201
|
Garcia-Sampedro A, Gaggia G, Ney A, Mahamed I, Acedo P. The State-of-the-Art of Phase II/III Clinical Trials for Targeted Pancreatic Cancer Therapies. J Clin Med 2021; 10:566. [PMID: 33546207 PMCID: PMC7913382 DOI: 10.3390/jcm10040566] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a devastating disease with very poor prognosis. Currently, surgery followed by adjuvant chemotherapy represents the only curative option which, unfortunately, is only available for a small group of patients. The majority of pancreatic cancer cases are diagnosed at advanced or metastatic stage when surgical resection is not possible and treatment options are limited. Thus, novel and more effective therapeutic strategies are urgently needed. Molecular profiling together with targeted therapies against key hallmarks of pancreatic cancer appear as a promising approach that could overcome the limitations of conventional chemo- and radio-therapy. In this review, we focus on the latest personalised and multimodal targeted therapies currently undergoing phase II or III clinical trials. We discuss the most promising findings of agents targeting surface receptors, angiogenesis, DNA damage and cell cycle arrest, key signalling pathways, immunotherapies, and the tumour microenvironment.
Collapse
Affiliation(s)
| | | | | | | | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, London NW3 2QG, UK; (A.G.-S.); (G.G.); (A.N.); (I.M.)
| |
Collapse
|
202
|
Cachot A, Bilous M, Liu YC, Li X, Saillard M, Cenerenti M, Rockinger GA, Wyss T, Guillaume P, Schmidt J, Genolet R, Ercolano G, Protti MP, Reith W, Ioannidou K, de Leval L, Trapani JA, Coukos G, Harari A, Speiser DE, Mathis A, Gfeller D, Altug H, Romero P, Jandus C. Tumor-specific cytolytic CD4 T cells mediate immunity against human cancer. SCIENCE ADVANCES 2021; 7:7/9/eabe3348. [PMID: 33637530 PMCID: PMC7909889 DOI: 10.1126/sciadv.abe3348] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/14/2021] [Indexed: 05/04/2023]
Abstract
CD4 T cells have been implicated in cancer immunity for their helper functions. Moreover, their direct cytotoxic potential has been shown in some patients with cancer. Here, by mining single-cell RNA-seq datasets, we identified CD4 T cell clusters displaying cytotoxic phenotypes in different human cancers, resembling CD8 T cell profiles. Using the peptide-MHCII-multimer technology, we confirmed ex vivo the presence of cytolytic tumor-specific CD4 T cells. We performed an integrated phenotypic and functional characterization of these cells, down to the single-cell level, through a high-throughput nanobiochip consisting of massive arrays of picowells and machine learning. We demonstrated a direct, contact-, and granzyme-dependent cytotoxic activity against tumors, with delayed kinetics compared to classical cytotoxic lymphocytes. Last, we found that this cytotoxic activity was in part dependent on SLAMF7. Agonistic engagement of SLAMF7 enhanced cytotoxicity of tumor-specific CD4 T cells, suggesting that targeting these cells might prove synergistic with other cancer immunotherapies.
Collapse
Affiliation(s)
- Amélie Cachot
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Mariia Bilous
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Yen-Cheng Liu
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Xiaokang Li
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Margaux Saillard
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Mara Cenerenti
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, CH-1211, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, CH-1066, Switzerland
| | - Georg Alexander Rockinger
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Tania Wyss
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Philippe Guillaume
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Julien Schmidt
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Raphaël Genolet
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Giuseppe Ercolano
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, CH-1211, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, CH-1066, Switzerland
| | - Maria Pia Protti
- Tumor Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Walter Reith
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, CH-1211, Switzerland
| | - Kalliopi Ioannidou
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, CH-1011, Switzerland
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, CH-1011, Switzerland
| | - Joseph A Trapani
- Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne 3000, Australia
| | - George Coukos
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Alexandre Harari
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Daniel E Speiser
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Alexander Mathis
- Harvard University, Cambridge, MA, USA
- Center for Neuroprosthetics, Center for Intelligent Systems, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, CH-1015, Switzerland
| | - David Gfeller
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Hatice Altug
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Pedro Romero
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, CH-1211, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, CH-1066, Switzerland
| |
Collapse
|
203
|
Abstract
Despite high mortality rates, molecular understanding of metastasis remains limited. It can be regulated by both pro- and anti-metastasis genes. The metastasis suppressor, breast cancer metastasis suppressor 1 (BRMS1), has been positively correlated with patient outcomes, but molecular functions are still being characterized. BRMS1 has been implicated in focal adhesion kinase (FAK), epidermal growth factor receptor (EGFR), and NF-κB signaling pathways. We review evidence that BRMS1 regulates these vast signaling pathways through chromatin remodeling as a member of mSin3 histone deacetylase complexes.
Collapse
|
204
|
Schmiechen ZC, Stromnes IM. Mechanisms Governing Immunotherapy Resistance in Pancreatic Ductal Adenocarcinoma. Front Immunol 2021; 11:613815. [PMID: 33584701 PMCID: PMC7876239 DOI: 10.3389/fimmu.2020.613815] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/10/2020] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal malignancy with an overall 5-year survival rate of 10%. Disease lethality is due to late diagnosis, early metastasis and resistance to therapy, including immunotherapy. PDA creates a robust fibroinflammatory tumor microenvironment that contributes to immunotherapy resistance. While previously considered an immune privileged site, evidence demonstrates that in some cases tumor antigen-specific T cells infiltrate and preferentially accumulate in PDA and are central to tumor cell clearance and long-term remission. Nonetheless, PDA can rapidly evade an adaptive immune response using a myriad of mechanisms. Mounting evidence indicates PDA interferes with T cell differentiation into potent cytolytic effector T cells via deficiencies in naive T cell priming, inducing T cell suppression or promoting T cell exhaustion. Mechanistic research indicates that immunotherapy combinations that change the suppressive tumor microenvironment while engaging antigen-specific T cells is required for treatment of advanced disease. This review focuses on recent advances in understanding mechanisms limiting T cell function and current strategies to overcome immunotherapy resistance in PDA.
Collapse
Affiliation(s)
- Zoe C. Schmiechen
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Ingunn M. Stromnes
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN, United States
- Center for Genome Engineering, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
205
|
Freen-van Heeren JJ. Toll-like receptor-2/7-mediated T cell activation: An innate potential to augment CD8 + T cell cytokine production. Scand J Immunol 2021; 93:e13019. [PMID: 33377182 DOI: 10.1111/sji.13019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/10/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022]
Abstract
CD8+ T cells are critical to combat pathogens and eradicate malignantly transformed cells. To exert their effector function and kill target cells, T cells produce copious amounts of effector molecules, including the pro-inflammatory cytokines interferon γ, tumour necrosis factor α and interleukin 2. TCR triggering alone is sufficient to induce cytokine secretion by effector and memory CD8+ T cells. However, T cells can also be directly activated by pathogen-derived molecules, such as through the triggering of Toll-like receptors (TLRs). TLR-mediated pathogen sensing by T cells results in the production of only interferon γ. However, in particular when the antigen load on target cells is low, or when TCR affinity to the antigen is limited, antigen-experienced T cells can benefit from costimulatory signals. TLR stimulation can also function in a costimulatory fashion to enhance TCR triggering. Combined TCR and TLR triggering enhances the proliferation, memory formation and effector function of T cells, resulting in enhanced production of interferon γ, tumour necrosis factor α and interleukin 2. Therefore, TLR ligands or the exploitation of TLR signalling could provide novel opportunities for immunotherapy approaches. In fact, CD19 CAR T cells bearing an intracellular TLR2 costimulatory domain were recently employed to treat cancer patients in a clinical trial. Here, the current knowledge regarding TLR2/7 stimulation-induced cytokine production by T cells is reviewed. Specifically, the transcriptional and post-transcriptional pathways engaged upon TLR2/7 sensing and TLR2/7 signalling are discussed. Finally, the potential uses of TLRs to enhance the anti-tumor effector function of T cells are explored.
Collapse
|
206
|
Optimal combination treatment regimens of vaccine and radiotherapy augment tumor-bearing host immunity. Commun Biol 2021; 4:78. [PMID: 33469123 PMCID: PMC7815836 DOI: 10.1038/s42003-020-01598-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 12/14/2020] [Indexed: 01/29/2023] Open
Abstract
A major obstacle to immunotherapy is insufficient infiltration of effector immune cells into the tumor microenvironment. Radiotherapy greatly reduces tumor burden but relapses often occur. Here we show that the immunosuppressive tumor microenvironment was gradually established by recruiting Tregs after radiation. Despite tumors being controlled after depletion of Tregs in the irradiated area, improvement of mice survival remained poor. A much better antitumor effect was achieved with vaccination followed by radiation than other treatments. Vaccination followed by radiation recruited more effector T cells in tumor regions, which responded to high levels of chemokines. Sequential combination of vaccination and radiotherapy could elicit distinct host immune responses. Our study demonstrated that optimal combination of irradiation and vaccination is required to achieve effective antitumor immune responses. We propose a combination regimen that could be easily translated into the clinic and offer an opportunity for rational combination therapies design in cancer treatment.
Collapse
|
207
|
Wang Y, Mohseni M, Grauel A, Diez JE, Guan W, Liang S, Choi JE, Pu M, Chen D, Laszewski T, Schwartz S, Gu J, Mansur L, Burks T, Brodeur L, Velazquez R, Kovats S, Pant B, Buruzula G, Deng E, Chen JT, Sari-Sarraf F, Dornelas C, Varadarajan M, Yu H, Liu C, Lim J, Hao HX, Jiang X, Malamas A, LaMarche MJ, Geyer FC, McLaughlin M, Costa C, Wagner J, Ruddy D, Jayaraman P, Kirkpatrick ND, Zhang P, Iartchouk O, Aardalen K, Cremasco V, Dranoff G, Engelman JA, Silver S, Wang H, Hastings WD, Goldoni S. SHP2 blockade enhances anti-tumor immunity via tumor cell intrinsic and extrinsic mechanisms. Sci Rep 2021; 11:1399. [PMID: 33446805 PMCID: PMC7809281 DOI: 10.1038/s41598-021-80999-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
SHP2 is a ubiquitous tyrosine phosphatase involved in regulating both tumor and immune cell signaling. In this study, we discovered a novel immune modulatory function of SHP2. Targeting this protein with allosteric SHP2 inhibitors promoted anti-tumor immunity, including enhancing T cell cytotoxic function and immune-mediated tumor regression. Knockout of SHP2 using CRISPR/Cas9 gene editing showed that targeting SHP2 in cancer cells contributes to this immune response. Inhibition of SHP2 activity augmented tumor intrinsic IFNγ signaling resulting in enhanced chemoattractant cytokine release and cytotoxic T cell recruitment, as well as increased expression of MHC Class I and PD-L1 on the cancer cell surface. Furthermore, SHP2 inhibition diminished the differentiation and inhibitory function of immune suppressive myeloid cells in the tumor microenvironment. SHP2 inhibition enhanced responses to anti-PD-1 blockade in syngeneic mouse models. Overall, our study reveals novel functions of SHP2 in tumor immunity and proposes that targeting SHP2 is a promising strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Ye Wang
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Morvarid Mohseni
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Angelo Grauel
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Javier Estrada Diez
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Wei Guan
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Simon Liang
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Jiyoung Elizabeth Choi
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Minying Pu
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Dongshu Chen
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Tyler Laszewski
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Stephanie Schwartz
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Jane Gu
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Leandra Mansur
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Tyler Burks
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Lauren Brodeur
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Roberto Velazquez
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Steve Kovats
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Bhavesh Pant
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Giri Buruzula
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Emily Deng
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Julie T Chen
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Farid Sari-Sarraf
- Analytical Sciences & Imaging, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Christina Dornelas
- Analytical Sciences & Imaging, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Malini Varadarajan
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Haiyan Yu
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Chen Liu
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Joanne Lim
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Huai-Xiang Hao
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Xiaomo Jiang
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Anthony Malamas
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Matthew J LaMarche
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Felipe Correa Geyer
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Margaret McLaughlin
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Carlotta Costa
- Oncology Disease Area, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Joel Wagner
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - David Ruddy
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Pushpa Jayaraman
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | | | - Pu Zhang
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Oleg Iartchouk
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Kimberly Aardalen
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Viviana Cremasco
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Glenn Dranoff
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Jeffrey A Engelman
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Serena Silver
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Hongyun Wang
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - William D Hastings
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| | - Silvia Goldoni
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
208
|
Gutierrez WR, Scherer A, McGivney GR, Brockman QR, Knepper-Adrian V, Laverty EA, Roughton GA, Dodd RD. Divergent immune landscapes of primary and syngeneic Kras-driven mouse tumor models. Sci Rep 2021; 11:1098. [PMID: 33441747 PMCID: PMC7806664 DOI: 10.1038/s41598-020-80216-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
Immune cells play critical functions in cancer, and mice with intact immune systems are vital to understanding tumor immunology. Both genetically engineered mouse models (GEMMs) and syngeneic cell transplant approaches use immunocompetent mice to define immune-dependent events in tumor development and progression. Due to their rapid and reproducible nature, there is expanded interest in developing new syngeneic tools from established primary tumor models. However, few studies have examined the extent that syngeneic tumors reflect the immune profile of their originating primary models. Here, we describe comprehensive immunophenotyping of two well-established GEMMs and four new syngeneic models derived from these parental primary tumors. To our knowledge, this is the first systematic analysis comparing immune landscapes between primary and orthotopic syngeneic tumors. These models all use the same well-defined human-relevant driver mutations, arise at identical orthotopic locations, and are generated in mice of the same background strain. This allows for a direct and focused comparison of tumor immune landscapes in carefully controlled mouse models. We identify key differences between the immune infiltrate of GEMM models and their corresponding syngeneic tumors. Most notable is the divergence of T cell populations, with different proportions of CD8+ T cells and regulatory T cells across several models. We also observe immune variation across syngeneic tumors derived from the same primary model. These findings highlight the importance of immune variance across mouse modeling approaches, which has strong implications for the design of rigorous and reproducible translational studies.
