications, especially when extending the analysis to include various covariates and thresholds, we present the Cancer Gene Prognosis Atlas (CGPA). This platform greatly enhances gene-centric biomarker research across cancer types by offering an interactive and user-friendly interface for highly customized, in-depth prognostic analysis. CGPA notably supports data-driven exploration of gene pairs and gene-hallmark relationships, elucidating key composite biological mechanisms like synthetic lethality and immunosuppression. It further expands its capabilities to assess multi-gene panels using both public and user-provided data, facilitating a seamless mechanism-to-machine analysis. Additionally, CGPA features a designated portal for discovering prognostic gene modules using curated cancer immunotherapy data. Ultimately, CGPA's comprehensive, accessible tools allow cancer researchers, including those without statistical expertise, to precisely investigate the prognostic landscape of genes, customizing the model to fit specific research hypotheses and enhancing biomarker discovery and validation through a synergy of mechanistic and data-driven strategies., Competing Interests: Conflict of interest: The authors declare no potential conflicts of interest.
- Published
- 2024
- Full Text
- View/download PDF
7. A pan-cancer gamma delta T cell repertoire.
- Author
-
Yu X, Song L, Cen L, Cao B, Tao R, Shen Y, Abate-Daga D, Rodriguez PC, Conejo-Garcia JR, and Wang X
- Abstract
This report presents the largest collection of gamma-delta T cell receptor (γδ TCR) reads in human cancer to date, analyzing about 11,000 patient tumor samples across 33 cancer types using the TRUST4 algorithm. Despite γδ T cells being a small fraction of the T cell population, they play a key role in both innate and adaptive immunity. Our comprehensive analysis reveals their significant presence across all cancer types, specifically highlighting the diverse spectrum and clonality patterns of their γδ receptors. This research highlights the complex roles of γδ T cells in tumor tissues and their potential as prognostic biomarkers. We also demonstrate the utility of T cell receptor gamma (TRG) and delta (TRD) gene expression values from standard RNA-seq data. Ultimately, our work establishes a fundamental resource for future tumor-infiltrating γδ T cell research and may facilitate the development of novel γδ-T-cell-based therapeutic strategies. Together, we demonstrate the strong diversity and prognostic potential of γδ T cells in multiple cancer types.
- Published
- 2024
- Full Text
- View/download PDF
8. Orthotopic Models Using New, Murine Lung Adenocarcinoma Cell Lines Simulate Human Non-Small Cell Lung Cancer Treated with Immunotherapy.
- Author
-
Knott EP, Kim EY, Kim EQ, Freire R, Medina JA, Wang Y, Chen CB, Wu C, Wangpaichitr M, Conejo-Garcia JR, and Lim DC
- Subjects
- Animals, Humans, Cell Line, Tumor, Mice, Disease Models, Animal, Female, Immunotherapy methods, Lung Neoplasms immunology, Lung Neoplasms therapy, Lung Neoplasms pathology, Lung Neoplasms genetics, Carcinoma, Non-Small-Cell Lung therapy, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung genetics, Adenocarcinoma of Lung immunology, Adenocarcinoma of Lung therapy, Adenocarcinoma of Lung pathology, Adenocarcinoma of Lung genetics
- Abstract
Understanding tumor-host immune interactions and the mechanisms of lung cancer response to immunotherapy is crucial. Current preclinical models used to study this often fall short of capturing the complexities of human lung cancer and lead to inconclusive results. To bridge the gap, we introduce two new murine monoclonal lung cancer cell lines for use in immunocompetent orthotopic models. We demonstrate how our cell lines exhibit immunohistochemical protein expression (TTF-1, NapA, PD-L1) and common driver mutations (KRAS, p53, and p110α) seen in human lung adenocarcinoma patients, and how our orthotopic models respond to combination immunotherapy in vivo in a way that closely mirrors current clinical outcomes. These new lung adenocarcinoma cell lines provide an invaluable, clinically relevant platform for investigating the intricate dynamics between tumor and the immune system, and thus potentially contributes to a deeper understanding of immunotherapeutic approaches to lung cancer treatment.
- Published
- 2024
- Full Text
- View/download PDF
9. Lifetime Exposure to Cigarette Smoke, B-Cell Tumor Immune Infiltration, and Immunoglobulin Abundance in Ovarian Tumors.
- Author
-
Hathaway CA, Townsend MK, Wang T, Vinci C, Jake-Schoffman DE, Hecht JL, Saeed-Vafa D, Moran Segura C, Nguyen JV, Conejo-Garcia JR, Fridley BL, and Tworoger SS
- Subjects
- Humans, Female, Middle Aged, Adult, B-Lymphocytes immunology, Immunoglobulins blood, Lymphocytes, Tumor-Infiltrating immunology, Aged, Cigarette Smoking adverse effects, Cigarette Smoking immunology, Ovarian Neoplasms immunology, Ovarian Neoplasms pathology, Ovarian Neoplasms epidemiology
- Abstract
Background: Cigarette smoke exposure has been linked to systemic immune dysfunction, including for B-cell and immunoglobulin (Ig) production, and poor outcomes in patients with ovarian cancer. No study has evaluated the impact of smoke exposure across the life-course on B-cell infiltration and Ig abundance in ovarian tumors., Methods: We measured markers of B and plasma cells and Ig isotypes using multiplex immunofluorescence on 395 ovarian cancer tumors in the Nurses' Health Study (NHS)/NHSII. We conducted beta-binomial analyses evaluating odds ratios (OR) and 95% confidence intervals (CI) for positivity of immune markers by cigarette exposure among cases and Cox proportional hazards models to evaluate hazard ratios (HR) and 95% CI for developing tumors with low (
- Published
- 2024
- Full Text
- View/download PDF
10. Jagged2 targeting in lung cancer activates anti-tumor immunity via Notch-induced functional reprogramming of tumor-associated macrophages.
- Author
-
Mandula JK, Sierra-Mondragon RA, Jimenez RV, Chang D, Mohamed E, Chang S, Vazquez-Martinez JA, Cao Y, Anadon CM, Lee SB, Das S, Rocha-Munguba L, Pham VM, Li R, Tarhini AA, Furqan M, Dalton W, Churchman M, Moran-Segura CM, Nguyen J, Perez B, Kojetin DJ, Obermayer A, Yu X, Chen A, Shaw TI, Conejo-Garcia JR, and Rodriguez PC
- Subjects
- Animals, Humans, Mice, Calcium-Binding Proteins metabolism, Calcium-Binding Proteins genetics, Cell Line, Tumor, Jagged-1 Protein metabolism, Jagged-1 Protein genetics, Macrophages immunology, Macrophages metabolism, Mice, Inbred C57BL, Receptor, Notch1 metabolism, Receptor, Notch1 genetics, Receptors, Notch metabolism, Signal Transduction, Tumor Escape immunology, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung metabolism, Interferon Regulatory Factors metabolism, Interferon Regulatory Factors genetics, Jagged-2 Protein metabolism, Jagged-2 Protein genetics, Jagged-2 Protein immunology, Lung Neoplasms immunology, Lung Neoplasms pathology, Lung Neoplasms genetics, Mice, Knockout, Tumor-Associated Macrophages immunology, Tumor-Associated Macrophages metabolism
- Abstract
Signaling through Notch receptors intrinsically regulates tumor cell development and growth. Here, we studied the role of the Notch ligand Jagged2 on immune evasion in non-small cell lung cancer (NSCLC). Higher expression of JAG2 in NSCLC negatively correlated with survival. In NSCLC pre-clinical models, deletion of Jag2, but not Jag1, in cancer cells attenuated tumor growth and activated protective anti-tumor T cell responses. Jag2
-/- lung tumors exhibited higher frequencies of macrophages that expressed immunostimulatory mediators and triggered T cell-dependent anti-tumor immunity. Mechanistically, Jag2 ablation promoted Nr4a-mediated induction of Notch ligands DLL1/4 on cancer cells. DLL1/4-initiated Notch1/2 signaling in macrophages induced the expression of transcription factor IRF4 and macrophage immunostimulatory functionality. IRF4 expression was required for the anti-tumor effects of Jag2 deletion in lung tumors. Antibody targeting of Jagged2 inhibited tumor growth and activated IRF4-driven macrophage-mediated anti-tumor immunity. Thus, Jagged2 orchestrates immunosuppressive systems in NSCLC that can be overcome to incite macrophage-mediated anti-tumor immunity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
11. Glucose-driven histone lactylation promotes the immunosuppressive activity of monocyte-derived macrophages in glioblastoma.
- Author
-
De Leo A, Ugolini A, Yu X, Scirocchi F, Scocozza D, Peixoto B, Pace A, D'Angelo L, Liu JKC, Etame AB, Rughetti A, Nuti M, Santoro A, Vogelbaum MA, Conejo-Garcia JR, Rodriguez PC, and Veglia F
- Subjects
- Animals, Mice, Humans, Cell Line, Tumor, Brain Neoplasms immunology, Brain Neoplasms metabolism, Brain Neoplasms pathology, Glucose Transporter Type 1 metabolism, Glucose Transporter Type 1 genetics, Interleukin-10 metabolism, Glycolysis, Microglia metabolism, Microglia immunology, Mice, Inbred C57BL, T-Lymphocytes immunology, T-Lymphocytes metabolism, Immune Tolerance, Glioblastoma immunology, Glioblastoma metabolism, Glioblastoma pathology, Histones metabolism, Macrophages immunology, Macrophages metabolism, Glucose metabolism
- Abstract
Immunosuppressive macrophages restrict anti-cancer immunity in glioblastoma (GBM). Here, we studied the contribution of microglia (MGs) and monocyte-derived macrophages (MDMs) to immunosuppression and mechanisms underlying their regulatory function. MDMs outnumbered MGs at late tumor stages and suppressed T cell activity. Molecular and functional analysis identified a population of glycolytic MDM expressing GLUT1 with potent immunosuppressive activity. GBM-derived factors promoted high glycolysis, lactate, and interleukin-10 (IL-10) production in MDMs. Inhibition of glycolysis or lactate production in MDMs impaired IL-10 expression and T cell suppression. Mechanistically, intracellular lactate-driven histone lactylation promoted IL-10 expression, which was required to suppress T cell activity. GLUT1 expression on MDMs was induced downstream of tumor-derived factors that activated the PERK-ATF4 axis. PERK deletion in MDM abrogated histone lactylation, led to the accumulation of intratumoral T cells and tumor growth delay, and, in combination with immunotherapy, blocked GBM progression. Thus, PERK-driven glucose metabolism promotes MDM immunosuppressive activity via histone lactylation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Introduction to the special issue: B cells in cancer immunosurveillance.
