1
|
Sun F, Yan P, Xiao Y, Zhang H, Shapiro SD, Xiao G, Qu Z. Improving PD-1 blockade plus chemotherapy for complete remission of lung cancer by nanoPDLIM2. eLife 2024; 12:RP89638. [PMID: 39718207 DOI: 10.7554/elife.89638] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) and their combination with other therapies such as chemotherapy, fail in most cancer patients. We previously identified the PDZ-LIM domain-containing protein 2 (PDLIM2) as a bona fide tumor suppressor that is repressed in lung cancer to drive cancer and its chemo and immunotherapy resistance, suggesting a new target for lung cancer therapy improvement. In this study, human clinical samples and data were used to investigate PDLIM2 genetic and epigenetic changes in lung cancer. Using an endogenous mouse lung cancer model faithfully recapitulating refractory human lung cancer and a clinically feasible nano-delivery system, we investigated the therapeutic efficacy, action mechanism, and safety of systemically administrated PDLIM2 expression plasmids encapsulated in nanoparticles (nanoPDLIM2) and its combination with PD-1 antibody and chemotherapeutic drugs. Our analysis indicate that PDLIM2 repression in human lung cancer involves both genetic deletion and epigenetic alteration. NanoPDLIM2 showed low toxicity, high tumor specificity, antitumor activity, and greatly improved the efficacy of anti-PD-1 and chemotherapeutic drugs, with complete tumor remission in most mice and substantial tumor reduction in the remaining mice by their triple combination. Mechanistically, nanoPDLIM2 increased major histocompatibility complex class I (MHC-I) expression, suppressed multi-drug resistance 1 (MDR1) induction and survival genes and other tumor-related genes expression in tumor cells, and enhanced lymphocyte tumor infiltration, turning the cold tumors hot and sensitive to ICIs and rendering them vulnerable to chemotherapeutic drugs and activated tumor-infiltrating lymphocytes (TILs) including those unleashed by ICIs. These studies established a clinically applicable PDLIM2-based combination therapy with great efficacy for lung cancer and possibly other cold cancers.
Collapse
Affiliation(s)
- Fan Sun
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Pengrong Yan
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Yadong Xiao
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, United States
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, United States
- Norris Comprehensive Cancer Center, Hastings Center for Pulmonary Research, Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, United States
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, United States
| | - Hongqiao Zhang
- Norris Comprehensive Cancer Center, Hastings Center for Pulmonary Research, Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, United States
| | - Steven D Shapiro
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, United States
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, United States
| | - Gutian Xiao
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, United States
- Norris Comprehensive Cancer Center, Hastings Center for Pulmonary Research, Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, United States
| | - Zhaoxia Qu
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, United States
- Norris Comprehensive Cancer Center, Hastings Center for Pulmonary Research, Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, United States
| |
Collapse
|
2
|
Zhang J, Wang L, Guo H, Kong S, Li W, He Q, Ding L, Yang B. The role of Tim-3 blockade in the tumor immune microenvironment beyond T cells. Pharmacol Res 2024; 209:107458. [PMID: 39396768 DOI: 10.1016/j.phrs.2024.107458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/22/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024]
Abstract
Numerous preclinical studies have demonstrated the inhibitory function of T cell immunoglobulin mucin domain-containing protein 3 (Tim-3) on T cells as an inhibitory receptor, leading to the clinical development of anti-Tim-3 blocking antibodies. However, recent studies have shown that Tim-3 is expressed not only on T cells but also on multiple cell types in the tumor microenvironment (TME), including dendritic cells (DCs), natural killer (NK) cells, macrophages, and tumor cells. Therefore, Tim-3 blockade in the immune microenvironment not only affect the function of T cells but also influence the functions of other cells. For example, Tim-3 blockade can enhance the ability of DCs to regulate innate and adaptive immunity. The role of Tim-3 blockade in NK cells function is controversial, as it can enhance the antitumor function of NK cells under certain conditions while having the opposite effect in other situations. Additionally, Tim-3 blockade can promote the reversal of macrophage polarization from the M2 phenotype to the M1 phenotype. Furthermore, Tim-3 blockade can inhibit tumor development by suppressing the proliferation and metastasis of tumor cells. In summary, increasing evidence has shown that Tim-3 in other cell types also plays a critical role in the efficacy of anti-Tim-3 therapy. Understanding the function of anti-Tim-3 therapy in non-T cells can help elucidate the diverse responses observed in clinical patients, leading to better development of relevant therapeutic strategies. This review aims to discuss the role of Tim-3 in the TME and emphasize the impact of Tim-3 blockade in the tumor immune microenvironment beyond T cells.