Collapse
Grants
- P30 CA086862 NCI NIH HHS
- T32 GM007337 NIGMS NIH HHS
- T32 GM067795 NIGMS NIH HHS
- Pharmacology Training Grant, University of Iowa, United States
- Medical Scientist Training Program, University of Iowa, United States
- Holden Comprehensive Cancer Center, University of Iowa, United States
- Sarcoma Multidisciplinary Oncology Group, University of Iowa, United States
- NCI Core Grant, Holden Comprehensive Cancer Center, University of Iowa, United States
Collapse
Affiliation(s)
- Wade R Gutierrez
- Cancer Biology Graduate Program, Carver College of Medicine, University of Iowa, 285 Newton Rd, 3269C CBRB, Iowa City, IA, 52246, USA
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Amanda Scherer
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Gavin R McGivney
- Cancer Biology Graduate Program, Carver College of Medicine, University of Iowa, 285 Newton Rd, 3269C CBRB, Iowa City, IA, 52246, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Qierra R Brockman
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA
| | | | - Emily A Laverty
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Grace A Roughton
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Rebecca D Dodd
- Cancer Biology Graduate Program, Carver College of Medicine, University of Iowa, 285 Newton Rd, 3269C CBRB, Iowa City, IA, 52246, USA.
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
- Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA.
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
209
|
George JT, Levine H. Implications of Tumor-Immune Coevolution on Cancer Evasion and Optimized Immunotherapy. Trends Cancer 2021; 7:373-383. [PMID: 33446448 DOI: 10.1016/j.trecan.2020.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Cancer represents a diverse collection of diseases characterized by heterogeneous cell populations that dynamically evolve in their environment. As painfully evident in cases of treatment failure and recurrence, this general feature makes identifying long-term successful therapies difficult. It is now well-established that the adaptive immune system recognizes and eliminates cancer cells, and various immunotherapeutic strategies have emerged to augment this effect. These therapies, while promising, often fail as a result of immune-specific cancer evasion. Increasingly available empirical evidence details both cancer and immune system populations pre- and post-treatment, providing rich opportunity for mathematical models of the tumor-immune interaction and subsequent co-evolution. Integrated mathematical and experimental efforts bear immediate relevance for optimized therapies and will undoubtedly accelerate our understanding of this emergent field.
Collapse
Affiliation(s)
- Jason T George
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA.
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA; Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Physics, Northeastern University, Boston, MA, USA.
| |
Collapse
|
210
|
Full-length HLA sequencing in adult T cell leukemia-lymphoma uncovers multiple gene alterations. Leukemia 2021; 35:2998-3001. [PMID: 34518643 PMCID: PMC8478651 DOI: 10.1038/s41375-021-01403-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
|
211
|
Lakoduk AM, Lee CF, Chen PH. Gain-of-"endocytic' function in mutant p53 cancer cells. Int J Biochem Cell Biol 2020; 131:105905. [PMID: 33359084 DOI: 10.1016/j.biocel.2020.105905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/07/2020] [Accepted: 12/15/2020] [Indexed: 11/24/2022]
Abstract
Beyond its well-known canonical function as a tumor suppressor, p53 is also involved in numerous cellular processes through altered transcription under both normal and pathological conditions. The functional diversity of p53 outputs is complex and dependent on cell context. However, the underlying mechanisms responsible for this diversity remain largely unclear. The emerging evidence of p53 mutations involved in regulating endocytic trafficking and signaling, in tandem to promote malignancy (invasion, exosome biogenesis and immune evasion), sheds light on possible mechanisms behind the p53-driven complexity. The interrelated nature of endocytic trafficking and receptor signaling that form dynamic and adaptable feedback loops - either positive or negative - functions to modulate multiple cellular outputs. Biasing the tunable endocytic trafficking and receptor signaling network by mutant p53 expands the purview of p53, allowing its contribution to diverse and aggressive phenotypes. In this review, we explore recent studies in which the novel role of mutant p53 in altering endocytic trafficking to bias receptor signaling and drive transforming phenotypes is revealed. Understanding the complex crosstalk of mutant p53, endocytic trafficking and receptor signaling will allow the development of therapies to selectively target p53-altered endocytic processes.
Collapse
Affiliation(s)
- Ashley M Lakoduk
- Department of Biological Sciences, The University of Texas at Dallas, Dallas, TX, 75080, United States
| | - Cheng-Fan Lee
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Ping-Hung Chen
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
| |
Collapse
|
212
|
Nafea H, Youness RA, Abou-Aisha K, Gad MZ. LncRNA HEIH/miR-939-5p interplay modulates triple-negative breast cancer progression through NOS2-induced nitric oxide production. J Cell Physiol 2020; 236:5362-5372. [PMID: 33368266 DOI: 10.1002/jcp.30234] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/17/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022]
Abstract
This study aimed to unravel the regulatory role of noncoding RNAs (ncRNA) on the nitric oxide (NO) machinery system in triple-negative breast cancer (TNBC) patients and to further assess the influence of NO-modulating ncRNAs on TNBC progression, immunogenic profile, and the tumor microenvironment (TME). The results revealed miR-939-5p and lncRNA HEIH as novel ncRNAs modulating NO machinery in TNBC. MiR-939-5p, an underexpressed microRNA (miRNA) in BC patients, showed an inhibitory effect on NOS2 and NOS3 transcript levels on TNBC cells. In contrast, HEIH was found to be markedly upregulated in TNBC patients and showed a modulatory role on miR-939-5p/NOS2/NO axis. Functionally, miR-939-5p was characterized as a tumor suppressor miRNA while HEIH was categorized as a novel oncogenic lncRNA in TNBC. Finally, knocking down of HEIH resulted in improvement of immunogenic profile of TNBC cells through inducing MICA/B and suppressing the immune checkpoint inhibitor PDL1. In the same context, knockdown of HEIH resulted in the alleviation of the immune-suppressive TME by repressing interleukin-10 and tumor necrosis factor-α levels. In conclusion, this study identifies miR-939-5p as a tumor suppressor miRNA while HEIH as an oncogenic lncRNA exhibiting its effect through miR-939-5p/NOS2/NO axis. Therefore, repressing BC hallmarks, improving TNBC immunogenic profile, and trimming TME.
Collapse
Affiliation(s)
- Heba Nafea
- Department of Biochemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo City, Egypt
| | - Rana A Youness
- Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo City, Egypt
| | - Khaled Abou-Aisha
- Department of Microbiology and Immunology, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo City, Egypt
| | - Mohamed Z Gad
- Department of Biochemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, New Cairo City, Egypt
| |
Collapse
|
213
|
Charap AJ, Enokida T, Brody R, Sfakianos J, Miles B, Bhardwaj N, Horowitz A. Landscape of natural killer cell activity in head and neck squamous cell carcinoma. J Immunother Cancer 2020; 8:jitc-2020-001523. [PMID: 33428584 PMCID: PMC7754625 DOI: 10.1136/jitc-2020-001523] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) encompasses a set of cancers arising from the epithelia of the upper aerodigestive tract, accounting for a significant burden of disease worldwide due to the disease’s mortality, morbidity, and predilection for recurrence. Prognosis of HNSCC in the recurrent and/or metastatic (R/M-HNSCC) setting is especially poor and effective treatment options increasingly rely on modulating T-cell antitumor responses. Still, immunotherapy response rates are generally low, prompting the exploration of novel strategies that incorporate other effector cells within the tumor microenvironment. Within the last decade, important advances have been made leveraging the powerful innate antitumor function of natural killer (NK) cells to treat solid tumors, including head and neck squamous cell carcinoma. NK cells are hybrid innate-adaptive effector cells capable of directly eliminating tumor cells in addition to initiating adaptive antitumor immune responses. In the setting of HNSCC, NK cells are important for tumor surveillance and control, and NK cell infiltration has repeatedly been associated with a favorable prognosis. Yet, HNSCC-infiltrating NK cells are susceptible to an array of immune evasion strategies employed by tumors that must be overcome to fully realize the antitumor potential of NK cells. We believe that a conceptual framework informed by the basic biological understanding of the mechanisms underlying NK cell activation can improve treatment of HNSCC, in part by selecting for patients most likely to respond to NK cell-based immunotherapy. Herein, we review the activity of NK cells in HNSCC, paying special attention to the role of environmental and genetic determinants of NK cell antitumor function. Moreover, we explore the evidence that NK cells are a crucial determinant of the efficacy of both established and emerging treatments for HNSCC.
Collapse
Affiliation(s)
- Andrew J Charap
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Tomohiro Enokida
- Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rachel Brody
- Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John Sfakianos
- Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Brett Miles
- Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nina Bhardwaj
- Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Amir Horowitz
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
214
|
Albertsmeier M, Altendorf-Hofmann A, Lindner LH, Issels RD, Kampmann E, Dürr HR, Schubert-Fritschle G, Angele MK, Kirchner T, Jungbluth AA, Knösel T. Cancer Testis Antigens and Immunotherapy: Expression of PRAME Is Associated with Prognosis in Soft Tissue Sarcoma. Cancers (Basel) 2020; 12:E3612. [PMID: 33287125 PMCID: PMC7761656 DOI: 10.3390/cancers12123612] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
(1) Background: PRAME, NY-ESO-1, and SSX2 are cancer testis antigens (CTAs), which are expressed in testicular germ cells with re-expression in numerous cancer types. Their ability to elicit humoral and cellular immune responses have rendered them promising targets for cancer immunotherapy, but they have never been studied in a large and well-characterised cohort of soft tissue sarcomas (STS). (2) Methods: On a protein level, we examined PRAME, NY-ESO-1, and SSX2 expression in tumour tissues of 249 high-risk STS using immunohistochemistry. We correlated expression levels with clinicopathological parameters including tumour-infiltrating lymphocyte (TIL) counts, grading, and long-term survival. (3) Results: Expression of PRAME, NY-ESO-1, and SSX2 was observed in 25 (10%), 19 (8%), and 11 (4%) of 249 specimens with distinct patterns for histo-subtypes. Expression of PRAME was associated with shorter patient survival (p = 0.005) and higher grade (G2 vs. G3, p = 0.001), while NY-ESO-1 expression was correlated with more favourable survival (p = 0.037) and lower grade (G2 vs. G3, p = 0.029). Both PRAME and NY-ESO-1 expression were more frequent in STS with low TIL counts. In multivariate analysis, high PRAME and low SSX2 expression levels as well as metastatic disease and non-radical resections were independent predictors of shorter overall survival. (4) Conclusions: CTAs PRAME, NY-ESO-1, and SSX2 show distinct expression patterns in different STS subtypes. These results demonstrate their prognostic relevance and may guide future immunotherapeutic approaches in STS.