- Author
-
Conejo-Garcia JR and Rodriguez PC
- Subjects
- Humans, Monitoring, Immunologic, Killer Cells, Natural, B-Lymphocytes, Neoplasms therapy
- Published
- 2024
- Full Text
- View/download PDF
13. Harnessing γδ T Cells against Human Gynecologic Cancers.
- Author
-
Conejo-Garcia JR, Anadon CM, Lopez-Bailon LU, and Chaurio RA
- Abstract
Immuno-oncology has traditionally focused on conventional MHC-restricted αβ T cells. Yet, unconventional γδ T cells, which kill tumor cells in an MHC-unrestricted manner, display characteristics of effector activity and stemness without exhaustion and are nearly universally observed in human gynecologic malignancies, correlating with improved outcomes. These cells do not have a clear counterpart in mice but are also found in the healthy female reproductive tract. Interventions that modulate their in vivo activity, or cellular therapies utilizing γδ T cells as an allogeneic, "off-the-shelf" platform (e.g., for chimeric antigen receptor expression) hold significant potential against challenging tumors like ovarian cancer, which has been stubbornly resistant to the immune checkpoint inhibitors that change the landscape of other human tumors. Here, we discuss recent discoveries on the specific populations of γδ T cells that infiltrate human gynecologic cancers, their anti-tumor activity, and the prospect of redirecting their effector function against tumor cells to develop a new generation of immunotherapies that extends beyond the traditional αβ T cell-centric view of the field.
- Published
- 2024
- Full Text
- View/download PDF
14. WITHDRAWN: Impact of spatial clustering of cytotoxic and tumor infiltrating lymphocytes on overall survival in women with high grade serous ovarian cancer.
- Author
-
Soupir AC, Townsend MK, Hathaway CA, Nguyen J, Moran Segura C, Saeed-Vafa D, Ospina OE, Peres LC, Conejo-Garcia JR, Terry KL, Tworoger SS, and Fridley BL
- Abstract
The authors have withdrawn their manuscript owing to incorrect handling of multiple measures in the survival analyses. Therefore, the authors do not wish this work to be cited as reference for the project. If you have any questions, please contact the corresponding author.
- Published
- 2024
- Full Text
- View/download PDF
15. Antibodies target intracellular oncodrivers through PIGR-mediated transcytosis.
- Author
-
Biswas S, Anadon CM, and Conejo-Garcia JR
- Subjects
- Immunoglobulin A, Antibodies, Transcytosis
- Published
- 2024
- Full Text
- View/download PDF
16. Immune landscape in molecular subtypes of human papillomavirus-negative head and neck cancer.
- Author
-
Xie M, Chaudhary R, Slebos RJC, Lee K, Song F, Poole MI, Hoening DS, Noel LC, Hernandez-Prera JC, Conejo-Garcia JR, Chung CH, and Tan AC
- Subjects
- Humans, Squamous Cell Carcinoma of Head and Neck genetics, Human Papillomavirus Viruses, Immunotherapy, Tumor Microenvironment, Papillomavirus Infections complications, Papillomavirus Infections genetics, Head and Neck Neoplasms genetics
- Abstract
Head and neck squamous cell carcinomas (HNSCC) remain a poorly understood disease clinically and immunologically. HPV is a known risk factor of HNSCC associated with better outcome, whereas HPV-negative HNSCC are more heterogeneous in outcome. Gene expression signatures have been developed to classify HNSCC into four molecular subtypes (classical, basal, mesenchymal, and atypical). However, the molecular underpinnings of treatment response and the immune landscape for these molecular subtypes are largely unknown. Herein, we described a comprehensive immune landscape analysis in three independent HNSCC cohorts (>700 patients) using transcriptomics data. We assigned the HPV
- HNSCC patients into these four molecular subtypes and characterized the tumor microenvironment using deconvolution method. We determined that atypical and mesenchymal subtypes have greater immune enrichment and exhibit a T-cell exhaustion phenotype, compared to classical and basal subtypes. Further analyses revealed different B cell maturation and antibody isotypes enrichment patterns, and distinct immune microenvironment crosstalk in the atypical and mesenchymal subtypes. Taken together, our study suggests that treatments that enhances B cell activity may benefit patients with HNSCC of the atypical subtypes. The rationale can be utilized in the design of future precision immunotherapy trials based on the molecular subtypes of HPV- HNSCC., (© 2023 The Authors. Molecular Carcinogenesis published by Wiley Periodicals LLC.)- Published
- 2024
- Full Text
- View/download PDF
17. Transgelin 2 guards T cell lipid metabolic programming and anti-tumor function.
- Author
-
Hwang SM, Awasthi D, Jeong J, Sandoval TA, Chae CS, Ramos Y, Tan C, Falco MM, McBain IT, Mishra B, Ivashkiv LB, Zamarin D, Cantillo E, Chapman-Davis E, Holcomb K, Morales DK, Rodriguez PC, Conejo-Garcia JR, Kaczocha M, Vähärautio A, Song M, and Cubillos-Ruiz JR
- Abstract
Mounting effective immunity against pathogens and tumors relies on the successful metabolic programming of T cells by extracellular fatty acids
1-3 . During this process, fatty-acid-binding protein 5 (FABP5) imports lipids that fuel mitochondrial respiration and sustain the bioenergetic requirements of protective CD8+ T cells4,5 . Importantly, however, the mechanisms governing this crucial immunometabolic axis remain unexplored. Here we report that the cytoskeletal organizer Transgelin 2 (TAGLN2) is necessary for optimal CD8+ T cell fatty acid uptake, mitochondrial respiration, and anti-cancer function. We found that TAGLN2 interacts with FABP5, enabling the surface localization of this lipid importer on activated CD8+ T cells. Analysis of ovarian cancer specimens revealed that endoplasmic reticulum (ER) stress responses elicited by the tumor microenvironment repress TAGLN2 in infiltrating CD8+ T cells, enforcing their dysfunctional state. Restoring TAGLN2 expression in ER-stressed CD8+ T cells bolstered their lipid uptake, mitochondrial respiration, and cytotoxic capacity. Accordingly, chimeric antigen receptor T cells overexpressing TAGLN2 bypassed the detrimental effects of tumor-induced ER stress and demonstrated superior therapeutic efficacy in mice with metastatic ovarian cancer. Our study unveils the role of cytoskeletal TAGLN2 in T cell lipid metabolism and highlights the potential to enhance cellular immunotherapy in solid malignancies by preserving the TAGLN2-FABP5 axis., Competing Interests: COMPETING INTERESTS / DISCLOSURES J.R.C.-R. holds patents on the use immune modulators for OvCa treatment and serves as scientific consultant for Moderna, Immagene B.V., and Autoimmunity Biologic Solutions, Inc. D.Z. reports institutional grants from Merck, Genentech, AstraZeneca, Plexxikon, and Synthekine, and personal fees from AstraZeneca, Xencor, Memgen, Takeda, Synthekine, Immunos, Tessa Therapeutics, Miltenyi, and Calidi Biotherapeutics. D.Z. owns a patent on use of oncolytic Newcastle Disease Virus for cancer therapy. J.R.C.-G. has stock options in Compass Therapeutics, Anixa Biosciences, and Alloy Therapeutics; receives licensing fees from Anixa Biosciences for the patent of FSHCER T cells; receives honorarium from Alloy Therapeutics; and intellectual property with Compass Therapeutics and Anixa Biosciences; and is co-founder of Cellepus Therapeutics, a company that develops allogeneic gamma/delta CAR T cells. All other authors declare no potential conflicts of interest.- Published
- 2023
- Full Text
- View/download PDF
18. Targeting intracellular oncoproteins with dimeric IgA promotes expulsion from the cytoplasm and immune-mediated control of epithelial cancers.
- Author
-
Biswas S, Mandal G, Anadon CM, Chaurio RA, Lopez-Bailon LU, Nagy MZ, Mine JA, Hänggi K, Sprenger KB, Innamarato P, Harro CM, Powers JJ, Johnson J, Fang B, Eysha M, Nan X, Li R, Perez BA, Curiel TJ, Yu X, Rodriguez PC, and Conejo-Garcia JR
- Subjects
- Humans, CD8-Positive T-Lymphocytes metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Cytoplasm metabolism, Immunoglobulin A metabolism, Carcinoma
- Abstract
Dimeric IgA (dIgA) can move through cells via the IgA/IgM polymeric immunoglobulin receptor (PIGR), which is expressed mainly on mucosal epithelia. Here, we studied the ability of dIgA to target commonly mutated cytoplasmic oncodrivers. Mutation-specific dIgA, but not IgG, neutralized KRAS
G12D within ovarian carcinoma cells and expelled this oncodriver from tumor cells. dIgA binding changed endosomal trafficking of KRASG12D from accumulation in recycling endosomes to aggregation in the early/late endosomes through which dIgA transcytoses. dIgA targeting of KRASG12D abrogated tumor cell proliferation in cell culture assays. In vivo, KRASG12D -specific dIgA1 limited the growth of KRASG12D -mutated ovarian and lung carcinomas in a manner dependent on CD8+ T cells. dIgA specific for IDH1R132H reduced colon cancer growth, demonstrating effective targeting of a cytoplasmic oncodriver not associated with surface receptors. dIgA targeting of KRASG12D restricted tumor growth more effectively than small-molecule KRASG12D inhibitors, supporting the potential of this approach for the treatment of human cancers., Competing Interests: Declaration of interests J.R.C.-G. has stock options in Compass Therapeutics, Anixa Biosciences, and Alloy Therapeutics; has sponsored research with Anixa Biosciences; receives honorarium from Alloy Therapeutics; and has intellectual property with Compass Therapeutics and Anixa Biosciences; all outside the submitted work. J.R.C.-G. and S.B. have filed a patent application for intracellular targeting of oncodrivers using dIgA. B.A.P. has completed Advisory Board with AstraZeneca and has Research Support from BMS. R.L.: clinical trial protocol committee—CG Oncology; Scientific adviser/consultant—BMS, Ferring, Fergene, Arquer Diagnostics., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
19. The relationship of lifetime history of depression on the ovarian tumor immune microenvironment.
- Author
-
Hathaway CA, Townsend MK, Conejo-Garcia JR, Fridley BL, Moran Segura C, Nguyen JV, Armaiz-Pena GN, Sasamoto N, Saeed-Vafa D, Terry KL, Kubzansky LD, and Tworoger SS
- Subjects
- Female, Humans, Case-Control Studies, Depression, Biomarkers, Tumor Microenvironment, Ovarian Neoplasms pathology, Carcinoma
- Abstract
Background: Depression is associated with a higher ovarian cancer risk. Prior work suggests that depression can lead to systemic immune suppression, which could potentially alter the anti-tumor immune response., Methods: We evaluated the association of pre-diagnosis depression with features of the anti-tumor immune response, including T and B cells and immunoglobulins, among women with ovarian tumor tissue collected in three studies, the Nurses' Health Study (NHS; n = 237), NHSII (n = 137) and New England Case-Control Study (NECC; n = 215). Women reporting depressive symptoms above a clinically relevant cut-point, antidepressant use, or physician diagnosis of depression at any time prior to diagnosis of ovarian cancer were considered to have pre-diagnosis depression. Multiplex immunofluorescence was performed on tumor tissue microarrays to measure immune cell infiltration. In pooled analyses, we estimated odds ratios (OR) and 95% confidence intervals (CI) for the positivity of tumor immune cells using a beta-binomial model comparing those with and without depression. We used Bonferroni corrections to adjust for multiple comparisons., Results: We observed no statistically significant association between depression status and any immune markers at the Bonferroni corrected p-value of 0.0045; however, several immune markers were significant at a nominal p-value of 0.05. Specifically, there were increased odds of having recently activated cytotoxic (CD3
+ CD8+ CD69+ ) and exhausted-like T cells (CD3+ Lag3+ ) in tumors of women with vs. without depression (OR = 1.36, 95 %CI = 1.09-1.69 and OR = 1.24, 95 %CI = 1.01-1.53, respectively). Associations were comparable when considering high grade serous tumors only (comparable ORs = 1.33, 95 %CI = 1.05-1.69 and OR = 1.25, 95 %CI = 0.99-1.58, respectively). There were decreased odds of having tumor infiltrating plasma cells (CD138+ ) in women with vs. without depression (OR = 0.54, 95 %CI = 0.33-0.90), which was similar among high grade serous carcinomas, although not statistically significant. Depression was also related to decreased odds of having naïve and memory B cells (CD20+ : OR = 0.54, 95 %CI = 0.30-0.98) and increased odds of IgG (OR = 1.22, 95 %CI = 0.97-1.53) in high grade serous carcinomas., Conclusion: Our results provide suggestive evidence that depression may influence ovarian cancer outcomes through changes in the tumor immune microenvironment, including increasing T cell activation and exhaustion and reducing antibody-producing B cells. Further studies with clinical measures of depression and larger samples are needed to confirm these results., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
20. A Phase II Study of Durvalumab for Bacillus Calmette-Guerin (BCG) Unresponsive Urothelial Carcinoma In Situ of the Bladder.