Collapse
Affiliation(s)
- Jie Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Longsheng Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hongjie Guo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shijia Kong
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wen Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China; School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China.
| |
Collapse
|
3
|
Gang X, Yan J, Li X, Shi S, Xu L, Liu R, Cai L, Li H, Zhao M. Immune checkpoint inhibitors rechallenge in non-small cell lung cancer: Current evidence and future directions. Cancer Lett 2024; 604:217241. [PMID: 39260670 DOI: 10.1016/j.canlet.2024.217241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/23/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Immunotherapy, remarkably immune checkpoint inhibitors (ICIs), has significantly altered the treatment landscape for non-small cell lung cancer (NSCLC). Despite their success, the discontinuation of ICIs therapy may occur due to factors such as prior treatment completion, disease progression during ICIs treatment, or immune-related adverse events (irAEs). As numerous studies highlight the dynamic nature of immune responses and the sustained benefits of ICIs, ICIs rechallenge has become an attractive and feasible option. However, the decision-making process for ICIs rechallenge in clinical settings is complicated by numerous uncertainties. This review systematically analyses existing clinical research evidence, classifying ICIs rechallenge into distinct clinical scenarios, exploring methods to overcome ICIs resistance in rechallenge instances, and identifying biomarkers to select patients likely to benefit from rechallenge. By integrating recent studies and new technologies, we offer crucial recommendations for future clinical trial design and provide a practical guideline to maximize the therapeutic benefits of immunotherapy for NSCLC patients.
Collapse
Affiliation(s)
- Xiaoyu Gang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Jinshan Yan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xin Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Sha Shi
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Lu Xu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ruotong Liu
- Clinical Medicine, Shenyang Medical College, Shenyang, 110001, China
| | - Lutong Cai
- Psychological Medicine, Shenyang Medical College, Shenyang, 110001, China
| | - Heming Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China; Guangdong Association of Clinical Trials (GACT)/Chinese Thoracic Oncology Group (CTONG) and Guangdong Provincial Key Lab of Translational Medicine in Lung Cancer, Guangzhou, 510000, China.
| | - Mingfang Zhao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
4
|
Wang C, Wang L. Resistance mechanisms and potential therapeutic strategies in relapsed or refractory natural killer/T cell lymphoma. Chin Med J (Engl) 2024; 137:2308-2324. [PMID: 39175124 PMCID: PMC11441923 DOI: 10.1097/cm9.0000000000003152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Indexed: 08/24/2024] Open
Abstract
ABSTRACT Natural killer/T cell lymphoma (NKTCL) is a malignant tumor originating from NK or T cells, characterized by its highly aggressive and heterogeneous nature. NKTCL is predominantly associated with Epstein-Barr virus infection, disproportionately affecting Asian and Latin American populations. Owing to the application of asparaginase and immunotherapy, clinical outcomes have improved significantly. However, for patients in whom first-line treatment fails, the prognosis is exceedingly poor. Overexpression of multidrug resistance genes, abnormal signaling pathways, epigenetic modifications and active Epstein-Barr virus infection may be responsible for resistance. This review summarized the mechanisms of resistance for NKTCL and proposed potential therapeutic approaches.