Collapse
Affiliation(s)
- Markus Albertsmeier
- Department of General, Visceral and Transplantation Surgery, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistr. 15, 81377 Munich, Germany;
| | - Annelore Altendorf-Hofmann
- Department of General, Visceral and Vascular Surgery, Friedrich-Schiller Universität Jena, Am Klinikum 1, 07743 Jena, Germany;
| | - Lars H. Lindner
- Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.L.); (R.D.I.); (E.K.)
| | - Rolf D. Issels
- Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.L.); (R.D.I.); (E.K.)
| | - Eric Kampmann
- Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.L.); (R.D.I.); (E.K.)
| | - Hans-Roland Dürr
- Musculoskeletal Oncology, Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistr. 15, 81377 Munich, Germany;
| | - Gabriele Schubert-Fritschle
- Munich Cancer Registry (MCR) of the Munich Tumour Centre (TZM), Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistr. 15, 81377 Munich, Germany;
| | - Martin K. Angele
- Department of General, Visceral and Transplantation Surgery, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistr. 15, 81377 Munich, Germany;
| | - Thomas Kirchner
- Institute of Pathology, Ludwig-Maximilians-Universität (LMU) Munich, Thalkirchner Str. 36, 80337 Munich, Germany;
| | - Achim A. Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 1275, USA;
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-Universität (LMU) Munich, Thalkirchner Str. 36, 80337 Munich, Germany;
| |
Collapse
|
215
|
Yin P, Gui L, Wang C, Yan J, Liu M, Ji L, Wang Y, Ma B, Gao WQ. Targeted Delivery of CXCL9 and OX40L by Mesenchymal Stem Cells Elicits Potent Antitumor Immunity. Mol Ther 2020; 28:2553-2563. [PMID: 32827461 DOI: 10.1016/j.ymthe.2020.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/21/2020] [Accepted: 08/05/2020] [Indexed: 12/31/2022] Open
Abstract
Major obstacles in immunotherapies include toxicities associated with systemic administration of therapeutic agents, as well as low tumor lymphocyte infiltration that hampers the efficacies. In this study, we report a mesenchymal stem cell (MSC)-based immunotherapeutic strategy in which MSCs specifically deliver T/natural killer (NK) cell-targeting chemokine CXCL9 and immunostimulatory factor OX40 ligand (OX40L)/tumor necrosis factor superfamily member 4 (TNFSF4) to tumor sites in syngeneic subcutaneous and azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced spontaneous colon cancer mouse models. This approach generated potent local antitumor immunity by increasing the ratios of tumor-infiltrating CD8+ T and NK cells and production of antitumor cytokines and cytolytic proteins in the tumor microenvironment. Moreover, it improved the efficacy of programmed death-1 (PD-1) blockade in a syngeneic mouse model and significantly suppressed the growth of major histocompatibility complex class I (MHC class I)-deficient tumors. Our MSC-based immunotherapeutic strategy simultaneously recruits and activates immune effector cells at the tumor site, thus overcoming the problems with toxicities of systemic therapeutic agents and low lymphocyte infiltration of solid tumors.
Collapse
Affiliation(s)
- Pan Yin
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liming Gui
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Caihong Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jingjing Yan
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Min Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lu Ji
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - You Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bin Ma
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
216
|
Evolving insights into the genomic complexity and immune landscape of diffuse large B-cell lymphoma: opportunities for novel biomarkers. Mod Pathol 2020; 33:2422-2436. [PMID: 32620919 DOI: 10.1038/s41379-020-0616-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022]
Abstract
Recently, comprehensive genomic analyses have allowed a better molecular characterization of diffuse large B-cell lymphoma (DLBCL), offering novel opportunities in patient risk stratification and management. In the era of precision medicine, this has allowed us to move closer toward a more promising therapeutic outcome in the setting of DLBCL. In this review, we highlight the newly reported heterogeneous mutational landscapes of DLBCL (from two whole-exome sequencing studies, and from a more recent work targeting a 293-gene of a hematologic malignancy-designed panel. Altogether, these studies provide further evidence of the clinical applicability of genomic tests. We also briefly review established biomarkers in DLBCL (e.g., MYC and TP53), and our understanding of the germinal center cell reaction, including its epigenetic regulation, emphasizing some of the key epigenetic modifiers that play a role in lymphomagenesis, with available therapeutic targets. In addition, we present current data regarding the role of immune landscapes in DLBCL (inflamed versus non-inflamed), how the recently defined molecular DLBCL subtypes may affect the cellular composition of the tumor microenvironment and the function of the immune cells, and how this new knowledge may result in promising therapeutic approaches in the near future.
Collapse
|
217
|
Potts MA, McDonald JA, Sutherland KD, Herold MJ. Critical cancer vulnerabilities identified by unbiased CRISPR/Cas9 screens inform on efficient cancer Immunotherapy. Eur J Immunol 2020; 50:1871-1884. [PMID: 33202035 DOI: 10.1002/eji.202048712] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/21/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022]
Abstract
The mutational landscape of human cancers is highly complex. While next generation sequencing aims to comprehensively catalogue somatic alterations in tumor cells, it fails to delineate driver from passenger mutations. Functional genomic approaches, particularly CRISPR/Cas9, enable both gene discovery, and annotation of gene function. Indeed, recent CRISPR/Cas9 technologies have flourished with the development of more sophisticated and versatile platforms capable of gene knockouts to high throughput genome wide editing of a single nucleotide base. With new platforms constantly emerging, it can be challenging to navigate what CRISPR tools are available and how they can be effectively applied to understand cancer biology. This review provides an overview of current and emerging CRISPR technologies and their power to model cancer and identify novel treatments. Specifically, how CRISPR screening approaches have been exploited to enhance immunotherapies through the identification of tumor intrinsic and extrinsic mechanisms to escape immune recognition will be discussed.
Collapse
Affiliation(s)
- Margaret A Potts
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Jackson A McDonald
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Kate D Sutherland
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
218
|
Gallo C, Barra G, Saponaro M, Manzo E, Fioretto L, Ziaco M, Nuzzo G, d’Ippolito G, De Palma R, Fontana A. A New Bioassay Platform Design for the Discovery of Small Molecules with Anticancer Immunotherapeutic Activity. Mar Drugs 2020; 18:E604. [PMID: 33260400 PMCID: PMC7760914 DOI: 10.3390/md18120604] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/18/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy takes advantage of the immune system to prevent, control, and eliminate neoplastic cells. The research in the field has already led to major breakthroughs to treat cancer. In this work, we describe a platform that integrates in vitro bioassays to test the immune response and direct antitumor effects for the preclinical discovery of anticancer candidates. The platform relies on the use of dendritic cells that are professional antigen-presenting cells (APC) able to activate T cells and trigger a primary adaptive immune response. The experimental procedure is based on two phenotypic assays for the selection of chemical leads by both a panel of nine tumor cell lines and growth factor-dependent immature mouse dendritic cells (D1). The positive hits are then validated by a secondary test on human monocyte-derived dendritic cells (MoDCs). The aim of this approach is the selection of potential immunotherapeutic small molecules from natural extracts or chemical libraries.
Collapse
Affiliation(s)
- Carmela Gallo
- Bio-Organic Chemistry Unit, CNR-Institute of Biomolecular Chemistry, Via Campi Flegrei 34, 80078 Naples, Italy; (G.B.); (M.S.); (E.M.); (G.N.); (G.d.); (R.D.P.)
| | - Giusi Barra
- Bio-Organic Chemistry Unit, CNR-Institute of Biomolecular Chemistry, Via Campi Flegrei 34, 80078 Naples, Italy; (G.B.); (M.S.); (E.M.); (G.N.); (G.d.); (R.D.P.)
| | - Marisa Saponaro
- Bio-Organic Chemistry Unit, CNR-Institute of Biomolecular Chemistry, Via Campi Flegrei 34, 80078 Naples, Italy; (G.B.); (M.S.); (E.M.); (G.N.); (G.d.); (R.D.P.)
| | - Emiliano Manzo
- Bio-Organic Chemistry Unit, CNR-Institute of Biomolecular Chemistry, Via Campi Flegrei 34, 80078 Naples, Italy; (G.B.); (M.S.); (E.M.); (G.N.); (G.d.); (R.D.P.)
| | - Laura Fioretto
- Consorzio Italbiotec, Via Fantoli, 16/15, 20138 Milan, Italy;
| | - Marcello Ziaco
- BioSearch Srl., Villa Comunale c/o Stazione Zoologica “A.Dohrn”, 80121 Naples, Italy;
| | - Genoveffa Nuzzo
- Bio-Organic Chemistry Unit, CNR-Institute of Biomolecular Chemistry, Via Campi Flegrei 34, 80078 Naples, Italy; (G.B.); (M.S.); (E.M.); (G.N.); (G.d.); (R.D.P.)
| | - Giuliana d’Ippolito
- Bio-Organic Chemistry Unit, CNR-Institute of Biomolecular Chemistry, Via Campi Flegrei 34, 80078 Naples, Italy; (G.B.); (M.S.); (E.M.); (G.N.); (G.d.); (R.D.P.)
| | - Raffaele De Palma
- Bio-Organic Chemistry Unit, CNR-Institute of Biomolecular Chemistry, Via Campi Flegrei 34, 80078 Naples, Italy; (G.B.); (M.S.); (E.M.); (G.N.); (G.d.); (R.D.P.)
- Internal Medicine, Clinical Immunology and Translational Medicine, University of Genova and IRCCS-Hospital S. Martino, 16132 Genova, Italy
| | - Angelo Fontana
- Bio-Organic Chemistry Unit, CNR-Institute of Biomolecular Chemistry, Via Campi Flegrei 34, 80078 Naples, Italy; (G.B.); (M.S.); (E.M.); (G.N.); (G.d.); (R.D.P.)
- Department of Biology, University of Naples Federico II, Via Vicinale Cupa Cintia 21, 80126 Naples, Italy
| |
Collapse
|
219
|
Berendsen MR, Stevens WBC, van den Brand M, van Krieken JH, Scheijen B. Molecular Genetics of Relapsed Diffuse Large B-Cell Lymphoma: Insight into Mechanisms of Therapy Resistance. Cancers (Basel) 2020; 12:E3553. [PMID: 33260693 PMCID: PMC7760867 DOI: 10.3390/cancers12123553] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
The majority of patients with diffuse large B-cell lymphoma (DLBCL) can be treated successfully with a combination of chemotherapy and the monoclonal anti-CD20 antibody rituximab. Nonetheless, approximately one-third of the patients with DLBCL still experience relapse or refractory (R/R) disease after first-line immunochemotherapy. Whole-exome sequencing on large cohorts of primary DLBCL has revealed the mutational landscape of DLBCL, which has provided a framework to define novel prognostic subtypes in DLBCL. Several studies have investigated the genetic alterations specifically associated with R/R DLBCL, thereby uncovering molecular pathways linked to therapy resistance. Here, we summarize the current state of knowledge regarding the genetic alterations that are enriched in R/R DLBCL, and the corresponding pathways affected by these gene mutations. Furthermore, we elaborate on their potential role in mediating therapy resistance, also in connection with findings in other B-cell malignancies, and discuss alternative treatment options. Hence, this review provides a comprehensive overview on the gene lesions and molecular mechanisms underlying R/R DLBCL, which are considered valuable parameters to guide treatment.