- Author
-
Li R, Sexton WJ, Dhillon J, Berglund A, Naidu S, Borjas G, Rose K, Kim Y, Wang X, Conejo-Garcia JR, Jain RK, Poch MA, Spiess PE, Pow-Sang J, Gilbert SM, and Zhang J
- Subjects
- Humans, BCG Vaccine adverse effects, Urinary Bladder pathology, Antibodies, Monoclonal adverse effects, Neoplasm Invasiveness, Administration, Intravesical, Adjuvants, Immunologic therapeutic use, Neoplasm Recurrence, Local, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms pathology, Carcinoma, Transitional Cell, Carcinoma in Situ drug therapy, Carcinoma in Situ pathology
- Abstract
Purpose: Immune checkpoint blockade holds promise for treating bacillus Calmette-Guerin (BCG)-unresponsive non-muscle-invasive bladder cancer (NMIBC). In this phase II study, we investigated the safety and efficacy of durvalumab, a human IgG1 monoclonal antibody, against BCG-unresponsive carcinoma in situ (CIS)., Patients and Methods: Patients with BCG-unresponsive CIS-containing NMIBC received durvalumab IV at 1,500 mg every 4 weeks for up to 12 months. The primary endpoint was complete response (CR) rate at month 6, defined by negative cystoscopy, urine cytology, and absence of high-grade recurrence on bladder mapping biopsy. The null hypothesis specified a CR rate of 18% and alternative hypothesis of 40%. According to the Simon two-stage design, if ≤3/13 patients achieved CR during stage 1, the trial is stopped due to futility., Results: Between March 8, 2017, and January 24, 2020, 17 patients were accrued whereas 4 withdrew from study treatment after bladder biopsy at month 3 was positive for CIS. Two of 17 (12%) achieved a CR at month 6, with duration of response of 10 and 18 months, respectively. A single grade 3 lipase elevation was attributed to durvalumab, and immune-related adverse events were observed in 7/17 (41%) patients. Only 1/17 patients had high programmed death-ligand 1 expression pretreatment. On RNA sequencing, complement activation genes were elevated posttreatment, along with enrichment of tumor-associated macrophage signature., Conclusions: Durvalumab monotherapy conferred minimal efficacy in treating BCG-unresponsive CIS of the bladder, with 6-month CR of 12%. Complement activation is a potential mechanism behind treatment resistance., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
21. Sézary syndrome originates from heavily mutated hematopoietic progenitors.
- Author
-
Harro CM, Sprenger KB, Chaurio RA, Powers JJ, Innamarato P, Anadon CM, Zhang Y, Biswas S, Mandal G, Mine JA, Cortina C, Nagy MZ, Martin AL, Handley KF, Borjas GJ, Chen PL, Pinilla-Ibarz J, Sokol L, Yu X, and Conejo-Garcia JR
- Subjects
- Humans, Dipeptidyl Peptidase 4, Receptors, Antigen, T-Cell, Sezary Syndrome genetics, Sezary Syndrome pathology, Skin Neoplasms genetics, Skin Neoplasms pathology, Mycosis Fungoides genetics, Mycosis Fungoides pathology, Lymphoma, T-Cell, Cutaneous genetics
- Abstract
The pathogenesis of cutaneous T-cell lymphoma (CTCL) remains unclear. Using single-cell RNA or T-cell receptor (TCR) sequencing of 32 619 CD3+CD4+ and CD26+/CD7+ and 29 932 CD3+CD4+ and CD26-/CD7- lymphocytes from the peripheral blood of 7 patients with CTCL, coupled to single-cell ATAC-sequencing of 26,411 CD3+CD4+ and CD26+/CD7+ and 33 841 CD3+CD4+ and CD26-/CD7- lymphocytes, we show that tumor cells in Sézary syndrome and mycosis fungoides (MF) exhibit different phenotypes and trajectories of differentiation. When compared to MF, Sézary cells exhibit narrower repertoires of TCRs and exhibit clonal enrichment. Surprisingly, we identified ≥200 mutations in hematopoietic stem cells from multiple patients with Sézary syndrome. Mutations in key oncogenes were also present in peripheral Sézary cells, which also showed the hallmarks of recent thymic egression. Together our data suggest that CTCL arises from mutated lymphocyte progenitors that acquire TCRs in the thymus, which complete their malignant transformation in the periphery., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
22. Virtual alignment of pathology image series for multi-gigapixel whole slide images.
- Author
-
Gatenbee CD, Baker AM, Prabhakaran S, Swinyard O, Slebos RJC, Mandal G, Mulholland E, Andor N, Marusyk A, Leedham S, Conejo-Garcia JR, Chung CH, Robertson-Tessi M, Graham TA, and Anderson ARA
- Subjects
- Technology, Microscopy methods, Software
- Abstract
Interest in spatial omics is on the rise, but generation of highly multiplexed images remains challenging, due to cost, expertise, methodical constraints, and access to technology. An alternative approach is to register collections of whole slide images (WSI), generating spatially aligned datasets. WSI registration is a two-part problem, the first being the alignment itself and the second the application of transformations to huge multi-gigapixel images. To address both challenges, we developed Virtual Alignment of pathoLogy Image Series (VALIS), software which enables generation of highly multiplexed images by aligning any number of brightfield and/or immunofluorescent WSI, the results of which can be saved in the ome.tiff format. Benchmarking using publicly available datasets indicates VALIS provides state-of-the-art accuracy in WSI registration and 3D reconstruction. Leveraging existing open-source software tools, VALIS is written in Python, providing a free, fast, scalable, robust, and easy-to-use pipeline for registering multi-gigapixel WSI, facilitating downstream spatial analyses., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
23. Dose-Limiting Pulmonary Toxicity in a Phase 1/2 Study of Radiation and Chemotherapy with Ipilimumab Followed by Nivolumab for Patients With Stage 3 Unresectable Non-Small Cell Lung Cancer.
- Author
-
Liveringhouse CL, Latifi K, Asous AG, Lam NB, Rosenberg SA, Dilling TJ, MacMillan GV, Chiappori AA, Haura EB, Creelan B, Gray JE, Tanvetyanon T, Shafique MR, Saltos AN, Weiner AA, Clarke J, Kelsey CR, Kim S, Caudell JJ, Rose TA, Conejo-Garcia JR, Li J, Schell MJ, Antonia SJ, and Perez BA
- Subjects
- Humans, Nivolumab adverse effects, Ipilimumab therapeutic use, Prospective Studies, Antineoplastic Combined Chemotherapy Protocols adverse effects, Neoplasm Staging, Carcinoma, Non-Small-Cell Lung drug therapy, Melanoma pathology, Lung Neoplasms drug therapy
- Abstract
Purpose: We hypothesized that concurrent ipilimumab with chemoradiationtherapy (chemoRT) followed by maintenance nivolumab would be safe for patients with unresectable stage III non-small cell lung cancer (NSCLC). We aimed to assess the safety (phase 1) and the 12-month progression-free survival (PFS) (phase 2) in a multi-institution prospective trial., Methods and Materials: Eligible patients had unresectable stage III NSCLC. The treatment included platinum doublet chemotherapy with concurrent thoracic radiation therapy to 60 Gy in 30 fractions and ipilimumab (1 mg/kg) delivered during weeks 1 and 4. After chemoRT, maintenance nivolumab (480 mg) was given every 4 weeks for up to 12 cycles. Adverse events (AEs) were assessed according to the Common Terminology Criteria for Adverse Events, version 5.0. Survival analyses were performed with Kaplan Meier (KM) methods and log-rank tests., Results: The trial was discontinued early after enrolling 19 patients without proceeding to the phase 2 component because of unacceptable toxicity. Sixteen patients (84%) had grade ≥3 (G3+) possible treatment-related toxicity, most commonly pulmonary AEs (n = 8, 42%). Fourteen patients (74%) discontinued study therapy early because of AEs (n = 12, 63%) or patient choice (n = 2, 11%). Eleven patients (58%) experienced G2+ pulmonary toxicity with median time to onset 4.1 months (95% CI 2.6-not reached [NR]), and 12-month freedom from G2+ pulmonary toxicity 37% (95% CI, 16-59). Five patients had G5 AEs, including 3 with G5 pulmonary AEs (1 respiratory failure with pneumonitis and pulmonary embolism, 1 pneumonia/chronic obstructive pulmonary disease exacerbation, 1 pulmonary fibrosis). Despite toxicities, the median PFS was 19.2 months (95% CI 6.1-NR) and the median overall survival was NR (95% CI 6.1-NR) with median follow-up of 30.1 months by the reverse KM method., Conclusions: Concurrent ipilimumab with chemoRT for unresectable stage III NSCLC is associated with pulmonary toxicity that may limit opportunities for improved outcomes. Future studies aiming to incorporate ipilimumab or other anti-CTLA4 therapies into management of unresectable stage III NSCLC should consider careful measures to minimize toxicity risk., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. A Novel Humanized Immune Stroma PDX Cancer Model for Therapeutic Studies.