Collapse
Affiliation(s)
- Chengji Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
5
|
Song X, Qu Z. NF-κB1 deficiency promotes macrophage-derived adrenal tumors but decreases neurofibromas in HTLV-I LTR-Tax transgenic mice. PLoS One 2024; 19:e0303138. [PMID: 38722890 PMCID: PMC11081228 DOI: 10.1371/journal.pone.0303138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
Human T-cell leukemia virus type I (HTLV-I) is an oncogenic virus whose infection can cause diverse diseases, most notably adult T-cell leukemia/lymphoma (ATL or ATLL), an aggressive and fatal malignancy of CD4 T cells. The oncogenic ability of HTLV-I is mostly attributed to the viral transcriptional transactivator Tax. Tax alone is sufficient to induce specific tumors in mice depending on the promotor used to drive Tax expression, thereby being used to understand HTLV-I tumorigenesis and model the tumor types developed in Tax transgenic mice. Tax exerts its oncogenic role predominantly by activating the cellular transcription factor NF-κB. Here, we report that genetic deletion of NF-κB1, the prototypic member of the NF-κB family, promotes adrenal medullary tumors but suppresses neurofibromas in mice with transgenic Tax driven by the HTLV-I Long Terminal Repeat (LTR) promoter. The adrenal tumors are derived from macrophages. Neoplastic macrophages also infiltrate the spleen and lymph nodes, causing splenomegaly and lymphadenopathy in mice. Nevertheless, the findings could be human relevant, because macrophages are important target cells of HTLV-I infection and serve as a virus reservoir in vivo. Moreover, the spleen, lymph nodes and adrenal glands are the most common sites of tumor cell infiltration in HTLV-I-infected patients. These data provide new mechanistic insights into the complex interaction between Tax and NF-κB, therefore improving our understanding of HTLV-I oncogenic pathogenesis. They also expand our knowledge and establish a new animal model of macrophage neoplasms and adrenal tumors.
Collapse
Affiliation(s)
- Xinxin Song
- Department of Microbiology and Molecular Genetics, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Zhaoxia Qu
- Department of Microbiology and Molecular Genetics, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
- Department of Molecular Microbiology and Immunology, Hastings Center for Pulmonary Research, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States of America
| |
Collapse
|
6
|
Sun F, Xiao Y, Shapiro SD, Qu Z, Xiao G. Critical and distinct roles of cell type-specific NF-κB2 in lung cancer. JCI Insight 2024; 9:e164188. [PMID: 38385745 DOI: 10.1172/jci.insight.164188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Different from the well-studied canonical NF-κB member RelA, the role of the noncanonical NF-κB member NF-κB2 in solid tumors, and lung cancer in particular, is poorly understood. Here we report that in contrast to the tumor-promoting role of RelA, NF-κB2 intrinsic to lung epithelial and tumor cells had no marked effect on lung tumorigenesis and progression. On the other hand, NF-κB2 limited dendritic cell number and activation in the lung but protected lung macrophages and drove them to promote lung cancer through controlling activation of noncanonical and canonical NF-κB, respectively. NF-κB2 was also required for B cell maintenance and T cell activation. The antitumor activity of lymphocyte NF-κB2 was dominated by the protumor function of myeloid NF-κB2; thus, NF-κB2 has an overall tumor-promoting activity. These studies reveal a cell type-dependent role for NF-κB2 in lung cancer and help understand the complexity of NF-κB action and lung cancer pathogenesis for better design of NF-κB-targeted therapy against this deadliest cancer.
Collapse
Affiliation(s)
- Fan Sun
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yadong Xiao
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Norris Comprehensive Cancer Center, Hastings Center for Pulmonary Research, Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Steven D Shapiro
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Zhaoxia Qu
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Norris Comprehensive Cancer Center, Hastings Center for Pulmonary Research, Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Gutian Xiao
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Norris Comprehensive Cancer Center, Hastings Center for Pulmonary Research, Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| |
Collapse
|
7
|
Sakhi H, Arabi M, Ghaemi A, Movafagh A, Sheikhpour M. Oncolytic viruses in lung cancer treatment: a review article. Immunotherapy 2024; 16:75-97. [PMID: 38112057 DOI: 10.2217/imt-2023-0124] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023] Open
Abstract
Lung cancer has a high morbidity rate worldwide due to its resistance to therapy. So new treatment options are needed to improve the outcomes of lung cancer treatment. This study aimed to evaluate the effectiveness of oncolytic viruses (OVs) as a new type of cancer treatment. In this study, 158 articles from PubMed and Scopus from 1994 to 2022 were reviewed on the effectiveness of OVs in the treatment of lung cancer. The oncolytic properties of eight categories of OVs and their interactions with treatment options were investigated. OVs can be applied as a promising immunotherapy option, as they are reproduced selectively in different types of cancer cells, cause tumor cell lysis and trigger efficient immune responses.