Collapse
Affiliation(s)
- Madeleine R. Berendsen
- Department of Pathology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (M.R.B.); (M.v.d.B.); (J.H.v.K.)
- Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| | - Wendy B. C. Stevens
- Department of Hematology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands;
| | - Michiel van den Brand
- Department of Pathology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (M.R.B.); (M.v.d.B.); (J.H.v.K.)
- Pathology-DNA, Rijnstate Hospital, 6815AD Arnhem, The Netherlands
| | - J. Han van Krieken
- Department of Pathology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (M.R.B.); (M.v.d.B.); (J.H.v.K.)
| | - Blanca Scheijen
- Department of Pathology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (M.R.B.); (M.v.d.B.); (J.H.v.K.)
- Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| |
Collapse
|
220
|
Cortés-Selva D, Dasgupta B, Singh S, Grewal IS. Innate and Innate-Like Cells: The Future of Chimeric Antigen Receptor (CAR) Cell Therapy. Trends Pharmacol Sci 2020; 42:45-59. [PMID: 33250273 DOI: 10.1016/j.tips.2020.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Conventional αβ CAR-T cell-based approaches have revolutionized the field of cancer immunotherapy, but hurdles remain, especially for solid tumors. Novel strategies in conjunction with alternative cell types are therefore required for effective CAR-based therapies. In this respect, innate and innate-like cells with unique immune properties, such as natural killer (NK) cells, NKT cells, γδ T cells, and macrophages, are promising alternatives to αβ CAR-T adoptive therapy. We review the applicability of these cells in the context of CAR therapy, focusing on therapies under development, the advantages of these approaches relative to conventional CAR-T cells, and their potential in allogeneic therapies. We also discuss the inherent limitations of these cell types and approaches, and outline numerous strategies to overcome the associated obstacles.
Collapse
Affiliation(s)
- Diana Cortés-Selva
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Bidisha Dasgupta
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Sanjaya Singh
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Iqbal S Grewal
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA.
| |
Collapse
|
221
|
Abstract
Initially identified as a T lymphocyte-elicited inhibitor of macrophage motility, macrophage migration inhibitory factor (MIF) has since been found to be expressed by nearly every immune cell type examined and overexpressed in most solid and hematogenous malignant cancers. It is localized to both extracellular and intracellular compartments and physically interacts with more than a dozen different cell surface and intracellular proteins. Although classically associated with and characterized as a mediator of pro-inflammatory innate immune responses, more recent studies demonstrate that, in malignant disease settings, MIF contributes to anti-inflammatory, immune evasive, and immune tolerant phenotypes in both innate and adaptive immune cell types. This review will summarize the studies describing MIF in tumor-specific innate and adaptive immune responses and attempt to reconcile these various pleiotropic functions in normal physiology.
Collapse
Affiliation(s)
- Jordan T. Noe
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, United States
- J.G. Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Robert A. Mitchell
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, United States
- J.G. Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, United States
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| |
Collapse
|
222
|
Wieczorek E, Garstka MA. Recurrent bladder cancer in aging societies: Importance of major histocompatibility complex class I antigen presentation. Int J Cancer 2020; 148:1808-1820. [PMID: 33105025 DOI: 10.1002/ijc.33359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/24/2022]
Abstract
Aging is associated with an insufficient immune response that may lead to the initiation and progression of various malignancies. Bladder cancer (BC), prevalent in elderly patients, predominantly presents as recurrent nonmuscle invasive BC that requires further treatment. There is much interest in the activation of patients' immune cells with the focus on CD8+ T cells. Successful therapy should also ensure the efficient presentation of BC antigens by major histocompatibility complex (MHC) class I molecules. The purpose of this systematic review is to present the existing literature on the role of MHC class I in BC research and therapy. The bibliographic databases PubMed and Web of Science were searched for articles published between January 2009 and September 2020 that addressed MHC class I relationship to BC. We searched for available relevant publications on MHC class I and its role and regulation in BC, aging and MHC class I importance in BC immunotherapy. Based on the provided evidence, we propose that the loss of MHC class I expression in BC may lead to its recurrence after the transurethral resection and unresponsiveness to Bacillus Calmette-Guerin immunotherapy. We discuss different ways to enhance MHC class I antigen presentation to CD8+ T cells in BC treatment. The immune status characterized by MHC class I expression patterns and cancer-infiltrating immune cells may provide valuable prognostic information about which patients may benefit from transurethral resection of BC and additional immunotherapy.
Collapse
Affiliation(s)
- Edyta Wieczorek
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Malgorzata A Garstka
- Core Research Laboratory, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China.,Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China.,Department of Tumor and Immunology, Precision Medical Institute, Western China Science and Technology Innovation Port, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
223
|
Krawczyk PA, Laub M, Kozik P. To Kill But Not Be Killed: Controlling the Activity of Mammalian Pore-Forming Proteins. Front Immunol 2020; 11:601405. [PMID: 33281828 PMCID: PMC7691655 DOI: 10.3389/fimmu.2020.601405] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/20/2020] [Indexed: 01/01/2023] Open
Abstract
Pore-forming proteins (PFPs) are present in all domains of life, and play an important role in host-pathogen warfare and in the elimination of cancers. They can be employed to deliver specific effectors across membranes, to disrupt membrane integrity interfering with cell homeostasis, and to lyse membranes either destroying intracellular organelles or entire cells. Considering the destructive potential of PFPs, it is perhaps not surprising that mechanisms controlling their activity are remarkably complex, especially in multicellular organisms. Mammalian PFPs discovered to date include the complement membrane attack complex (MAC), perforins, as well as gasdermins. While the primary function of perforin-1 and gasdermins is to eliminate infected or cancerous host cells, perforin-2 and MAC can target pathogens directly. Yet, all mammalian PFPs are in principle capable of generating pores in membranes of healthy host cells which-if uncontrolled-could have dire, and potentially lethal consequences. In this review, we will highlight the strategies employed to protect the host from destruction by endogenous PFPs, while enabling timely and efficient elimination of target cells.
Collapse
Affiliation(s)
- Patrycja A Krawczyk
- MRC Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Marco Laub
- MRC Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Patrycja Kozik
- MRC Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
224
|
Knott EL, Leidenheimer NJ. A Targeted Bioinformatics Assessment of Adrenocortical Carcinoma Reveals Prognostic Implications of GABA System Gene Expression. Int J Mol Sci 2020; 21:ijms21228485. [PMID: 33187258 PMCID: PMC7697095 DOI: 10.3390/ijms21228485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare but deadly cancer for which few treatments exist. Here, we have undertaken a targeted bioinformatics study of The Cancer Genome Atlas (TCGA) ACC dataset focusing on the 30 genes encoding the γ-aminobutyric acid (GABA) system—an under-studied, evolutionarily-conserved system that is an emerging potential player in cancer progression. Our analysis identified a subset of ACC patients whose tumors expressed a distinct GABA system transcriptome. Transcript levels of ABAT (encoding a key GABA shunt enzyme), were upregulated in over 40% of tumors, and this correlated with several favorable clinical outcomes including patient survival; while enrichment and ontology analysis implicated two cancer-related biological pathways involved in metastasis and immune response. The phenotype associated with ABAT upregulation revealed a potential metabolic heterogeneity among ACC tumors associated with enhanced mitochondrial metabolism. Furthermore, many GABAA receptor subunit-encoding transcripts were expressed, including two (GABRB2 and GABRD) prognostic for patient survival. Transcripts encoding GABAB receptor subunits and GABA transporters were also ubiquitously expressed. The GABA system transcriptome of ACC tumors is largely mirrored in the ACC NCI-H295R cell line, suggesting that this cell line may be appropriate for future functional studies investigating the role of the GABA system in ACC cell growth phenotypes and metabolism.
Collapse
|
225
|
Land CA, Musich PR, Haydar D, Krenciute G, Xie Q. Chimeric antigen receptor T-cell therapy in glioblastoma: charging the T cells to fight. J Transl Med 2020; 18:428. [PMID: 33176788 PMCID: PMC7659102 DOI: 10.1186/s12967-020-02598-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant brain cancer that invades normal brain tissue and impedes surgical eradication, resulting in early local recurrence and high mortality. In addition, most therapeutic agents lack permeability across the blood brain barrier (BBB), further reducing the efficacy of chemotherapy. Thus, effective treatment against GBM requires tumor specific targets and efficient intracranial drug delivery. With the most recent advances in immunotherapy, genetically engineered T cells with chimeric antigen receptors (CARs) are becoming a promising approach for treating cancer. By transducing T lymphocytes with CAR constructs containing a tumor-associated antigen (TAA) recognition domain linked to the constant regions of a signaling T cell receptor, CAR T cells may recognize a predefined TAA with high specificity in a non-MHC restricted manner, and is independent of antigen processing. Active T cells can travel across the BBB, providing additional advantage for drug delivery and tumor targeting. Here we review the CAR design and technical innovations, the major targets that are in pre-clinical and clinical development with a focus on GBM, and multiple strategies developed to improve CAR T cell efficacy.
Collapse
Affiliation(s)
- Craig A. Land
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Phillip R. Musich
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Dalia Haydar
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Qian Xie
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| |
Collapse
|
226
|
Cheng CC, Lin HC, Chiang YW, Chang J, Sie ZL, Yang BL, Lim KH, Peng CL, Ho AS, Chang YF. Nicotine exhausts CD8 + T cells against tumor cells through increasing miR-629-5p to repress IL2RB-mediated granzyme B expression. Cancer Immunol Immunother 2020; 70:1351-1364. [PMID: 33146402 DOI: 10.1007/s00262-020-02770-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023]
Abstract
The mechanism exhausting CD8+ T cells is not completely clear against tumors. Literature has demonstrated that cigarette smoking disables the immunological activity, so we propose nicotine is able to exhaust CD8+ T cells. The CD8+ T cells from healthy volunteers with and without cigarette smoking and the capacity of CD8+ T cells against tumor cells were investigated. RNAseq was used to investigate the gene profiling expression in CD8+ T cells. Meanwhile, small RNAseq was also used to search novel microRNAs involved in the exhaustion of CD8+ T cells. The effect of nicotine exhausting CD8+ T cells was investigated in vitro and in the humanized tumor xenografts in vivo. We found that CD8+ T cells were able to reduce cell viability in lung cancer HCC827 and A549 cells, that secreted granzyme B, but CD8+ T cells from the healthy cigarette smokers lost anti-HCC827 effect. Moreover, nicotine suppressed the anti-HCC827 effect of CD8+ T cells. RNAseq revealed lower levels of IL2RB and GZMB in the exhausted CD8+ T cells. We identified that miR-629-5p was increased by nicotine, that targeted IL2RB. Transfection of miR-629-5p mimic reduced IL2RB and GZMB levels. We further validated that nicotine reduced granzyme B levels using a nuclear imaging technique, and demonstrated that nicotine exhausted peripheral blood mononuclear cells against HCC827 growth in the humanized tumor xenografts. This study demonstrated that nicotine exhausted CD8+ T cells against HCC827 cells through increasing miR-629-5p to suppress IL2RB.