- Author
-
Yang D, Beddows I, Tang H, Cascio S, McGonigal SC, Bai S, Johnson BK, Powers JJ, Acharya R, Bao R, Bruno TC, Soong TR, Conejo-Garcia JR, Shen H, Bility MT, and Buckanovich RJ
- Abstract
Standard preclinical human tumor models lack a human tumor stroma. However, as stroma contributes to therapeutic resistance, the lack of human stroma may make current models less stringent for testing new therapies. To address this, using patient-derived tumor cells, patient derived cancer-associated mesenchymal stem/progenitor cells, and human endothelial cells, we created a Human Stroma-Patient Derived Xenograft (HS-PDX) tumor model. HS-PDX, compared to the standard PDX model, demonstrate greater resistance to targeted therapy and chemotherapy, and better reflect patient response to therapy. Furthermore, HS-PDX can be grown in mice with humanized bone marrow to create humanized immune stroma patient-derived xenograft (HIS-PDX) models. The HIS-PDX model contains human connective tissues, vascular and immune cell infiltrates. RNA sequencing analysis demonstrated a 94-96% correlation with primary human tumor. Using this model, we demonstrate the impact of human tumor stroma on response to CAR-T cell therapy and immune checkpoint inhibitor therapy. We show an immunosuppressive role for human tumor stroma and that this model can be used to identify immunotherapeutic combinations to overcome stromally mediated immunosuppression. Combined, our data confirm a critical role for human stoma in therapeutic response and indicate that HIS-PDX can be an important tool for preclinical drug testing., Statement of Significance: We developed a tumor model with human stromal, vascular, and immune cells. This model mirrors patient response to chemotherapy, targeted therapy, and immunotherapy, and can be used to study therapy resistance.
- Published
- 2023
- Full Text
- View/download PDF
25. Actionable spontaneous antibody responses antagonize malignant progression in ovarian carcinoma.
- Author
-
Handley KF, Mehta S, Martin AL, Biswas S, Maharaj K, Nagy MZ, Mine JA, Cortina C, Yu X, Sprenger K, Mandal G, Innamarato P, Powers JJ, Harro CM, Chaurio RA, Anadon CM, Shahzad MM, Flores I, and Conejo-Garcia JR
- Subjects
- Humans, Female, Animals, Mice, Apoptosis, Antibody Formation, Cell Line, Tumor, Carcinoma, Ovarian Epithelial, Immunoglobulin A metabolism, Syntenins metabolism, Ovarian Neoplasms pathology, Endometriosis, Carcinoma, Endometrioid pathology, Adenocarcinoma, Clear Cell pathology
- Abstract
Objective: To demonstrate that shared antibody responses in endometriosis and endometriosis-associated ovarian cancer spontaneously antagonize malignant progression and can be leveraged to develop future immunotherapies., Methods: B cells from cyopreserved clear cell ovarian carcinoma (CCC, n = 2), endometrioid ovarian carcinoma (EC, n = 2), and endometriomas (n = 2) were isolated, activated, and EBV-immortalized. Antibodies were purified from B cell supernatants and used for screening arrays containing most of the human proteome. Targets were prioritized based on accessibility (transmembrane or secreted proteins), expression in endometriosis and cancer, and concurrent IgA and IgG responses. We focused on antibodies targeting tumor-promoting syndecan binding protein (SDCBP) to demonstrate anti-tumor activity. Immunoblots and qPCR were performed to assess SDCBP expression in ovarian cancer and endometriosis cell lines and tumor samples. Recombinant IgG4 was generated using the variable heavy and light chains of dominant B cell receptors (BCRs) reacting against the extracellular domain of SDCBP, and used in in vivo studies in human CCC- and high-grade serous ovarian carcinoma (HGSOC)-bearing immunodeficient mice., Results: Nine accessible proteins detected by both IgA and IgG were identified in all samples - including SDCBP, which is expressed in ovarian carcinomas of multiple histologies. Administration of α-SDCBP IgG4 in OVCAR3 (HGSOC), TOV21G and RMG-I (CCC) tumor-bearing mice significantly decreased tumor volume compared to control irrelevant IgG4., Conclusions: Spontaneous antibody responses exert suboptimal but measurable immune pressure against malignant progression in ovarian carcinomas. Using tumor-derived antibodies for developing novel immunotherapeutics warrants further investigation., Competing Interests: Declaration of Competing Interest KFH, SB, and JRCG report a provisional patent for “Utilization of immortalized B cells to identify SDCBP as a novel therapeutic target in ovarian carcinoma.” AM reports funding from Hearing the Ovarian Cancer Whisper, Ocala Royal Dames, Inc. JRCG has stock options in Compass Therapeutics, Anixa Biosciences and Alloy Therapeutics; has sponsored research with Anixa Biosciences; receives honorarium from Alloy Therapeutics; and has intellectual property with Compass Therapeutics and Anixa Biosciences; all outside the submitted work., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Measurement of Ovarian Tumor Immune Profiles by Multiplex Immunohistochemistry: Implications for Epidemiologic Studies.
- Author
-
Hathaway CA, Conejo-Garcia JR, Fridley BL, Rosner B, Saeed-Vafa D, Moran Segura C, Nguyen JV, Hecht JL, Sasamoto N, Terry KL, Tworoger SS, and Townsend MK
- Subjects
- Aged, Child, Female, Humans, Biomarkers, Biomarkers, Tumor metabolism, Epidemiologic Studies, Immunohistochemistry, Prospective Studies, Tumor Microenvironment, Ovarian Neoplasms epidemiology
- Abstract
Background: Despite the immunogenic nature of many ovarian tumors, treatment with immune checkpoint therapies has not led to substantial improvements in ovarian cancer survival. To advance population-level research on the ovarian tumor immune microenvironment, it is critical to understand methodologic issues related to measurement of immune cells on tissue microarrays (TMA) using multiplex immunofluorescence (mIF) assays., Methods: In two prospective cohorts, we collected formalin-fixed, paraffin-embedded ovarian tumors from 486 cases and created seven TMAs. We measured T cells, including several sub-populations, and immune checkpoint markers on the TMAs using two mIF panels. We used Spearman correlations, Fisher exact tests, and multivariable-adjusted beta-binomial models to evaluate factors related to immune cell measurements in TMA tumor cores., Results: Between-core correlations of intratumoral immune markers ranged from 0.52 to 0.72, with more common markers (e.g., CD3+, CD3+CD8+) having higher correlations. Correlations of immune cell markers between the whole core, tumor area, and stromal area were high (range 0.69-0.97). In multivariable-adjusted models, odds of T-cell positivity were lower in clear cell and mucinous versus type II tumors (ORs, 0.13-0.48) and, for several sub-populations, were lower in older tissue (sample age > 30 versus ≤ 10 years; OR, 0.11-0.32)., Conclusions: Overall, high correlations between cores for immune markers measured via mIF support the use of TMAs in studying ovarian tumor immune infiltration, although very old samples may have reduced antigenicity., Impact: Future epidemiologic studies should evaluate differences in the tumor immune response by histotype and identify modifiable factors that may alter the tumor immune microenvironment., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
27. Anti-4-1BB immunotherapy enhances systemic immune effects of radiotherapy to induce B and T cell-dependent anti-tumor immune activation and improve tumor control at unirradiated sites.
- Author
-
Martin AL, Powell C, Nagy MZ, Innamarato P, Powers J, Nichols D, Anadon CM, Chaurio RA, Kim S, Wang MH, Gong B, Wang X, Scheutz TJ, Antonia SJ, Conejo-Garcia JR, and Perez BA
- Subjects
- Humans, Immunotherapy, Tumor Necrosis Factor Receptor Superfamily, Member 9, Lymphocyte Activation, Tumor Microenvironment, CD8-Positive T-Lymphocytes, Neoplasms radiotherapy
- Abstract
Radiation therapy (RT) can prime and boost systemic anti-tumor effects via STING activation, resulting in enhanced tumor antigen presentation and antigen recognition by T cells. It is increasingly recognized that optimal anti-tumor immune responses benefit from coordinated cellular (T cell) and humoral (B cell) responses. However, the nature and functional relevance of the RT-induced immune response are controversial, beyond STING signaling, and agonistic interventions are lacking. Here, we show that B and CD4
+ T cell accumulation at tumor beds in response to RT precedes the arrival of CD8+ T cells, and both cell types are absolutely required for abrogated tumor growth in non-irradiated tumors. Further, RT induces increased expression of 4-1BB (CD137) in both T and B cells; both in preclinical models and in a cohort of patients with small cell lung cancer treated with thoracic RT. Accordingly, the combination of RT and anti-41BB therapy leads to increased immune cell infiltration in the tumor microenvironment and significant abscopal effects. Thus, 4-1BB therapy enhances radiation-induced tumor-specific immune responses via coordinated B and T cell responses, thereby preventing malignant progression at unirradiated tumor sites. These findings provide a rationale for combining RT and 4-1bb therapy in future clinical trials., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)- Published
- 2023
- Full Text
- View/download PDF
28. EGFR Inhibition by Cetuximab Modulates Hypoxia and IFN Response Genes in Head and Neck Squamous Cell Carcinoma.
- Author
-
Chaudhary R, Slebos RJC, Noel LC, Song F, Poole MI, Hoening DS, Hernandez-Prera JC, Conejo-Garcia JR, Guevara-Patino JA, Wang X, Xie M, Tan AC, and Chung CH
- Subjects
- Humans, Squamous Cell Carcinoma of Head and Neck drug therapy, Cetuximab pharmacology, ErbB Receptors genetics, CD8-Positive T-Lymphocytes metabolism, Hypoxia genetics, Tumor Microenvironment genetics, Carcinoma, Squamous Cell drug therapy, Head and Neck Neoplasms drug therapy
- Abstract
Head and neck squamous cell carcinoma (HNSCC) has one of the most hypoxic and immunosuppressive tumor microenvironments (TME) among solid tumors. However, there is no proven therapeutic strategy to remodel the TME to be less hypoxic and proinflammatory. In this study, we classified tumors according to a Hypoxia-Immune signature, characterized the immune cells in each subgroup, and analyzed the signaling pathways to identify a potential therapeutic target that can remodel the TME. We confirmed that hypoxic tumors had significantly higher numbers of immunosuppressive cells, as evidenced by a lower ratio of CD8
+ T cells to FOXP3+ regulatory T cells, compared with nonhypoxic tumors. Patients with hypoxic tumors had worse outcomes after treatment with pembrolizumab or nivolumab, anti-programmed cell death-1 inhibitors. Our expression analysis also indicated that hypoxic tumors predominantly increased the expression of the EGFR and TGFβ pathway genes. Cetuximab, an anti-EGFR inhibitor, decreased the expression of hypoxia signature genes, suggesting that it may alleviate the effects of hypoxia and remodel the TME to become more proinflammatory. Our study provides a rationale for treatment strategies combining EGFR-targeted agents and immunotherapy in the management of hypoxic HNSCC., Significance: While the hypoxic and immunosuppressive TME of HNSCC has been well described, comprehensive evaluation of the immune cell components and signaling pathways contributing to immunotherapy resistance has been poorly characterized. We further identified additional molecular determinants and potential therapeutic targets of the hypoxic TME to fully leverage currently available targeted therapies that can be administered with immunotherapy., (© 2023 The Authors; Published by the American Association for Cancer Research.)- Published