Collapse
Affiliation(s)
- Hanie Sakhi
- Department of Mycobacteriology & Pulmonary Research, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mohadeseh Arabi
- Department of Mycobacteriology & Pulmonary Research, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Amir Ghaemi
- Department of Virology, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Abolfazl Movafagh
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, 1983969411, Iran
| | - Mojgan Sheikhpour
- Department of Mycobacteriology & Pulmonary Research, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| |
Collapse
|
8
|
Bigos KJA, Quiles CG, Lunj S, Smith DJ, Krause M, Troost EGC, West CM, Hoskin P, Choudhury A. Tumour response to hypoxia: understanding the hypoxic tumour microenvironment to improve treatment outcome in solid tumours. Front Oncol 2024; 14:1331355. [PMID: 38352889 PMCID: PMC10861654 DOI: 10.3389/fonc.2024.1331355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Hypoxia is a common feature of solid tumours affecting their biology and response to therapy. One of the main transcription factors activated by hypoxia is hypoxia-inducible factor (HIF), which regulates the expression of genes involved in various aspects of tumourigenesis including proliferative capacity, angiogenesis, immune evasion, metabolic reprogramming, extracellular matrix (ECM) remodelling, and cell migration. This can negatively impact patient outcomes by inducing therapeutic resistance. The importance of hypoxia is clearly demonstrated by continued research into finding clinically relevant hypoxia biomarkers, and hypoxia-targeting therapies. One of the problems is the lack of clinically applicable methods of hypoxia detection, and lack of standardisation. Additionally, a lot of the methods of detecting hypoxia do not take into consideration the complexity of the hypoxic tumour microenvironment (TME). Therefore, this needs further elucidation as approximately 50% of solid tumours are hypoxic. The ECM is important component of the hypoxic TME, and is developed by both cancer associated fibroblasts (CAFs) and tumour cells. However, it is important to distinguish the different roles to develop both biomarkers and novel compounds. Fibronectin (FN), collagen (COL) and hyaluronic acid (HA) are important components of the ECM that create ECM fibres. These fibres are crosslinked by specific enzymes including lysyl oxidase (LOX) which regulates the stiffness of tumours and induces fibrosis. This is partially regulated by HIFs. The review highlights the importance of understanding the role of matrix stiffness in different solid tumours as current data shows contradictory results on the impact on therapeutic resistance. The review also indicates that further research is needed into identifying different CAF subtypes and their exact roles; with some showing pro-tumorigenic capacity and others having anti-tumorigenic roles. This has made it difficult to fully elucidate the role of CAFs within the TME. However, it is clear that this is an important area of research that requires unravelling as current strategies to target CAFs have resulted in worsened prognosis. The role of immune cells within the tumour microenvironment is also discussed as hypoxia has been associated with modulating immune cells to create an anti-tumorigenic environment. Which has led to the development of immunotherapies including PD-L1. These hypoxia-induced changes can confer resistance to conventional therapies, such as chemotherapy, radiotherapy, and immunotherapy. This review summarizes the current knowledge on the impact of hypoxia on the TME and its implications for therapy resistance. It also discusses the potential of hypoxia biomarkers as prognostic and predictive indictors of treatment response, as well as the challenges and opportunities of targeting hypoxia in clinical trials.