Collapse
Affiliation(s)
- Chun-Chia Cheng
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Linkou, Taiwan
| | - Hsin-Chi Lin
- Division of Gastroenterology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Ya-Wen Chiang
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, Mackay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Jungshan Chang
- Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Zong-Lin Sie
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Linkou, Taiwan
| | - Bi-Ling Yang
- Division of Gastroenterology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Ken-Hong Lim
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, Mackay Memorial Hospital, Tamsui District, New Taipei City, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Cheng-Liang Peng
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Ai-Sheng Ho
- Division of Gastroenterology, Cheng Hsin General Hospital, Taipei, Taiwan.
| | - Yi-Fang Chang
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan. .,Department of Medical Research, Laboratory of Good Clinical Research Center, Mackay Memorial Hospital, Tamsui District, New Taipei City, Taiwan. .,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.
| |
Collapse
|
227
|
Graham JC, Hillegass J, Schulze G. Considerations for setting occupational exposure limits for novel pharmaceutical modalities. Regul Toxicol Pharmacol 2020; 118:104813. [PMID: 33144077 PMCID: PMC7605856 DOI: 10.1016/j.yrtph.2020.104813] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/13/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022]
Abstract
In order to develop new and effective medicines, pharmaceutical companies must be modality agnostic. As science reveals an enhanced understanding of biological processes, new therapeutic modalities are becoming important in developing breakthrough therapies to treat both rare and common diseases. As these new modalities progress, concern and uncertainty arise regarding their safe handling by the researchers developing them, employees manufacturing them and nurses administering them. This manuscript reviews the available literature for emerging modalities (including oligonucleotides, monoclonal antibodies, fusion proteins and bispecific antibodies, antibody-drug conjugates, peptides, vaccines, genetically modified organisms, and several others) and provides considerations for occupational health and safety-oriented hazard identification and risk assessments to enable timely, consistent and well-informed hazard identification, hazard communication and risk-management decisions. This manuscript also points out instances where historical exposure control banding systems may not be applicable (e.g. oncolytic viruses, biologics) and where other occupational exposure limit systems are more applicable (e.g. Biosafety Levels, Biologic Control Categories). Review of toxicology and pharmacology information for novel therapeutic modalities. Identification of occupational hazards associated with novel therapeutic modalities. Occupational hazards and exposure risks differ across pharmaceutical modalities. Occupational exposure control banding systems are not one size fits all. Banding system variations offer benefits while enabling proper exposure controls.
Collapse
Affiliation(s)
- Jessica C Graham
- Bristol Myers Squibb, 1 Squibb Drive, New Brunswick, NJ, 08903, USA.
| | - Jedd Hillegass
- Bristol Myers Squibb, 1 Squibb Drive, New Brunswick, NJ, 08903, USA
| | - Gene Schulze
- Bristol Myers Squibb, 1 Squibb Drive, New Brunswick, NJ, 08903, USA
| |
Collapse
|
228
|
Biological Selenium Nano-particles Modify Immune Responses of Macrophages Exposed to Bladder Tumor Antigens. J CLUST SCI 2020. [DOI: 10.1007/s10876-020-01920-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
229
|
Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 2020; 20:651-668. [PMID: 32433532 PMCID: PMC7238960 DOI: 10.1038/s41577-020-0306-5] [Citation(s) in RCA: 2268] [Impact Index Per Article: 453.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
The T lymphocyte, especially its capacity for antigen-directed cytotoxicity, has become a central focus for engaging the immune system in the fight against cancer. Basic science discoveries elucidating the molecular and cellular biology of the T cell have led to new strategies in this fight, including checkpoint blockade, adoptive cellular therapy and cancer vaccinology. This area of immunological research has been highly active for the past 50 years and is now enjoying unprecedented bench-to-bedside clinical success. Here, we provide a comprehensive historical and biological perspective regarding the advent and clinical implementation of cancer immunotherapeutics, with an emphasis on the fundamental importance of T lymphocyte regulation. We highlight clinical trials that demonstrate therapeutic efficacy and toxicities associated with each class of drug. Finally, we summarize emerging therapies and emphasize the yet to be elucidated questions and future promise within the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Alex D Waldman
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jill M Fritz
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael J Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- Clinical Genomics Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
230
|
Bullock TNJ. Fundamentals of Cancer Immunology and Their Application to Cancer Vaccines. Clin Transl Sci 2020; 14:120-131. [PMID: 32770735 PMCID: PMC7877844 DOI: 10.1111/cts.12856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/12/2020] [Indexed: 12/22/2022] Open
Abstract
The capacity of the immune system to influence tumor progression has been a long-standing notion that first generated clinical traction over a 100 years ago when Dr. William Coley injected disaggregated bacterial components into sarcomas and noted that the ensuing inflammation commonly associated with tumor regression.1 Since then, our understanding of the individual components and the overall interaction of the immune system has expanded exponentially. This has led to the development of a robust understanding of how components of innate and adaptive immunity recognize and respond to tumors and leveraging this information for the development of tumor immunotherapies. However, clinical failures have also deepened our knowledge of how tumors might adapt/be selected to avoid or inhibit immune responses, which, in turn, has led to the further iteration of immunotherapies. In this tutorial, the established elements of tumor immunity are explained, and areas where our knowledge base is too thin is highlighted. The principles of tumor immunity that guide the development of cancer vaccines are further illustrated, and potential considerations of how to integrate cancer vaccines with conventional therapies and other immunotherapies are proposed.
Collapse
Affiliation(s)
- Timothy N J Bullock
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
231
|
Ivanova M, Shivarov V. HLA genotyping meets response to immune checkpoint inhibitors prediction: A story just started. Int J Immunogenet 2020; 48:193-200. [PMID: 33112034 DOI: 10.1111/iji.12517] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/06/2020] [Accepted: 10/13/2020] [Indexed: 12/27/2022]
Abstract
The implementation of the immune checkpoint blockade as a therapeutic option in contemporary oncology is one of the significant immunological achievements in the last century. Constantly accumulating evidence suggests that the response to immune checkpoint inhibitors (ICIs) is not universal. Therefore, it is critical to identify determinants for response, resistance and adverse effects of immune checkpoint therapy that could be developed as prognostic and predictive markers. Recent large scale analyses of cancer genome data revealed the key role of HLA class I and class II molecules in cancer immunoediting, and it appears that HLA diversity can predict response to ICIs. In the present review, we summarize the emerging data on the role of HLA germline variations as a marker for response to ICIs.
Collapse
Affiliation(s)
- Milena Ivanova
- Department of Clinical Immunology, University Hospital Alexandrovska, Medical University Sofia, Sofia, Bulgaria
| | - Velizar Shivarov
- Department of Genetics, St. Kliment Ohridski University, Sofia, Bulgaria
| |
Collapse
|
232
|
Thompson JC, Davis C, Deshpande C, Hwang WT, Jeffries S, Huang A, Mitchell TC, Langer CJ, Albelda SM. Gene signature of antigen processing and presentation machinery predicts response to checkpoint blockade in non-small cell lung cancer (NSCLC) and melanoma. J Immunother Cancer 2020; 8:jitc-2020-000974. [PMID: 33028693 PMCID: PMC7542663 DOI: 10.1136/jitc-2020-000974] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2020] [Indexed: 12/31/2022] Open
Abstract
Background Limited data exist on the role of alterations in HLA Class I antigen processing and presentation machinery in mediating response to immune checkpoint blockade (ICB). Methods This retrospective cohort study analyzed transcriptional profiles from pre-treatment tumor samples of 51 chemotherapy-refractory advanced non-small cell lung cancer (NSCLC) patients and two independent melanoma cohorts treated with ICB. An antigen processing machinery (APM) score was generated utilizing eight genes associated with APM (B2M, CALR, NLRC5, PSMB9, PSME1, PSME3, RFX5, and HSP90AB1). Associations were made for therapeutic response, progression-free survival (PFS) and overall survival (OS). Results In NSCLC, the APM score was significantly higher in responders compared with non-responders (p=0.0001). An APM score above the median value for the cohort was associated with improved PFS (HR 0.34 (0.18 to 0.64), p=0.001) and OS (HR 0.44 (0.23 to 0.83), p=0.006). The APM score was correlated with an inflammation score based on the established T-cell-inflamed resistance gene expression profile (Pearson’s r=0.58, p<0.0001). However, the APM score better predicted response to ICB relative to the inflammation score with area under a receiving operating characteristics curve of 0.84 and 0.70 for PFS and OS, respectively. In a cohort of 14 high-risk resectable stage III/IV melanoma patients treated with neoadjuvant anti-PD1 ICB, a higher APM score was associated with improved disease-free survival (HR: 0.08 (0.01 to 0.50), p=0.0065). In an additional independent melanoma cohort of 27 metastatic patients treated with ICB, a higher APM score was associated with improved OS (HR 0.29 (0.09 to 0.89), p=0.044). Conclusion Our data demonstrate that defects in antigen presentation may be an important feature in predicting outcomes to ICB in both lung cancer and melanoma.
Collapse
Affiliation(s)
- Jeffrey C Thompson
- Pulmonary and Critical Care, Thoracic Oncology Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christiana Davis
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Charuhas Deshpande
- Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Wei-Ting Hwang
- Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Seth Jeffries
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alexander Huang
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tara C Mitchell
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Corey J Langer
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Steven M Albelda
- Pulmonary and Critical Care, Thoracic Oncology Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
233
|
Sabbatino F, Liguori L, Polcaro G, Salvato I, Caramori G, Salzano FA, Casolaro V, Stellato C, Dal Col J, Pepe S. Role of Human Leukocyte Antigen System as A Predictive Biomarker for Checkpoint-Based Immunotherapy in Cancer Patients. Int J Mol Sci 2020; 21:ijms21197295. [PMID: 33023239 PMCID: PMC7582904 DOI: 10.3390/ijms21197295] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022] Open
Abstract
Recent advances in cancer immunotherapy have clearly shown that checkpoint-based immunotherapy is effective in a small subgroup of cancer patients. However, no effective predictive biomarker has been identified so far. The major histocompatibility complex, better known in humans as human leukocyte antigen (HLA), is a very polymorphic gene complex consisting of more than 200 genes. It has a crucial role in activating an appropriate host immune response against pathogens and tumor cells by discriminating self and non-self peptides. Several lines of evidence have shown that down-regulation of expression of HLA class I antigen derived peptide complexes by cancer cells is a mechanism of tumor immune escape and is often associated to poor prognosis in cancer patients. In addition, it has also been shown that HLA class I and II antigen expression, as well as defects in the antigen processing machinery complex, may predict tumor responses in cancer immunotherapy. Nevertheless, the role of HLA in predicting tumor responses to checkpoint-based immunotherapy is still debated. In this review, firstly, we will describe the structure and function of the HLA system. Secondly, we will summarize the HLA defects and their clinical significance in cancer patients. Thirdly, we will review the potential role of the HLA as a predictive biomarker for checkpoint-based immunotherapy in cancer patients. Lastly, we will discuss the potential strategies that may restore HLA function to implement novel therapeutic strategies in cancer patients.
Collapse
Affiliation(s)
- Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry ’Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (F.S.); (G.P.); (I.S.); (F.A.S.); (V.C.); (C.S.); (S.P.)
- Oncology Unit, AOU San Giovanni di Dio e Ruggi D’Aragona, 84131 Salerno, Italy
| | - Luigi Liguori
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Giovanna Polcaro
- Department of Medicine, Surgery and Dentistry ’Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (F.S.); (G.P.); (I.S.); (F.A.S.); (V.C.); (C.S.); (S.P.)
| | - Ilaria Salvato
- Department of Medicine, Surgery and Dentistry ’Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (F.S.); (G.P.); (I.S.); (F.A.S.); (V.C.); (C.S.); (S.P.)
- Pulmonary Unit, Department of Biomedical Sciences, Dentistry, Morphological and Functional Imaging (BIOMORF), University of Messina, 98125 Messina, Italy;
| | - Gaetano Caramori
- Pulmonary Unit, Department of Biomedical Sciences, Dentistry, Morphological and Functional Imaging (BIOMORF), University of Messina, 98125 Messina, Italy;
| | - Francesco A. Salzano
- Department of Medicine, Surgery and Dentistry ’Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (F.S.); (G.P.); (I.S.); (F.A.S.); (V.C.); (C.S.); (S.P.)
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry ’Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (F.S.); (G.P.); (I.S.); (F.A.S.); (V.C.); (C.S.); (S.P.)
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry ’Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (F.S.); (G.P.); (I.S.); (F.A.S.); (V.C.); (C.S.); (S.P.)
| | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry ’Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (F.S.); (G.P.); (I.S.); (F.A.S.); (V.C.); (C.S.); (S.P.)
- Correspondence: ; Tel.: +39-08996-5210
| | - Stefano Pepe
- Department of Medicine, Surgery and Dentistry ’Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (F.S.); (G.P.); (I.S.); (F.A.S.); (V.C.); (C.S.); (S.P.)