- 2023
- Full Text
- View/download PDF
29. Bcl11b sustains multipotency and restricts effector programs of intestinal-resident memory CD8 + T cells.
- Author
-
Helm EY, Zelenka T, Cismasiu VB, Islam S, Silvane L, Zitti B, Holmes TD, Drashansky TT, Kwiatkowski AJ, Tao C, Dean J, Obermayer AN, Chen X, Keselowsky BG, Zhang W, Huo Z, Zhou L, Sheridan BS, Conejo-Garcia JR, Shaw TI, Bryceson YT, and Avram D
- Subjects
- Mice, Animals, Cell Differentiation, Intestines, Tumor Suppressor Proteins metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, CD8-Positive T-Lymphocytes metabolism, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
The networks of transcription factors (TFs) that control intestinal-resident memory CD8
+ T (TRM ) cells, including multipotency and effector programs, are poorly understood. In this work, we investigated the role of the TF Bcl11b in TRM cells during infection with Listeria monocytogenes using mice with post-activation, conditional deletion of Bcl11b in CD8+ T cells. Conditional deletion of Bcl11b resulted in increased numbers of intestinal TRM cells and their precursors as well as decreased splenic effector and circulating memory cells and precursors. Loss of circulating memory cells was in part due to increased intestinal homing of Bcl11b-/- circulating precursors, with no major alterations in their programs. Bcl11b-/- TRM cells had altered transcriptional programs, with diminished expression of multipotent/multifunctional (MP/MF) program genes, including Tcf7 , and up-regulation of the effector program genes, including Prdm1. Bcl11b also limits the expression of Ahr, another TF with a role in intestinal CD8+ TRM cell differentiation. Deregulation of TRM programs translated into a poor recall response despite TRM cell accumulation in the intestine. Reduced expression of MP/MF program genes in Bcl11b-/- TRM cells was linked to decreased chromatin accessibility and a reduction in activating histone marks at these loci. In contrast, the effector program genes displayed increased activating epigenetic status. These findings demonstrate that Bcl11b is a frontrunner in the tissue residency program of intestinal memory cells upstream of Tcf1 and Blimp1, promoting multipotency and restricting the effector program.- Published
- 2023
- Full Text
- View/download PDF
30. Dichotomous Nitric Oxide-Dependent Post-Translational Modifications of STAT1 Are Associated with Ipilimumab Benefits in Melanoma.
- Author
-
Garg SK, Sun J, Kim Y, Whiting J, Sarnaik A, Conejo-Garcia JR, Phelps M, Weber JS, Mulé JJ, and Markowitz J
- Abstract
Although Ipilimumab (anti-CTLA-4) is FDA-approved for stage III/IV melanoma adjuvant treatment, it is not used clinically in first-line therapy, given the superior relapse-free survival (RFS)/toxicity benefits of anti-PD-1 therapy. However, it is important to understand anti-CTLA-4's mechanistic contribution to combination anti-PD-1/CTLA-4 therapy and investigate anti-CTLA-4 therapy for BRAF-wild type melanoma cases reresected after previous adjuvant anti-PD-1 therapy. Our group published that nitric oxide (NO) increased within the immune effector cells among patients with longer RFS after adjuvant ipilimumab, whereas NO increased within the immune suppressor cells among patients with shorter RFS. Herein, we measured the post-translational modifications of STAT1 (nitration-nSTAT1 and phosphorylation-pSTAT1) that are important for regulating its activity via flow cytometry and mass spectrometry approaches. PBMCs were analyzed from 35 patients undergoing adjuvant ipilimumab treatment. Shorter RFS was associated with higher pSTAT1 levels before ( p = 0.007) and after ( p = 0.036) ipilimumab. Ipilimumab-treated patients with high nSTAT1 levels before and after therapy in PBMCs experienced decreased RFS, but the change in nSTAT1 levels before and after ipilimumab therapy was associated with longer RFS ( p = 0.01). The measurement of post-translational modifications in STAT1 may distinguish patients with prolonged RFS from ipilimumab and provide mechanistic insight into responses to ipilimumab combination regimens.
- Published
- 2023
- Full Text
- View/download PDF
31. Pharmacological tumor PDL1 depletion with chlorambucil treats ovarian cancer and melanoma: improves antitumor immunity and renders anti-PDL1-resistant tumors anti-PDL1-sensitive through NK cell effects.
- Author
-
Bai H, Padron AS, Deng Y, Liao YJ, Murray CJ, Ontiveros C, Kari SJ, Kancharla A, Kornepati AVR, Garcia M, Reyes RM, Gupta HB, Conejo-Garcia JR, and Curiel T
- Subjects
- Animals, Female, Humans, Mice, Chlorambucil pharmacology, Chlorambucil therapeutic use, Killer Cells, Natural, United States, B7-H1 Antigen immunology, Melanoma, Experimental, Ovarian Neoplasms drug therapy
- Abstract
Background: Tumor intracellular programmed cell death ligand-1 (PDL1) mediates pathologic signals that regulate clinical treatment responses distinctly from surface-expressed PDL1 targeted by αPDL1 immune checkpoint blockade antibodies., Methods: We performed a drug screen for tumor cell PDL1 depleting drugs that identified Food and Drug Administration (FDA)-approved chlorambucil and also 9-[2-(phosphonomethoxy)ethyl] guanine. We used in vitro and in vivo assays to evaluate treatment and signaling effects of pharmacological tumor PDL1 depletion focused on chlorambucil as FDA approved, alone or plus αPDL1., Results: PDL1-expressing mouse and human ovarian cancer lines and mouse melanoma were more sensitive to chlorambucil-mediated proliferation inhibition in vitro versus corresponding genetically PDL1-depleted lines. Orthotopic peritoneal PDL1-expressing ID8agg ovarian cancer and subcutaneous B16 melanoma tumors were more chlorambucil-sensitive in vivo versus corresponding genetically PDL1-depleted tumors. Chlorambucil enhanced αPDL1 efficacy in tumors otherwise αPDL1-refractory, and improved antitumor immunity and treatment efficacy in a natural killer cell-dependent manner alone and plus αPDL1. Chlorambucil-mediated PDL1 depletion was relatively tumor-cell selective in vivo, and treatment efficacy was preserved in PDL1KO hosts, demonstrating tumor PDL1-specific treatment effects. Chlorambucil induced PDL1-dependent immunogenic tumor cell death which could help explain immune contributions. Chlorambucil-mediated PDL1 reduction mechanisms were tumor cell-type-specific and involved transcriptional or post-translational mechanisms, including promoting PDL1 ubiquitination through the GSK3β/β-TRCP pathway. Chlorambucil-mediated tumor cell PDL1 depletion also phenocopied genetic PDL1 depletion in reducing tumor cell mTORC1 activation and tumor initiating cell content, and in augmenting autophagy, suggesting additional treatment potential., Conclusions: Pharmacological tumor PDL1 depletion with chlorambucil targets tumor-intrinsic PDL1 signaling that mediates treatment resistance, especially in αPDL1-resistant tumors, generates PDL1-dependent tumor immunogenicity and inhibits tumor growth in immune-dependent and independent manners. It could improve treatment efficacy of selected agents in otherwise treatment-refractory, including αPDL1-refractory cancers, and is rapidly clinically translatable., Competing Interests: Competing interests: TC and ARK have filed a patent on PDL1 depletion drugs., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
32. Lifetime Exposure to Cigarette Smoke and Risk of Ovarian Cancer by T-cell Tumor Immune Infiltration.
- Author
-
Hathaway CA, Wang T, Townsend MK, Vinci C, Jake-Schoffman DE, Saeed-Vafa D, Moran Segura C, Nguyen JV, Conejo-Garcia JR, Fridley BL, and Tworoger SS
- Subjects
- Adult, Humans, Female, Risk, T-Lymphocytes, Tumor Microenvironment, Cigarette Smoking, Ovarian Neoplasms epidemiology, Ovarian Neoplasms etiology
- Abstract
Background: Exposure to cigarette smoke, particularly in early life, is modestly associated with ovarian cancer risk and may impact systemic immunity and the tumor immune response. However, no studies have evaluated whether cigarette smoke exposure impacts the ovarian tumor immune microenvironment., Methods: Participants in the Nurses' Health Study (NHS) and NHSII reported on early life exposure to cigarette smoke and personal smoking history on questionnaires (n = 165,760). Multiplex immunofluorescence assays were used to measure markers of T cells and immune checkpoints in tumor tissue from 385 incident ovarian cancer cases. We used Cox proportional hazards models to evaluate HRs and 95% confidence intervals (CI) for developing ovarian tumors with a low (
- Published
- 2023
- Full Text
- View/download PDF
33. Neglected no more: B cell-mediated anti-tumor immunity.
- Author
-
Conejo-Garcia JR, Biswas S, Chaurio R, and Rodriguez PC
- Subjects
- Animals, Humans, Mice, Adaptive Immunity immunology, B-Lymphocytes immunology, Neoplasms immunology, Neoplasms therapy, T-Lymphocytes immunology, Tertiary Lymphoid Structures immunology
- Abstract
Immuno-oncology has traditionally focused on the cellular arm of the adaptive immune response, while attributing tumor-promoting activity to humoral responses in tumor-bearing hosts. This view stems from mouse models that do not necessarily recapitulate the antibody response process consistently observed in most human cancers. In recent years, the field has reconsidered the coordinated action of T and B cell responses in the context of anti-tumor immunity, as in any other immune response. Thus, recent studies in human cancer identify B cell responses with better outcome, typically in association with superior T cell responses. An area of particular interest is tertiary lymphoid structures, where germinal centers produce isotype switched antibodies and B cells and T lymphocytes interact with other immune cell types. The presence of these lymphoid structures is associated with better immunotherapeutic responses and remain poorly understood. Here, we discuss recent discoveries on how coordination between humoral and cellular responses is required for effective immune pressure against malignant progression, providing a perspective on the role of tertiary lymphoid structures and interventions to elicit their formation in unresectable tumors., (Copyright © 2022. Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
34. Protection of Regulatory T Cells from Fragility and Inactivation in the Tumor Microenvironment.
- Author
-
Zhang H, Tomar VS, Li J, Basavaraja R, Yan F, Gui J, McBrearty N, Costich TL, Beiting DP, Blanco MA, Conejo-Garcia JR, Saggu G, Berger A, Nefedova Y, Gabrilovich DI, and Fuchs SY
- Subjects
- Humans, Tumor Microenvironment, Neuropilin-1, Immunotherapy, T-Lymphocytes, Regulatory, Neoplasms
- Abstract
Fragility of regulatory T (Treg) cells manifested by the loss of neuropilin-1 (NRP1) and expression of IFNγ undermines the immune suppressive functions of Treg cells and contributes to the success of immune therapies against cancers. Intratumoral Treg cells somehow avoid fragility; however, the mechanisms by which Treg cells are protected from fragility in the tumor microenvironment are not well understood. Here, we demonstrate that the IFNAR1 chain of the type I IFN (IFN1) receptor was downregulated on intratumoral Treg cells. Downregulation of IFNAR1 mediated by p38α kinase protected Treg cells from fragility and maintained NRP1 levels, which were decreased in response to IFN1. Genetic or pharmacologic inactivation of p38α and stabilization of IFNAR1 in Treg cells induced fragility and inhibited their immune suppressive and protumorigenic activities. The inhibitor of sumoylation TAK981 (Subasumstat) upregulated IFNAR1, eliciting Treg fragility and inhibiting tumor growth in an IFNAR1-dependent manner. These findings describe a mechanism by which intratumoral Treg cells retain immunosuppressive activities and suggest therapeutic approaches for inducing Treg fragility and increasing the efficacy of immunotherapies., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
35. Case Report: Durable complete pathologic response and organ preservation following ipilimumab and nivolumab for locally advanced primary vaginal mucosal melanoma.