- Oncology Unit, AOU San Giovanni di Dio e Ruggi D’Aragona, 84131 Salerno, Italy
| |
Collapse
|
234
|
Winge-Main AK, Wälchli S, Inderberg EM. T cell receptor therapy against melanoma-Immunotherapy for the future? Scand J Immunol 2020; 92:e12927. [PMID: 32640053 DOI: 10.1111/sji.12927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022]
Abstract
Malignant melanoma has seen monumental changes in treatment options the last decade from the very poor results of dacarbazine treatment to the modern-day use of targeted therapies and immune checkpoint inhibitors. Melanoma has a high mutational burden making it more capable of evoking immune responses than many other tumours. Even when considering double immune checkpoint blockade with anti-CTLA-4 and anti-PD-1, we still have far to go in melanoma treatment as 50% of patients with metastatic disease do not respond to current treatment. Alternative immunotherapy should therefore be considered. Since melanoma has a high mutational burden, it is considered more immunogenic than many other tumours. T cell receptor (TCR) therapy could be a possible way forward, either alone or in combination, to improve the response rates of this deadly disease. Melanoma is one of the cancers where TCR therapy has been frequently applied. However, the number of antigens targeted remains fairly limited, although advanced personalized therapies aim at also targeting private mutations. In this review, we look at possible aspects of targeting TCR therapy towards melanoma and provide an implication of its use in the future.
Collapse
Affiliation(s)
- Anna K Winge-Main
- Department of Cellular Therapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Department of Cellular Therapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Department of Cellular Therapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
235
|
Gerbec ZJ, Hashemi E, Nanbakhsh A, Holzhauer S, Yang C, Mei A, Tsaih SW, Lemke A, Flister MJ, Riese MJ, Thakar MS, Malarkannan S. Conditional Deletion of PGC-1α Results in Energetic and Functional Defects in NK Cells. iScience 2020; 23:101454. [PMID: 32858341 PMCID: PMC7474003 DOI: 10.1016/j.isci.2020.101454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/30/2019] [Accepted: 08/10/2020] [Indexed: 01/07/2023] Open
Abstract
During an immune response, natural killer (NK) cells activate specific metabolic pathways to meet the increased energetic and biosynthetic demands associated with effector functions. Here, we found in vivo activation of NK cells during Listeria monocytogenes infection-augmented transcription of genes encoding mitochondria-associated proteins in a manner dependent on the transcriptional coactivator PGC-1α. Using an Ncr1Cre-based conditional knockout mouse, we found that PGC-1α was crucial for optimal NK cell effector functions and bioenergetics, as the deletion of PGC-1α was associated with decreased cytotoxic potential and cytokine production along with altered ADP/ATP ratios. Lack of PGC-1α also significantly impaired the ability of NK cells to control B16F10 tumor growth in vivo, and subsequent gene expression analysis showed that PGC-1α mediates transcription required to maintain mitochondrial activity within the tumor microenvironment. Together, these data suggest that PGC-1α-dependent transcription of specific target genes is required for optimal NK cell function during the response to infection or tumor growth.
Collapse
Affiliation(s)
- Zachary J. Gerbec
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Arash Nanbakhsh
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Sandra Holzhauer
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Chao Yang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ao Mei
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shirng-Wern Tsaih
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Angela Lemke
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael J. Flister
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew J. Riese
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Monica S. Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
236
|
Adenovirus Receptor Expression in Cancer and Its Multifaceted Role in Oncolytic Adenovirus Therapy. Int J Mol Sci 2020; 21:ijms21186828. [PMID: 32957644 PMCID: PMC7554712 DOI: 10.3390/ijms21186828] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Oncolytic adenovirus therapy is believed to be a promising way to treat cancer patients. To be able to target tumor cells with an oncolytic adenovirus, expression of the adenovirus receptor on the tumor cell is essential. Different adenovirus types bind to different receptors on the cell, of which the expression can vary between tumor types. Pre-existing neutralizing immunity to human adenovirus species C type 5 (HAdV-C5) has hampered its therapeutic efficacy in clinical trials, hence several adenoviral vectors from different species are currently being developed as a means to evade pre-existing immunity. Therefore, knowledge on the expression of appropriate adenovirus receptors on tumor cells is important. This could aid in determining which tumor types would benefit most from treatment with a certain oncolytic adenovirus type. This review provides an overview of the known receptors for human adenoviruses and how their expression on tumor cells might be differentially regulated compared to healthy tissue, before and after standardized anticancer treatments. Mechanisms behind the up- or downregulation of adenovirus receptor expression are discussed, which could be used to find new targets for combination therapy to enhance the efficacy of oncolytic adenovirus therapy. Additionally, the utility of the adenovirus receptors in oncolytic virotherapy is examined, including their role in viral spread, which might even surpass their function as primary entry receptors. Finally, future directions are offered regarding the selection of adenovirus types to be used in oncolytic adenovirus therapy in the fight against cancer.
Collapse
|
237
|
Zhu L, Yang X, Zhong D, Xie S, Shi W, Li Y, Hou X, HuaYao, Zhou H, Zhao M, Ding Z, Zhao X, Mo F, Yin S, Liu A, Lu X. Single-Domain Antibody-Based TCR-Like CAR-T: A Potential Cancer Therapy. J Immunol Res 2020; 2020:2454907. [PMID: 32964055 PMCID: PMC7492946 DOI: 10.1155/2020/2454907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/30/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022] Open
Abstract
Retargeting the antigen-binding specificity of T cells to intracellular antigens that are degraded and presented on the tumor surface by engineering chimeric antigen receptor (CAR), also named TCR-like antibody CAR-T, remains limited. With the exception of the commercialized CD19 CAR-T for hematological malignancies and other CAR-T therapies aiming mostly at extracellular antigens achieving great success, the rareness and scarcity of TCR-like CAR-T therapies might be due to their current status and limitations. This review provides the probable optimized initiatives for improving TCR-like CAR-T reprogramming and discusses single-domain antibodies administered as an alternative to conventional scFvs and secreted by CAR-T cells, which might be of great value to the development of CAR-T immunotherapies for intracellular antigens.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Epitopes, T-Lymphocyte/immunology
- Genetic Engineering
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Single-Chain Antibodies/immunology
- Single-Domain Antibodies/genetics
- Single-Domain Antibodies/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Lichen Zhu
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaomei Yang
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Dani Zhong
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- Department of Chemotherapy, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Shenxia Xie
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Wei Shi
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yangzi Li
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaoqiong Hou
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - HuaYao
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Huihui Zhou
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Minlong Zhao
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ziqiang Ding
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xinyue Zhao
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Fengzhen Mo
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Shihua Yin
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Aiqun Liu
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaoling Lu
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
238
|
Bell D, Bell A, Ferrarotto R, Glisson B, Takahashi Y, Fuller G, Weber R, Hanna E. High-grade sinonasal carcinomas and surveillance of differential expression in immune related transcriptome. Ann Diagn Pathol 2020; 49:151622. [PMID: 32927372 DOI: 10.1016/j.anndiagpath.2020.151622] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022]
Abstract
The skull base is the location of a wide variety of malignant tumors. Among them is sinonasal undifferentiated carcinoma (SNUC), a highly aggressive sinonasal neoplasm that was recently reclassified into subgroups of high-grade carcinomas with unique genomic events (e.g., SMARC-deficient carcinoma, nuclear protein in testis NUT carcinoma). Other high-grade carcinomas in this location are neuroendocrine carcinomas, sinonasal adenocarcinomas, and teratocarcinosarcomas. Given the rarity of these tumors, little transcriptomic data is available. The aim of this study was to characterize the immune-oncology gene expression profile in SNUC and other high-grade sinonasal carcinomas. Next-generation sequencing was performed in 30 high-grade sinonasal carcinoma samples using the HTG EdgeSeq Precision Immuno-Oncology Panel. Ingenuity pathway analysis was performed to understand the immunobiology, signaling, and functional perturbations during tumor development. The samples were divided into 3 groups: 21 SNUCs and SMARC-deficient sinonasal carcinomas; 5 high-grade neuroendocrine carcinomas (HGNECs), with small cell and large cell variants; and 4 high-grade sinonasal carcinomas (HGSNCs) of mixed histology (1 NUT carcinoma, 1 teratocarcinosarcoma, and 2 sinonasal adenocarcinomas). PRAME and ASCL1 emerged as upregulated transcripts with strong protein validation for SNUC and HGNEC; other upregulated candidates EZH2 and BRCA1 offer consideration for alternative targeted therapy, and downregulation of major histocompatibility complex molecules and chemokines represent another hurdle in the development of effective immunotherapy. This immune-oncology gene expression analysis of 3 groups of high-grade sinonasal carcinoma with emphasis on SNUC identified a number of differentially expressed transcripts reflecting effects on tumorigenesis. Identification of immune pathways should be further investigated for possible integration of immunotherapy into a multidisciplinary approach to these cancers and personalized treatment.
Collapse
Affiliation(s)
- Diana Bell
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States of America; Department Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States of America.
| | - Achim Bell
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States of America
| | - Renata Ferrarotto
- Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States of America
| | - Bonnie Glisson
- Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States of America
| | - Yoko Takahashi
- Department Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States of America
| | - Gregory Fuller
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States of America
| | - Randal Weber
- Department Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States of America
| | - Ehab Hanna
- Department Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, United States of America
| |
Collapse
|
239
|
Podoplanin as an Attractive Target of CAR T Cell Therapy. Cells 2020; 9:cells9091971. [PMID: 32858947 PMCID: PMC7564405 DOI: 10.3390/cells9091971] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 12/19/2022] Open
Abstract
To date, various kinds of cancer immunotherapy methods have been developed, but T cell immunotherapy is one of the most promising strategies. In general, T cell receptor (TCR) or chimeric antigen receptor (CAR) is used to modify the antigen specificity of T cells. CARs possess an underlying potential with treatment efficacy to treat a broad range of cancer patients compared with TCRs. Although a variety of CAR molecules have been developed so far, the clinical application for solid tumors is limited partly due to its adverse effect known as “on-target off-tumor toxicity”. Therefore, it is very important for CAR T cell therapy to target specific antigens exclusively expressed by malignant cells. Here, we review the application of T cell immunotherapy using specific antigen receptor molecules and discuss the possibility of the clinical application of podoplanin-targeted CAR derived from a cancer-specific monoclonal antibody (CasMab).
Collapse
|
240
|
Zhang Z, Lu S, Dunmall LSC, Wang Z, Cheng Z, Zhang Z, Yan W, Chu Y, Gao D, Wang N, Li Y, Wang J, Li Y, Ji Y, Shan D, Li K, Wang P, Dong Y, Dong J, Lemoine NR, Pei D, Zhang L, Wang Y. Treatment and Prevention of Lung Cancer Using a Virus-Infected Reprogrammed Somatic Cell-Derived Tumor Cell Vaccination (VIReST) Regime. Front Immunol 2020; 11:1996. [PMID: 32903551 PMCID: PMC7438408 DOI: 10.3389/fimmu.2020.01996] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/23/2020] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is one of the most commonly diagnosed cancer and despite therapeutic advances, mortality remains high. The long period of clinical latency associated with lung cancer provides an ideal window of opportunity to administer vaccines to at-risk individuals that can prevent tumor progression and initiate long-term anti-tumor immune surveillance. Here we describe a personalized vaccination regime that could be applied for both therapeutic and prophylactic prevention of lung cancer, based on the derivation of lung cancer cells from induced pluripotent stem cells. Stem cells from healthy mice were modified to express Cre-dependent KRASG12D and Trp53R172H prior to differentiation to lung progenitor cells. Subsequent viral delivery of Cre caused activation of exogenous driver mutations, resulting in transformation and development of lung cancer cells. iPSC-derived lung cancer cells were highly antigenically related to lung cancer cells induced in LSL-KRASG12D/+; Trp53R172H/+ transgenic mice and were antigenically unrelated to original pluripotent stem cells or pancreatic cancer cells derived using the same technological platform. For vaccination, induced lung cancer cells were infected with oncolytic Adenovirus or Vaccinia virus, to act as vaccine adjuvants, prior to delivery of vaccines sequentially to a murine inducible transgenic model of lung cancer. Application of this Virus-Infected, Reprogrammed Somatic cell-derived Tumor cell (VIReST) regime primed tumor-specific T cell responses that significantly prolonged survival in both subcutaneous post-vaccine challenge models and induced transgenic models of lung cancer, demonstrating that stem cell-derived prophylactic vaccines may be a feasible intervention for treatment or prevention of lung cancer development in at-risk individuals.