- Author
-
Tarhini AA, Hanayneh WB, Powers JJ, Segura CMM, Conejo-Garcia JR, Lam CA, Hakam A, and Hoffman MS
- Abstract
Optimal management of locally advanced vaginal mucosal melanoma is poorly understood because of its rarity and unique biology. Patients have a poor prognosis despite aggressive management approaches including pelvic exenteration and adjuvant radiation that carry major morbidities. We report a case of a patient in early 40's who experienced complete pathologic response and organ preservation following immunotherapy consisting of 3 cycles of ipilimumab and nivolumab. Treatment was complicated by a high-grade immune mediated hepatitis that eventually resolved with immunosuppressive therapy. Immune monitoring studies utilizing vaginal tumor biopsies showed evidence of enhanced infiltration by CD3+/CD8+ cytotoxic T-cells and increased expression of MHC-I/PD-L1 within the tumor microenvironment following immunotherapy. The patient continues to be without evidence of disease recurrence by radiologic and gynecologic examinations with more than 2 years of follow up from the time of immunotherapy initiation. To our knowledge, this is the only case report in the literature of a patient with locally advanced vaginal mucosal melanoma experiencing a durable complete pathologic response and organ preservation following immune checkpoint blockade as the only treatment approach., Competing Interests: AT reports grants contracted with the institution from Bristol Myers Squib, Genentech-Roche, Regeneron, Sanofi-Genzyme, Nektar, Clinigen, Merck, Acrotech, Pfizer, Checkmate, OncoSec. Consulting fees from Bristol Myers Squibb, Merck, Easai, Instil Bio, Clinigen, Regeneron, Sanofi-Genzyme, Novartis, Partner Therapeutics, Genentech/Roche, BioNTech. All outside the submitted work. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Tarhini, Hanayneh, Powers, Segura, Conejo-Garcia, Lam, Hakam and Hoffman.)
- Published
- 2022
- Full Text
- View/download PDF
36. Barriers and Opportunities for CAR T-Cell Targeting of Solid Tumors.
- Author
-
Conejo-Garcia JR and Guevara-Patino JA
- Subjects
- Humans, Immunotherapy, Adoptive, T-Lymphocytes metabolism, Tumor Microenvironment, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen metabolism, Neoplasms therapy, Neoplasms metabolism, Hematologic Neoplasms therapy, Hematologic Neoplasms metabolism
- Abstract
CAR T-cell therapy has transformed the treatment of hematological malignancies of the B cell lineage. However, the quest to fulfil the same promise for solid tumors is still in its infancy. This review summarizes some of the challenges that the field is trying to overcome for effective treatment of human carcinomas, including tumor heterogeneity, the paucity of truly tumor-specific targets, immunosuppression and metabolic restrictions at solid tumor beds, and defective T-cell trafficking. All these barriers are being currently investigated and, in some cases, targeted, by multiple independent groups. With clinical interventions against multiple human malignancies and different platforms under accelerated clinical development, the next few years will see an array of cellular therapies, including CAR T-cells, progressively becoming routine interventions to eliminate currently incurable diseases, as it happened with some hematological malignancies.
- Published
- 2022
- Full Text
- View/download PDF
37. Ablation of the endoplasmic reticulum stress kinase PERK induces paraptosis and type I interferon to promote anti-tumor T cell responses.
- Author
-
Mandula JK, Chang S, Mohamed E, Jimenez R, Sierra-Mondragon RA, Chang DC, Obermayer AN, Moran-Segura CM, Das S, Vazquez-Martinez JA, Prieto K, Chen A, Smalley KSM, Czerniecki B, Forsyth P, Koya RC, Ruffell B, Cubillos-Ruiz JR, Munn DH, Shaw TI, Conejo-Garcia JR, and Rodriguez PC
- Subjects
- Animals, Endoplasmic Reticulum Stress, Mice, Signal Transduction, T-Lymphocytes metabolism, Unfolded Protein Response, eIF-2 Kinase genetics, eIF-2 Kinase metabolism, Interferon Type I metabolism, Neoplasms
- Abstract
Activation of unfolded protein responses (UPRs) in cancer cells undergoing endoplasmic reticulum (ER) stress promotes survival. However, how UPR in tumor cells impacts anti-tumor immune responses remains poorly described. Here, we investigate the role of the UPR mediator pancreatic ER kinase (PKR)-like ER kinase (PERK) in cancer cells in the modulation of anti-tumor immunity. Deletion of PERK in cancer cells or pharmacological inhibition of PERK in melanoma-bearing mice incites robust activation of anti-tumor T cell immunity and attenuates tumor growth. PERK elimination in ER-stressed malignant cells triggers SEC61β-induced paraptosis, thereby promoting immunogenic cell death (ICD) and systemic anti-tumor responses. ICD induction in PERK-ablated tumors stimulates type I interferon production in dendritic cells (DCs), which primes CCR2-dependent tumor trafficking of common-monocytic precursors and their intra-tumor commitment into monocytic-lineage inflammatory Ly6C
+ CD103+ DCs. These findings identify how tumor cell-derived PERK promotes immune evasion and highlight the potential of PERK-targeting therapies in cancer immunotherapy., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
38. Reduced Satb1 expression predisposes CD4 + T conventional cells to Treg suppression and promotes transplant survival.
- Author
-
Gupta PK, Allocco JB, Fraipont JM, McKeague ML, Wang P, Andrade MS, McIntosh C, Chen L, Wang Y, Li Y, Andrade J, Conejo-Garcia JR, Chong AS, and Alegre ML
- Subjects
- Animals, Immunologic Memory genetics, Interleukin-2 metabolism, Mice, Mice, Inbred C57BL, Graft Survival genetics, Graft Survival immunology, Matrix Attachment Region Binding Proteins genetics, Matrix Attachment Region Binding Proteins metabolism, T-Lymphocytes, Regulatory immunology, Transcription Factors genetics, Transcription Factors metabolism, Transplantation Tolerance genetics, Transplantation Tolerance immunology
- Abstract
Limiting CD4
+ T cell responses is important to prevent solid organ transplant rejection. In a mouse model of costimulation blockade-dependent cardiac allograft tolerance, we previously reported that alloreactive CD4+ conventional T cells (Tconvs) develop dysfunction, losing proliferative capacity. In parallel, induction of transplantation tolerance is dependent on the presence of regulatory T cells (Tregs). Whether susceptibility of CD4+ Tconvs to Treg suppression is modulated during tolerance induction is unknown. We found that alloreactive Tconvs from transplant tolerant mice had augmented sensitivity to Treg suppression when compared with memory T cells from rejector mice and expressed a transcriptional profile distinct from these memory T cells, including down-regulated expression of the transcription factor Special AT-rich sequence-binding protein 1 (Satb1). Mechanistically, Satb1 deficiency in CD4+ T cells limited their expression of CD25 and IL-2, and addition of Tregs, which express higher levels of CD25 than Satb1-deficient Tconvs and successfully competed for IL-2, resulted in greater suppression of Satb1-deficient than wild-type Tconvs in vitro. In vivo, Satb1-deficient Tconvs were more susceptible to Treg suppression, resulting in significantly prolonged skin allograft survival. Overall, our study reveals that transplantation tolerance is associated with Tconvs' susceptibility to Treg suppression, via modulated expression of Tconv-intrinsic Satb1. Targeting Satb1 in the context of Treg-sparing immunosuppressive therapies might be exploited to improve transplant outcomes.- Published
- 2022
- Full Text
- View/download PDF
39. Protocol for the isolation of CD8+ tumor-infiltrating lymphocytes from human tumors and their characterization by single-cell immune profiling and multiome.
- Author
-
Anadon CM, Zhang C, Wang X, Cen L, Conejo-Garcia JR, and Yu X
- Subjects
- CD8-Positive T-Lymphocytes, Flow Cytometry, Humans, Lymphocytes, Tumor-Infiltrating, Neoplasms pathology
- Abstract
Understanding the heterogenicity of tumor-infiltraing lymphocyte (TIL) populations and the immunobiology in human cancer is a key to establish efficient immunotherapies. Here, we have established a protocol for the characterization of CD8
+ TILs in tumors by single-cell RNA-seq paired to VDJ profiling and chromatin structure including dissociation of tumor biopsies. We have also provided guidance for subsequent fluorescence-activated cell sorting (FACS), single-cell encapsulation, bioinformatics analysis, and troubleshooting. For complete details on the use and execution of this protocol, please refer to Anadon et al. (2022)., Competing Interests: J.R.C.-G. has stock options in Compass Therapeutics, Anixa Biosciences, and Alloy Therapeutics; has sponsored research with Anixa Biosciences; receives honorarium from Alloy Therapeutics and Leidos; and has intellectual property with Compass Therapeutics and Anixa Biosciences., (© 2022 The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
40. Belly Fat Weakens Immune Fitness.
- Author
-
Conejo-Garcia JR and Curiel TJ
- Subjects
- Abdominal Fat metabolism, Carcinoma, Ovarian Epithelial, Female, Humans, Lysophospholipids, Interferon Type I, Ovarian Neoplasms metabolism
- Abstract
Much work has been done to reduce cancer immunosuppression through inhibiting soluble proteins, surface molecules, and suppressive cells. This article shows an important role for the lipid lysophosphatidic acid, whose suppression shows promise as a novel cancer immunotherapeutic, demonstrated in ovarian cancer. See related article by Chae et al., 1904 (5)., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
41. Olfactory Receptor OR2H1 Is an Effective Target for CAR T Cells in Human Epithelial Tumors.
- Author
-
Martin AL, Anadon CM, Biswas S, Mine JA, Handley KF, Payne KK, Mandal G, Chaurio RA, Powers JJ, Sprenger KB, Rigolizzo KE, Innamarato P, Harro CM, Mehta S, Perez BA, Wenham RM, and Conejo-Garcia JR
- Subjects
- Female, Humans, Cell Line, Tumor, Immunotherapy, Adoptive, T-Lymphocytes, Lung Neoplasms genetics, Lung Neoplasms therapy, Neoplasms, Glandular and Epithelial metabolism, Ovarian Neoplasms metabolism, Receptors, Odorant metabolism
- Abstract
Although chimeric antigen receptor (CAR)-expressing T cells have proven success in hematologic malignancies, their effectiveness in solid tumors has been largely unsuccessful thus far. We found that some olfactory receptors are expressed in a variety of solid tumors of different histologic subtypes, with a limited pattern of expression in normal tissues. Quantification of OR2H1 expression by qRT-PCR and Western blot analysis of 17 normal tissues, 82 ovarian cancers of various histologies, eight non-small cell lung cancers (NSCLCs), and 17 breast cancers demonstrated widespread OR2H1 expression in solid epithelial tumors with expression in normal human tissues limited to the testis. CAR T cells recognizing the extracellular domain of the olfactory receptor OR2H1 were generated with a targeting motif identified through the screening of a phage display library and demonstrated OR2H1-specific cytotoxic killing in vitro and in vivo, using tumor cells with spontaneous expression of variable OR2H1 levels. Importantly, recombinant OR2H1 IgG generated with the VH/VL sequences of the CAR construct specifically detected OR2H1 protein signal in 60 human lung cancers, 40 ovarian carcinomas, and 73 cholangiocarcinomas, at positivity rates comparable with mRNA expression and without OR2H1 staining in 58 normal tissues. CRISPR/Cas9-mediated ablation of OR2H1 confirmed targeting specificity of the CAR and the tumor-promoting role of OR2H1 in glucose metabolism. Therefore, T cells redirected against OR2H1-expressing tumor cells represent a promising therapy against a broad range of epithelial cancers, likely with an admissible toxicity profile., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
42. Tumor Intrinsic PD-L1 Promotes DNA Repair in Distinct Cancers and Suppresses PARP Inhibitor-Induced Synthetic Lethality.