Collapse
Affiliation(s)
- Zhe Zhang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shuangshuang Lu
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S. Chard Dunmall
- Centre for Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Zhizhong Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhenguo Cheng
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhongxian Zhang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wenli Yan
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yongchao Chu
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Dongling Gao
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yang Li
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiwei Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yuenan Li
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yupei Ji
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Danyang Shan
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Keke Li
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Panpan Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yunshu Dong
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jianzeng Dong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Nick R. Lemoine
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Duanqing Pei
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Lirong Zhang
- School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
241
|
Casey W, Massey SE, Mishra B. How Signaling Games Explain Mimicry at Many Levels: From Viral Epidemiology to Human Sociology. RESEARCH SQUARE 2020:rs.3.rs-51959. [PMID: 32793895 PMCID: PMC7418725 DOI: 10.21203/rs.3.rs-51959/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mimicry is exhibited in multiple scales, ranging from molecular, to organismal, and then to human society. 'Batesian' type mimicry entails a conflict of interest between sender and receiver, reflected in a deceptive mimic signal. 'Mullerian' type mimicry occurs when there is perfect common interest between sender and receiver, manifested by an honest co-mimic signal. Using a signaling games approach, simulations show that invasion by Batesian mimics will make Mullerian mimicry unstable, in a coevolutionary chase. We use these results to better understand the deceptive strategies of SARS-CoV-2 and their key role in the COVID-19 pandemic. At the biomolecular level, we explain how cellularization promotes Mullerian molecular mimicry, and discourages Batesian molecular mimicry. A wide range of processes analogous to cellularization are presented; these might represent a manner of reducing oscillatory instabilities. Lastly, we identify examples of mimicry in human society, that might be addressed using a signaling game approach.
Collapse
|
242
|
Such L, Zhao F, Liu D, Thier B, Le-Trilling VTK, Sucker A, Coch C, Pieper N, Howe S, Bhat H, Kalkavan H, Ritter C, Brinkhaus R, Ugurel S, Köster J, Seifert U, Dittmer U, Schuler M, Lang KS, Kufer TA, Hartmann G, Becker JC, Horn S, Ferrone S, Liu D, Van Allen EM, Schadendorf D, Griewank K, Trilling M, Paschen A. Targeting the innate immunoreceptor RIG-I overcomes melanoma-intrinsic resistance to T cell immunotherapy. J Clin Invest 2020; 130:4266-4281. [PMID: 32427578 PMCID: PMC7410049 DOI: 10.1172/jci131572] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 05/07/2020] [Indexed: 12/20/2022] Open
Abstract
Understanding tumor resistance to T cell immunotherapies is critical to improve patient outcomes. Our study revealed a role for transcriptional suppression of the tumor-intrinsic HLA class I (HLA-I) antigen processing and presentation machinery (APM) in therapy resistance. Low HLA-I APM mRNA levels in melanoma metastases before immune checkpoint blockade (ICB) correlated with nonresponsiveness to therapy and poor clinical outcome. Patient-derived melanoma cells with silenced HLA-I APM escaped recognition by autologous CD8+ T cells. However, targeted activation of the innate immunoreceptor RIG-I initiated de novo HLA-I APM transcription, thereby overcoming T cell resistance. Antigen presentation was restored in interferon-sensitive (IFN-sensitive) but also immunoedited IFN-resistant melanoma models through RIG-I-dependent stimulation of an IFN-independent salvage pathway involving IRF1 and IRF3. Likewise, enhanced HLA-I APM expression was detected in RIG-Ihi (DDX58hi) melanoma biopsies, correlating with improved patient survival. Induction of HLA-I APM by RIG-I synergized with antibodies blocking PD-1 and TIGIT inhibitory checkpoints in boosting the antitumor T cell activity of ICB nonresponders. Overall, the herein-identified IFN-independent effect of RIG-I on tumor antigen presentation and T cell recognition proposes innate immunoreceptor targeting as a strategy to overcome intrinsic T cell resistance of IFN-sensitive and IFN-resistant melanomas and improve clinical outcomes in immunotherapy.
Collapse
Affiliation(s)
- Lina Such
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Fang Zhao
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Derek Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Beatrice Thier
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | | | - Antje Sucker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Christoph Coch
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Natalia Pieper
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Sebastian Howe
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Halime Kalkavan
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- Institute of Immunology, and
- Department of Medical Oncology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Immunology, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Cathrin Ritter
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- Department of Translational Skin Cancer Research, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robin Brinkhaus
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Johannes Köster
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrike Seifert
- Friedrich Loeffler Institute for Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Martin Schuler
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- Department of Medical Oncology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Thomas A. Kufer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, Stuttgart, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Jürgen C. Becker
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- Department of Translational Skin Cancer Research, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Susanne Horn
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- Rudolf Schönheimer Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| |
Collapse
|
243
|
Cho SX, Vijayan S, Yoo JS, Watanabe T, Ouda R, An N, Kobayashi KS. MHC class I transactivator NLRC5 in host immunity, cancer and beyond. Immunology 2020; 162:252-261. [PMID: 32633419 DOI: 10.1111/imm.13235] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022] Open
Abstract
The presentation of antigenic peptides by major histocompatibility complex (MHC) class I and class II molecules is crucial for activation of the adaptive immune system. The nucleotide-binding domain and leucine-rich repeat receptor family members CIITA and NLRC5 function as the major transcriptional activators of MHC class II and class I gene expression, respectively. Since the identification of NLRC5 as the master regulator of MHC class I and class-I-related genes, there have been major advances in understanding the function of NLRC5 in infectious diseases and cancer. Here, we discuss the biological significance and mechanism of NLRC5-dependent MHC class I expression.
Collapse
Affiliation(s)
- Steven X Cho
- Department of Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Saptha Vijayan
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, TX, USA
| | - Ji-Seung Yoo
- Department of Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshiyuki Watanabe
- Department of Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ryota Ouda
- Department of Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ning An
- Department of Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Koichi S Kobayashi
- Department of Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan.,Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, TX, USA
| |
Collapse
|
244
|
Kang N, Eccleston M, Clermont PL, Latarani M, Male DK, Wang Y, Crea F. EZH2 inhibition: a promising strategy to prevent cancer immune editing. Epigenomics 2020; 12:1457-1476. [PMID: 32938196 PMCID: PMC7607396 DOI: 10.2217/epi-2020-0186] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapies are revolutionizing the clinical management of a wide range of cancers. However, intrinsic or acquired unresponsiveness to immunotherapies does occur due to the dynamic cancer immunoediting which ultimately leads to immune escape. The evolutionarily conserved histone modifier enhancer of zeste 2 (EZH2) is aberrantly overexpressed in a number of human cancers. Accumulating studies indicate that EZH2 is a main driver of cancer cells' immunoediting and mediate immune escape through downregulating immune recognition and activation, upregulating immune checkpoints and creating an immunosuppressive tumor microenvironment. In this review, we overviewed the roles of EZH2 in cancer immunoediting, the preclinical and clinical studies of current pharmacologic EZH2 inhibitors and the prospects for EZH2 inhibitor and immunotherapy combination for cancer treatment.
Collapse
Affiliation(s)
- Ning Kang
- Department of Experimental Therapeutics, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Mark Eccleston
- Belgian Volition SPRL, Parc Scientifique Créalys, Rue Phocas Lejeune 22, BE-5032 Isnes, Belgium
| | - Pier-Luc Clermont
- Faculty of Medicine, Université Laval, 1050, avenue de la Médecine, Québec, QC, G1V 0A6, Canada
| | - Maryam Latarani
- Cancer Research Group, School of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - David Kingsley Male
- Cancer Research Group, School of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
- Department of Urologic Sciences, The Vancouver Prostate Centre, The University of British Columbia, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Francesco Crea
- Cancer Research Group, School of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| |
Collapse
|
245
|
Rodríguez-Lobato LG, Ganzetti M, Fernández de Larrea C, Hudecek M, Einsele H, Danhof S. CAR T-Cells in Multiple Myeloma: State of the Art and Future Directions. Front Oncol 2020; 10:1243. [PMID: 32850376 PMCID: PMC7399644 DOI: 10.3389/fonc.2020.01243] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/16/2020] [Indexed: 01/24/2023] Open
Abstract
Despite recent therapeutic advances, the prognosis of multiple myeloma (MM) patients remains poor. Thus, new strategies to improve outcomes are imperative. Chimeric antigen receptor (CAR) T-cell therapy has changed the treatment landscape of B-cell malignancies, providing a potentially curative option for patients who are refractory to standard treatment. Long-term remissions achieved in patients with acute lymphoblastic leukemia and Non-Hodgkin Lymphoma encouraged its further development in MM. B-cell maturation antigen (BCMA)-targeted CAR T-cells have established outstanding results in heavily pre-treated patients. However, several other antigens such as SLAMF7 and CD44v6 are currently under investigation with promising results. Idecabtagene vicleucel is expected to be approved soon for clinical use. Unfortunately, relapses after CAR T-cell infusion have been reported. Hence, understanding the underlying mechanisms of resistance is essential to promote prevention strategies and to enhance CAR T-cell efficacy. In this review we provide an update of the most recent clinical and pre-clinical data and we elucidate both, the potential and the challenges of CAR T-cell therapy in the future.
Collapse
Affiliation(s)
- Luis Gerardo Rodríguez-Lobato
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maya Ganzetti
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carlos Fernández de Larrea
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Michael Hudecek
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Sophia Danhof
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
246
|
Obajdin J, Davies DM, Maher J. Engineering of chimeric natural killer cell receptors to develop precision adoptive immunotherapies for cancer. Clin Exp Immunol 2020; 202:11-27. [PMID: 32544282 PMCID: PMC7488126 DOI: 10.1111/cei.13478] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/04/2020] [Accepted: 06/07/2020] [Indexed: 12/15/2022] Open
Abstract
Natural killer (NK) cells are innate immune effectors which play a crucial role in recognizing and eliminating virally infected and cancerous cells. They effectively distinguish between healthy and distressed self through the integration of signals delivered by germline‐encoded activating and inhibitory cell surface receptors. The frequent up‐regulation of stress markers on genetically unstable cancer cells has prompted the development of novel immunotherapies that exploit such innate receptors. One prominent example entails the development of chimeric antigen receptors (CAR) that detect cell surface ligands bound by NK receptors, coupling this engagement to the delivery of tailored immune activating signals. Here, we review strategies to engineer CARs in which specificity is conferred by natural killer group 2D (NKG2D) or other NK receptor types. Multiple preclinical studies have demonstrated the remarkable ability of chimeric NK receptor‐targeted T cells and NK cells to effectively and specifically eliminate cancer cells and to reject established tumour burdens. Importantly, such systems act not only acutely but, in some cases, they also incite immunological memory. Moreover, CARs targeted with the NKG2D ligand binding domain have also been shown to disrupt the tumour microenvironment, through the targeting of suppressive T regulatory cells, myeloid‐derived suppressor cells and tumour vasculature. Collectively, these findings have led to the initiation of early‐phase clinical trials evaluating both autologous and allogeneic NKG2D‐targeted CAR T cells in the haematological and solid tumour settings.
Collapse
Affiliation(s)
- J Obajdin
- School of Cancer and Pharmaceutical Sciences, CAR Mechanics Laboratory, Guy's Cancer Centre, King's College London, London, UK
| | - D M Davies
- School of Cancer and Pharmaceutical Sciences, CAR Mechanics Laboratory, Guy's Cancer Centre, King's College London, London, UK
| | - J Maher
- School of Cancer and Pharmaceutical Sciences, CAR Mechanics Laboratory, Guy's Cancer Centre, King's College London, London, UK.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, UK.,Department of Immunology, Eastbourne Hospital, Eastbourne, UK.,Leucid Bio Ltd, Guy's Hospital, London, UK
| |
Collapse
|
247
|
Domankevich V, Efrati M, Schmidt M, Glikson E, Mansour F, Shai A, Cohen A, Zilberstein Y, Flaisher E, Galalae R, Kelson I, Keisari Y. RIG-1-Like Receptor Activation Synergizes With Intratumoral Alpha Radiation to Induce Pancreatic Tumor Rejection, Triple-Negative Breast Metastases Clearance, and Antitumor Immune Memory in Mice. Front Oncol 2020; 10:990. [PMID: 32766128 PMCID: PMC7379859 DOI: 10.3389/fonc.2020.00990] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/19/2020] [Indexed: 12/19/2022] Open
Abstract
Diffusing alpha-emitting radiation therapy (DaRT) employs intratumoral Ra-224-coated seeds that efficiently destroy solid tumors by slowly releasing alpha-emitting atoms inside the tumor. In immunogenic tumor models, DaRT was shown to activate systemic antitumor immunity. Agonists of the membrane-bound toll-like receptors (TLRs) enhanced these effects and led to tumor rejection. Here, we examined the combination of DaRT with agents that activate a different type of pattern recognition receptors, the cytoplasmatic RIG1-like receptors (RLRs). In response to cytoplasmatic viral dsRNA, RLRs activate an antiviral immune response that includes the elevation of antigen presentation. Thus, it was postulated that in low-immunogenic tumor models, RLR activation in tumor cells prior to the induction of their death by DaRT will be superior compared to TLR activation. Intratumoral cytoplasmatic delivery of the dsRNA mimic polyIC by polyethylenimine (PEI), was used to activate RLR, while polyIC without PEI was used to activate TLR. PolyIC(PEI) prior to DaRT synergistically retarded 4T1 triple-negative breast tumors and metastasis development more efficiently than polyIC and rejected panc02 pancreatic tumors in some of the treated mice. Splenocytes from treated mice, adoptively transferred to naive mice in combination with 4T1 tumor cells, delayed tumor development compared to naïve splenocytes. Low-dose cyclophosphamide, known to reduce T regulatory cell number, enhanced the effect of DaRT and polyIC(PEI) and led to high long-term survival rates under neoadjuvant settings, which confirmed metastasis clearance. The epigenetic drug decitabine, known to activate RLR in low doses, was given intraperitoneally prior to DaRT and caused tumor growth retardation, similar to local polyIC(PEI). The systemic and/or local administration of RLR activators was also tested in the squamous cell carcinoma (SCC) tumor model SQ2, in which a delay in tumor re-challenge development was demonstrated. We conclude that RIG-I-like activation prior to intratumoral alpha radiation may serve as a potent combination technique to reduce both tumor growth and the spread of distant metastases in low-immunogenic and metastatic tumor models.
Collapse
Affiliation(s)
- Vered Domankevich
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel.,Alpha Tau Medical, Tel Aviv-Yafo, Israel
| | - Margalit Efrati
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel.,Alpha Tau Medical, Tel Aviv-Yafo, Israel
| | - Michael Schmidt
- Alpha Tau Medical, Tel Aviv-Yafo, Israel.,Sackler Faculty of Exact Sciences, School of Physics and Astronomy, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Eran Glikson
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel.,Department of Otolaryngology, Head and Neck Surgery, Sheba Medical Center, Tel HaShomer, Israel
| | - Fairuz Mansour
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Amit Shai
- Alpha Tau Medical, Tel Aviv-Yafo, Israel
| | - Adi Cohen
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Yael Zilberstein
- Sackler Cellular and Molecular Imaging Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | | | - Razvan Galalae
- MedAustron, Wiener Neustadt, Austria.,Medical Faculty, Christian-Albrechts University, Kiel, Germany
| | - Itzhak Kelson
- Sackler Faculty of Exact Sciences, School of Physics and Astronomy, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Yona Keisari
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
248
|
Youness RA, Gad AZ, Sanber K, Ahn YJ, Lee GJ, Khallaf E, Hafez HM, Motaal AA, Ahmed N, Gad MZ. Targeting hydrogen sulphide signaling in breast cancer. J Adv Res 2020; 27:177-190. [PMID: 33318876 PMCID: PMC7728592 DOI: 10.1016/j.jare.2020.07.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/06/2020] [Accepted: 07/12/2020] [Indexed: 02/08/2023] Open
Abstract
Introduction Hydrogen sulphide (H2S) has been established as a key member of the gasotransmitters family that recently showed a pivotal role in various pathological conditions including cancer. Objectives This study investigated the role of H2S in breast cancer (BC) pathogenesis, on BC immune recognition capacity and the consequence of targeting H2S using non-coding RNAs. Methods Eighty BC patients have been recruited for the study. BC cell lines were cultured and transfected using validated oligonucleotide delivery system. Gene and protein expression analysis was performed using qRT-PCR, western blot and flow-cytometry. In-vitro analysis for BC hallmarks was performed using MTT, BrdU, Modified Boyden chamber, migration and colony forming assays. H2S and nitric oxide (NO) levels were measured spectrophotometrically. Primary natural killer cells (NK cells) and T cell isolation and chimeric antigen receptor transduction (CAR T cells) were performed using appropriate kits. NK and T cells cytotoxicity was measured. Finally, computational target prediction analysis and binding confirmation analyses were performed using different software and dual luciferase assay kit, respectively. Results The H2S synthesizing enzymes, cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE), exhibited elevated levels in the clinical samples that correlated with tumor proliferation index. Knock-down of CBS and CSE in the HER2+ BC and triple negative BC (TNBC) cells resulted in significant attenuation of BC malignancy. In addition to increased susceptibility of HER2+ BC and TNBC to the cytotoxic activity of HER2 targeting CAR T cells and NK cells, respectively. Transcriptomic and phosphoprotein analysis revealed that H2S signaling is mediated through Akt in MCF7, STAT3 in MDA-MB-231 and miR-155/ NOS2/NO signaling in both cell lines. Lastly, miR-4317 was found to function as an upstream regulator of CBS and CSE synergistically abrogates the malignancy of BC cells. Conclusion These findings demonstrate the potential role of H2S signaling in BC pathogenesis and the potential of its targeting for disease mitigation.
Collapse
Key Words
- 41BBL, 41BB Ligand
- 51Cr-release, Chromium release assay
- BC, Breast Cancer
- Breast cancer
- CAR T cells
- CAR, Chimeric antigen receptor
- CBS, Cystathionine β-synthase
- CD80, Cluster of differentiation 80
- CD86, Cluster of differentiation 86
- CSE, Cystathionine γ-lyase
- CTL, Cytotoxic T lymphocyte
- H2S, Hydrogen sulphide
- HCC, Hepatocellular carcinoma
- HLA-DR, Human Leukocytic antigen DR
- Hydrogen sulphide
- IFN-γ, Interferon gamma
- KD, Knock down
- LDH, Lactate dehydrogenase Assay
- MICA/B, MHC class I polypeptide-related sequence A/B
- NK, Natural killer
- NKG2D, Natural Killer Group 2D
- NO, Nitric oxide
- NOS2, Inducible nitric oxide synthase-2
- NOS3, Endothelial nitric oxide synthase-3
- Natural killer cells
- Nitric oxide
- PD-L1, Programmed death-ligand 1
- PI3K/AKT signaling pathway
- Scr-miRNAs, Scrambled microRNAs
- Scr-siRNAs, Scrambled siRNAs
- TNBC, Triple negative breast cancer
- TNF-α, Tumor necrosis factor-α
- ULBP2/5/6, UL16 binding protein 2/5/6
- miR-155/NOS2/NO signaling pathway
- miR-4317
- miRNA, MicroRNA
- ncRNAs, Non-coding RNAs
- siRNAs, Small interfering RNAs
Collapse
Affiliation(s)
- Rana Ahmed Youness
- Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Egypt
| | - Ahmed Zakaria Gad
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA.,Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Khaled Sanber
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA.,Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yong Jin Ahn
- Department of Medical Engineering, Graduate School, Kyung Hee University, Seoul 130-701, Republic of Korea.,Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Gi-Ja Lee
- Department of Medical Engineering, Graduate School, Kyung Hee University, Seoul 130-701, Republic of Korea.,Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Emad Khallaf
- Department of General Surgery, Faculty of Medicine, Cairo University, 12613 Cairo, Egypt
| | - Hafez Mohamed Hafez
- Department of General Surgery, Faculty of Medicine, Cairo University, 12613 Cairo, Egypt
| | - Amira Abdel Motaal
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.,Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Egypt
| | - Nabil Ahmed
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA.,Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mohamed Zakaria Gad
- Department of Biochemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Egypt
| |
Collapse
|
249
|
Tumor Cell-Intrinsic Immunometabolism and Precision Nutrition in Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12071757. [PMID: 32630618 PMCID: PMC7409312 DOI: 10.3390/cancers12071757] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
One of the greatest challenges in the cancer immunotherapy field is the need to biologically rationalize and broaden the clinical utility of immune checkpoint inhibitors (ICIs). The balance between metabolism and immune response has critical implications for overcoming the major weaknesses of ICIs, including their lack of universality and durability. The last decade has seen tremendous advances in understanding how the immune system's ability to kill tumor cells requires the conspicuous metabolic specialization of T-cells. We have learned that cancer cell-associated metabolic activities trigger shifts in the abundance of some metabolites with immunosuppressory roles in the tumor microenvironment. Yet very little is known about the tumor cell-intrinsic metabolic traits that control the immune checkpoint contexture in cancer cells. Likewise, we lack a comprehensive understanding of how systemic metabolic perturbations in response to dietary interventions can reprogram the immune checkpoint landscape of tumor cells. We here review state-of-the-art molecular- and functional-level interrogation approaches to uncover how cell-autonomous metabolic traits and diet-mediated changes in nutrient availability and utilization might delineate new cancer cell-intrinsic metabolic dependencies of tumor immunogenicity. We propose that clinical monitoring and in-depth molecular evaluation of the cancer cell-intrinsic metabolic traits involved in primary, adaptive, and acquired resistance to cancer immunotherapy can provide the basis for improvements in therapeutic responses to ICIs. Overall, these approaches might guide the use of metabolic therapeutics and dietary approaches as novel strategies to broaden the spectrum of cancer patients and indications that can be effectively treated with ICI-based cancer immunotherapy.
Collapse
|
250
|
Wang X, Waschke BC, Woolaver RA, Chen SMY, Chen Z, Wang JH. HDAC inhibitors overcome immunotherapy resistance in B-cell lymphoma. Protein Cell 2020; 11:472-482. [PMID: 32162275 PMCID: PMC7305292 DOI: 10.1007/s13238-020-00694-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has been applied successfully to treat B-cell lymphomas in preclinical models or clinical settings. However, immunotherapy resistance is a major challenge for B-cell lymphoma treatment. To overcome this issue, combinatorial therapeutic strategies have been pursued to achieve a better efficacy for treating B-cell lymphomas. One of such strategies is to combine immunotherapy with histone deacetylase (HDAC) inhibitors. HDAC inhibitors can potentially increase tumor immunogenicity, promote anti-tumor immune responses, or reverse immunosuppressive tumor environments. Thus, the combination of HDAC inhibitors and immunotherapy has drawn much attention in current cancer treatment. However, not all HDAC inhibitors are created equal and their net effects are highly dependent on the specific inhibitors used and the HDACs they target. Hence, we suggest that optimal treatment efficacy requires personalized design and rational combination based on prognostic biomarkers and unique profiles of HDAC inhibitors. Here, we discuss the possible mechanisms by which B-cell lymphomas acquire immunotherapy resistance and the effects of HDAC inhibitors on tumor cells and immune cells that could help overcome immunotherapy resistance.
Collapse
Affiliation(s)
- Xiaoguang Wang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Brittany C Waschke
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Rachel A Woolaver
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Samantha M Y Chen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Zhangguo Chen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Jing H Wang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA.
| |
Collapse
|