- Author
-
Kornepati AVR, Boyd JT, Murray CE, Saifetiarova J, de la Peña Avalos B, Rogers CM, Bai H, Padron AS, Liao Y, Ontiveros C, Svatek RS, Hromas R, Li R, Hu Y, Conejo-Garcia JR, Vadlamudi RK, Zhao W, Dray E, Sung P, and Curiel TJ
- Subjects
- Animals, B7-H1 Antigen genetics, BRCA1 Protein genetics, Cell Line, Tumor, DNA Repair, Humans, Mice, Phthalazines pharmacology, Phthalazines therapeutic use, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Synthetic Lethal Mutations, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Neoplasms genetics
- Abstract
BRCA1-mediated homologous recombination is an important DNA repair mechanism that is the target of FDA-approved PARP inhibitors, yet details of BRCA1-mediated functions remain to be fully elucidated. Similarly, immune checkpoint molecules are targets of FDA-approved cancer immunotherapies, but the biological and mechanistic consequences of their application are incompletely understood. We show here that the immune checkpoint molecule PD-L1 regulates homologous recombination in cancer cells by promoting BRCA1 nuclear foci formation and DNA end resection. Genetic depletion of tumor PD-L1 reduced homologous recombination, increased nonhomologous end joining, and elicited synthetic lethality to PARP inhibitors olaparib and talazoparib in vitro in some, but not all, BRCA1 wild-type tumor cells. In vivo, genetic depletion of tumor PD-L1 rendered olaparib-resistant tumors sensitive to olaparib. In contrast, anti-PD-L1 immune checkpoint blockade neither enhanced olaparib synthetic lethality nor improved its efficacy in vitro or in wild-type mice. Tumor PD-L1 did not alter expression of BRCA1 or its cofactor BARD1 but instead coimmunoprecipitated with BARD1 and increased BRCA1 nuclear accumulation. Tumor PD-L1 depletion enhanced tumor CCL5 expression and TANK-binding kinase 1 activation in vitro, similar to known immune-potentiating effects of PARP inhibitors. Collectively, these data define immune-dependent and immune-independent effects of PARP inhibitor treatment and genetic tumor PD-L1 depletion. Moreover, they implicate a tumor cell-intrinsic, immune checkpoint-independent function of PD-L1 in cancer cell BRCA1-mediated DNA damage repair with translational potential, including as a treatment response biomarker., Significance: PD-L1 upregulates BRCA1-mediated homologous recombination, and PD-L1-deficient tumors exhibit BRCAness by manifesting synthetic lethality in response to PARP inhibitors, revealing an exploitable therapeutic vulnerability and a candidate treatment response biomarker. See related commentary by Hanks, p. 2069., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
43. γδ T cells share the spotlight in cancer.
- Author
-
Conejo-Garcia JR and Innamarato P
- Subjects
- Humans, Receptors, Antigen, T-Cell, gamma-delta, T-Lymphocytes, Neoplasms therapy
- Published
- 2022
- Full Text
- View/download PDF
44. T cell repertoire in peripheral blood as a potential biomarker for predicting response to concurrent cetuximab and nivolumab in head and neck squamous cell carcinoma.
- Author
-
Wang X, Muzaffar J, Kirtane K, Song F, Johnson M, Schell MJ, Li J, Yoder SJ, Conejo-Garcia JR, Guevara-Patino JA, Bonomi M, Bhateja P, Rocco JW, Steuer CE, Saba NF, and Chung CH
- Subjects
- Biomarkers, Cetuximab, Humans, Leukocytes, Mononuclear, Nivolumab pharmacology, Nivolumab therapeutic use, Receptors, Antigen, T-Cell, Squamous Cell Carcinoma of Head and Neck drug therapy, T-Lymphocytes, Head and Neck Neoplasms drug therapy, Papillomavirus Infections drug therapy
- Abstract
Background: T cell receptor (TCR) signaling profile is a fundamental property that underpins both adaptive and innate immunity in the host. Despite its potential clinical relevance, the TCR repertoire in peripheral blood has not been thoroughly explored for its value as an immunotherapy efficacy biomarker in head and neck squamous cell carcinoma (HNSCC). The purpose of the present study is to characterize and compare the TCR repertoire in peripheral blood mononuclear cells (PBMC) from patients with HNSCC treated with the combination of cetuximab and nivolumab., Methods: We used the immunoSEQ assay to sequence the TCR beta (TCR-B) chain repertoire from serially obtained PBMC at baseline and during the treatments from a total of 41 patients who received the combination (NCT03370276). Key TCR repertoire metrics, including diversity and clonality, were calculated and compared between patients with different therapy responses and clinical characteristics (eg, human papillomavirus (HPV) status and smoking history). Patient survival outcomes were compared according to patient groups stratified by the TCR-B clonotyping. To confirm the observed patterns in TCR spectrum, samples from patients who achieved complete response (CR) and partial response (PR) were further profiled with the immunoSEQ deep resolution assay., Results: Our data indicated that the patients who achieved CR and PR had an increased TCR sequence diversity in their baseline samples, this tendency being more pronounced in HPV-negative patients or those with a smoking history. Notably, the CR/PR group had the lowest proportion of patients with oligoclonal TCR clones (2 out of 8 patients), followed by the stable disease group (9 out of 20 patients) and lastly the progressive disease group (7 out of 10 patients). An overall trend toward favorable patient survival was also observed in the polyclonal group. Finally, we reported the shared TCR clones across patients within the same response group, as well as the shared clones by aligning immunoSEQ reads with TCR data retrieved from The Cancer Genome Atlas- head and neck squamous cell carcinoma (TCGA-HNSC) cohort., Conclusions: Our data suggest that, despite the great clinical heterogeneity of HNSCC and the limited responders in the present cohort, the peripheral TCR repertoires from pretreatment PBMC may be developed as biomarkers for the benefit of immunotherapy in HNSCC., Competing Interests: Competing interests: CHC—honoraria from Bristol Myers Squibb, CUE, Sanofi, Mirati, Merck, and Exelixis for ad hoc Scientific Advisory Board participation. CS—honoraria from Armo, Bergen Bio, AbbVie, and Lilly Oncology for ad hoc Scientific Advisory Board participation. NS—honoraria from Pfizer, Merck, Aduro, Rakuten, CUE, and Blupoint, Eisai, AstraZeneca, WebMD, Mirati, Reach MD, Vaccinex, Kura, Biontech, GSK, Aduro, Pfizer for ad hoc Scientific Advisory Board or Data Safety Monitoring Committee and research funding Bristol Myers Squibb and Exelixis. JRCG—Honorarium from Alloy Therapeutics and Anixa Biosciences; stock options in Alloy Therapeutics, Anixa Biosciences and Compass Therapeutics; intellectual property with Anixa Biosciences and Compass Therapeutics; sponsored research supported by Anixa Biosciences.The other authors do not have conflict of interest to declare., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
45. Ovarian cancer immunogenicity is governed by a narrow subset of progenitor tissue-resident memory T cells.
- Author
-
Anadon CM, Yu X, Hänggi K, Biswas S, Chaurio RA, Martin A, Payne KK, Mandal G, Innamarato P, Harro CM, Mine JA, Sprenger KB, Cortina C, Powers JJ, Costich TL, Perez BA, Gatenbee CD, Prabhakaran S, Marchion D, Heemskerk MHM, Curiel TJ, Anderson AR, Wenham RM, Rodriguez PC, and Conejo-Garcia JR
- Subjects
- CD8-Positive T-Lymphocytes, Female, Humans, Lymphocytes, Tumor-Infiltrating, Memory T Cells, Immunologic Memory, Ovarian Neoplasms
- Abstract
Despite repeated associations between T cell infiltration and outcome, human ovarian cancer remains poorly responsive to immunotherapy. We report that the hallmarks of tumor recognition in ovarian cancer-infiltrating T cells are primarily restricted to tissue-resident memory (TRM) cells. Single-cell RNA/TCR/ATAC sequencing of 83,454 CD3
+ CD8+ CD103+ CD69+ TRM cells and immunohistochemistry of 122 high-grade serous ovarian cancers shows that only progenitor (TCF1low ) tissue-resident T cells (TRMstem cells), but not recirculating TCF1+ T cells, predict ovarian cancer outcome. TRMstem cells arise from transitional recirculating T cells, which depends on antigen affinity/persistence, resulting in oligoclonal, trogocytic, effector lymphocytes that eventually become exhausted. Therefore, ovarian cancer is indeed an immunogenic disease, but that depends on ∼13% of CD8+ tumor-infiltrating T cells (∼3% of CD8+ clonotypes), which are primed against high-affinity antigens and maintain waves of effector TRM-like cells. Our results define the signature of relevant tumor-reactive T cells in human ovarian cancer, which could be applicable to other tumors with unideal mutational burden., Competing Interests: Declaration of interests J.R.C.-G. has stock options with Compass Therapeutics, Anixa Biosciences, and Alloy Therapeutics; receives honorarium from Anixa Biosciences, Alloy Therapeutics, and Leidos; and has sponsored research with Anixa Biosciences (all outside the submitted work). B.A.P. has served on the Advisory Board of AstraZeneca and has research support from BMS. J.R.C.-G. is currently an employee of STEMCELL Technologies (all work outside the submitted work). R.M.W. reports grants and personal fees from Merck, personal fees from Tesaro/GSK, personal fees from Genentech, personal fees from Legend Biotech, personal fees from AbbVie, personal fees from Astrazeneca, grants and stock from Ovation Diagnostics, personal fees from Clovis Oncology, and personal fees from Regeneron (all outside the submitted work)., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
46. Racial Differences in the Tumor Immune Landscape and Survival of Women with High-Grade Serous Ovarian Carcinoma.
- Author
-
Peres LC, Colin-Leitzinger C, Sinha S, Marks JR, Conejo-Garcia JR, Alberg AJ, Bandera EV, Berchuck A, Bondy ML, Christensen BC, Cote ML, Doherty JA, Moorman PG, Peters ES, Moran Segura C, Nguyen JV, Schwartz AG, Terry PD, Wilson CM, Fridley BL, and Schildkraut JM
- Subjects
- Ethnicity, Female, Humans, Lymphocytes, Tumor-Infiltrating, Race Factors, Ovarian Neoplasms
- Abstract
Background: Tumor-infiltrating lymphocytes (TIL) confer a survival benefit among patients with ovarian cancer; however, little work has been conducted in racially diverse cohorts., Methods: The current study investigated racial differences in the tumor immune landscape and survival of age- and stage-matched non-Hispanic Black and non-Hispanic White women with high-grade serous ovarian carcinoma (HGSOC) enrolled in two population-based studies (n = 121 in each racial group). We measured TILs (CD3+), cytotoxic T cells (CD3+CD8+), regulatory T cells (CD3+FoxP3+), myeloid cells (CD11b+), and neutrophils (CD11b+CD15+) via multiplex immunofluorescence. Multivariable Cox proportional hazard regression was used to estimate the association between immune cell abundance and survival overall and by race., Results: Overall, higher levels of TILs, cytotoxic T cells, myeloid cells, and neutrophils were associated with better survival in the intratumoral and peritumoral region, irrespective of tissue compartment (tumor, stroma). Improved survival was noted for T-regulatory cells in the peritumoral region and in the stroma of the intratumoral region, but no association for intratumoral T-regulatory cells. Despite similar abundance of immune cells across racial groups, associations with survival among non-Hispanic White women were consistent with the overall findings, but among non-Hispanic Black women, most associations were attenuated and not statistically significant., Conclusions: Our results add to the existing evidence that a robust immune infiltrate confers a survival advantage among women with HGSOC; however, non-Hispanic Black women may not experience the same survival benefit as non-Hispanic White women with HGSOC., Impact: This study contributes to our understanding of the immunoepidemiology of HGSOC in diverse populations., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
47. Tumor Expression Quantitative Trait Methylation Screening Reveals Distinct CpG Panels for Deconvolving Cancer Immune Signatures.
- Author
-
Yu X, Cen L, Chen YA, Markowitz J, Shaw TI, Tsai KY, Conejo-Garcia JR, and Wang X
- Subjects
- Biomarkers, Tumor genetics, CpG Islands genetics, Early Detection of Cancer, Epigenomics, Gene Expression Regulation, Neoplastic, Humans, Phenotype, Prognosis, Tumor Microenvironment genetics, DNA Methylation, Melanoma genetics, Melanoma pathology
- Abstract
DNA methylation signatures in tumors could serve as reliable biomarkers that are accessible in archival tissues for tracking the epigenetic dynamics shaped by both cancer cells and the tumor microenvironment. However, given the ultrahigh dimensionality and noncollapsible nature of the data, it remains challenging to screen all CpG sites to identify the most promising marker panels. In this article, we introduce the concept of tumor-based expression quantitative trait methylation (eQTM) for the prioritization and systematic mining of predictive biomarkers. In melanoma as a disease model, eQTM CpGs and genes represent new and efficient candidate targets to be investigated for both prognostic and immune status monitoring purposes. Three cis-eQTM CpGs (cg07786657, cg12446199, and cg00027570) were strongly associated with and can serve as surrogate biomarkers for the tumor immune cytolytic activity score (CYT). In addition, multiple eQTM genes could be further exploited for predicting immunoregulatory phenotypes. A targeted gene panel analysis identified one eQTM in TCF7 (cg25947408) as a novel candidate biomarker for uncoupling overall T-cell differentiation and exhaustion status in a tumor. The prognostic significance of this eQTM as an independent signature to CYT was validated by both The Cancer Genome Atlas and Moffitt melanoma cohort data. Overall, eQTMs represent a mechanistically distinct class of potential biomarkers that can be used to predict patient prognosis and immune status., Significance: This study provides a novel and promising approach to identify targeted epigenetic biomarkers in cancer and will spur further analysis in tumor immune phenotyping., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
48. Genomic and Single-Cell Landscape Reveals Novel Drivers and Therapeutic Vulnerabilities of Transformed Cutaneous T-cell Lymphoma.
- Author
-
Song X, Chang S, Seminario-Vidal L, de Mingo Pulido A, Tordesillas L, Song X, Reed RA, Harkins A, Whiddon S, Nguyen JV, Segura CM, Zhang C, Yoder S, Sayegh Z, Zhao Y, Messina JL, Harro CM, Zhang X, Conejo-Garcia JR, Berglund A, Sokol L, Zhang J, Rodriguez PC, Mulé JJ, Futreal AP, Tsai KY, and Chen PL
- Subjects
- Cell Transformation, Neoplastic, Ecosystem, Genomics, Humans, Lymphoma, T-Cell, Cutaneous drug therapy, Lymphoma, T-Cell, Cutaneous genetics, Skin Neoplasms drug therapy, Skin Neoplasms genetics, Skin Neoplasms metabolism
- Abstract
Abstract: Cutaneous T-cell lymphoma (CTCL) is a rare cancer of skin-homing T cells. A subgroup of patients develops large cell transformation with rapid progression to an aggressive lymphoma. Here, we investigated the transformed CTCL (tCTCL) tumor ecosystem using integrative multiomics spanning whole-exome sequencing (WES), single-cell RNA sequencing, and immune profiling in a unique cohort of 56 patients. WES of 70 skin biopsies showed high tumor mutation burden, UV signatures that are prognostic for survival, exome-based driver events, and most recurrently mutated pathways in tCTCL. Single-cell profiling of 16 tCTCL skin biopsies identified a core oncogenic program with metabolic reprogramming toward oxidative phosphorylation (OXPHOS), cellular plasticity, upregulation of MYC and E2F activities, and downregulation of MHC I suggestive of immune escape. Pharmacologic perturbation using OXPHOS and MYC inhibitors demonstrated potent antitumor activities, whereas immune profiling provided in situ evidence of intercellular communications between malignant T cells expressing macrophage migration inhibitory factor and macrophages and B cells expressing CD74., Significance: Our study contributes a key resource to the community with the largest collection of tCTCL biopsies that are difficult to obtain. The multiomics data herein provide the first comprehensive compendium of genomic alterations in tCTCL and identify potential prognostic signatures and novel therapeutic targets for an incurable T-cell lymphoma. This article is highlighted in the In This Issue feature, p. 1171., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
49. Interaction of bacterial genera associated with therapeutic response to immune checkpoint PD-1 blockade in a United States cohort.
- Author
-
Newsome RC, Gharaibeh RZ, Pierce CM, da Silva WV, Paul S, Hogue SR, Yu Q, Antonia S, Conejo-Garcia JR, Robinson LA, and Jobin C
- Subjects
- Animals, Humans, Immune Checkpoint Inhibitors, Mice, Programmed Cell Death 1 Receptor, United States, Carcinoma, Non-Small-Cell Lung, Gastrointestinal Microbiome, Lung Neoplasms drug therapy
- Abstract
Background: Recent studies show that human gut microbial composition can determine whether a patient is a responder or non-responder to immunotherapy but have not identified a common microbial signal shared by responding patients. The functional relationship between immunity, intestinal microbiota, and NSCLC response to immune checkpoint inhibitor/inhibition (ICI) in an American cohort remains unexplored., Methods: RNAlater-preserved fecal samples were collected from 65 pre-treatment (baseline) and post-treatment stage III/IV NSCLC patients undergoing ICI therapy, categorized as responders or non-responders according to RECIST criteria. Pooled and individual responder and non-responder microbiota were transplanted into a gnotobiotic mouse model of lung cancer and treated with ICIs. 16S rDNA and RNA sequencing was performed on patient fecal samples, 16S rDNA sequencing on mouse fecal samples, and flow cytometric analysis on mouse tumor tissue., Results: Responder patients have both a different microbial community structure than non-responders (P = 0.004) and a different bacterial transcriptome (PC2 = 0.03) at baseline. Taxa significantly enriched in responders include amplicon sequence variants (ASVs) belonging to the genera Ruminococcus, Akkermansia, and Faecalibacterium. Pooled and individual responder microbiota transplantation into gnotobiotic mice decreased tumor growth compared to non-responder colonized mice following ICI (P = 0.023, P = 0.019, P = 0.008, respectively). Responder tumors showed an increased anti-tumor cellular phenotype following ICI treatment. Responder mice are enriched with ASVs belonging to the genera Bacteroides, Blautia, Akkermansia, and Faecalibacterium. Overlapping taxa mapping between human and mouse cohorts correlated with tumor size and weight revealed a network highlighting responder-associated ASVs belonging to the genera Colidextribacter, Frisingicoccus, Marvinbryantia, and Blautia which have not yet been reported., Conclusions: The role of isolate-specific function and bacterial gene expression in gut microbial-driven responsiveness to ICI has been underappreciated. This work supports further investigation using isolate-driven models to characterize the mechanisms underlying this phenomenon., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
50. IgA-Dominated Humoral Immune Responses Govern Patients' Outcome in Endometrial Cancer.
- Author
-
Mandal G, Biswas S, Anadon CM, Yu X, Gatenbee CD, Prabhakaran S, Payne KK, Chaurio RA, Martin A, Innamarato P, Moran C, Powers JJ, Harro CM, Mine JA, Sprenger KB, Rigolizzo KE, Wang X, Curiel TJ, Rodriguez PC, Anderson AR, Saglam O, and Conejo-Garcia JR
- Subjects
- B-Lymphocytes metabolism, Female, Humans, Immunity, Humoral, Immunoglobulin A metabolism, Tumor Microenvironment, Endometrial Neoplasms pathology, Receptors, Polymeric Immunoglobulin genetics, Receptors, Polymeric Immunoglobulin metabolism
- Abstract
Recent studies suggest that B cells could play an important role in the tumor microenvironment. However, the role of humoral responses in endometrial cancer remains insufficiently investigated. Using a cohort of 107 patients with different histological subtypes of endometrial carcinoma, we evaluated the role of coordinated humoral and cellular adaptive immune responses in endometrial cancer. Concomitant accumulation of T, B, and plasma cells at tumor beds predicted better survival. However, only B-cell markers corresponded with prolonged survival specifically in high-grade endometrioid type and serous tumors. Immune protection was associated with class-switched IgA and, to a lesser extent, IgG. Expressions of polymeric immunoglobulin receptor (pIgR) by tumor cells and its occupancy by IgA were superior predictors of outcome and correlated with defects in methyl-directed DNA mismatch repair. Mechanistically, pIgR-dependent, antigen-independent IgA occupancy drove activation of inflammatory pathways associated with IFN and TNF signaling in tumor cells, along with apoptotic and endoplasmic reticulum stress pathways, while thwarting DNA repair mechanisms. Together, these findings suggest that coordinated humoral and cellular immune responses, characterized by IgA:pIgR interactions in tumor cells, determine the progression of human endometrial cancer as well as the potential for effective immunotherapies., Significance: This study provides new insights into the crucial role of humoral immunity in human endometrial cancer, providing a rationale for designing novel immunotherapies against this prevalent malignancy. See related commentary by Osorio and Zamarin, p. 766., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.