Collapse
Affiliation(s)
- Kamilla JA. Bigos
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Conrado G. Quiles
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Sapna Lunj
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Danielle J. Smith
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Mechthild Krause
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- School of Medicine, Technische Universitat Dresden, Dresden, Germany
| | - Esther GC. Troost
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- School of Medicine, Technische Universitat Dresden, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Institute of Radiooncology – OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Rossendorf, Germany
| | - Catharine M. West
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, United Kingdom
| | - Peter Hoskin
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Mount Vernon Cancer Centre, Northwood, United Kingdom
| | - Ananya Choudhury
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Christie Hospital NHS Foundation Trust, Manchester, Germany
| |
Collapse
|
9
|
Sun F, Yan P, Xiao Y, Zhang H, Shapiro SD, Xiao G, Qu Z. Improving PD-1 blockade plus chemotherapy for complete remission of lung cancer by nanoPDLIM2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.23.550248. [PMID: 37546791 PMCID: PMC10402062 DOI: 10.1101/2023.07.23.550248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Background Immune checkpoint inhibitors (ICIs) and their combination with other therapies such as chemotherapy, fail in most cancer patients. We previously identified the PDZ-LIM domain-containing protein 2 (PDLIM2) as a bona fide tumor suppressor that is repressed in lung cancer to drive cancer and its chemo and immunotherapy resistance, suggesting a new target for lung cancer therapy improvement. Methods Human clinical samples and data were used to investigate PDLIM2 genetic and epigenetic changes in lung cancer. Using an endogenous mouse lung cancer model faithfully recapitulating refractory human lung cancer and a clinically feasible nano-delivery system, we investigated the therapeutic efficacy, action mechanism, and safety of systemically administrated PDLIM2 expression plasmids encapsulated in nanoparticles (nanoPDLIM2) and its combination with PD-1 antibody and chemotherapeutic drugs. Results PDLIM2 repression in human lung cancer involves both genetic deletion and epigenetic alteration. NanoPDLIM2 showed low toxicity, high tumor specificity, antitumor activity, and greatly improved the efficacy of anti-PD-1 and chemotherapeutic drugs, with complete tumor remission in most mice and substantial tumor reduction in the remaining mice by their triple combination. Mechanistically, nanoPDLIM2 increased major histocompatibility complex class I (MHC-I) expression, suppressed multi-drug resistance 1 (MDR1) induction and survival genes and other tumor-related genes expression in tumor cells, and enhanced lymphocyte tumor infiltration, turning the cold tumors hot and sensitive to ICIs and rendering them vulnerable to chemotherapeutic drugs and activated tumor-infiltrating lymphocytes (TILs) including those unleashed by ICIs. Conclusions These studies established a clinically applicable PDLIM2-based combination therapy with great efficacy for lung cancer and possibly other cold cancers.
Collapse
|
10
|
Sun Y, Zhou X, Lucas E, Chen L, Zhang H, Chen H, Zhou F. Expression of B7-H3 and TIM-3 in gastric-type endocervical adenocarcinoma: prevalence, association with PD-L1 expression, and prognostic significance. J Pathol Clin Res 2024; 10:e345. [PMID: 37798754 PMCID: PMC10766062 DOI: 10.1002/cjp2.345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/23/2023] [Accepted: 09/03/2023] [Indexed: 10/07/2023]
Abstract
Gastric-type endocervical adenocarcinoma (GEA) is the second most common subtype of endocervical adenocarcinoma and has a poor prognosis. Anti-programmed death-1 and anti-programmed death-ligand 1 (PD-L1) inhibitors have emerged as a major treatment option for GEA; however, data on the expression of other immune checkpoints in GEA are limited. We analyzed the expression of T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and B7 homolog 3 protein (B7-H3) in 58 GEA and investigated their prognostic significance as well as association with PD-L1 expression and other known prognostic factors. Applying the tumor proportion score (TPS) with a cutoff of 1%, B7-H3 and TIM-3 were present in 48.3% and 17.2% of cases, respectively. Applying the combined positive score (CPS) with a cutoff of 1, TIM-3 expression was present in 70.7% of cases. Moreover, the expression of three checkpoints (B7-H3, TIM-3, and PD-L1) was incompletely overlapping. Patients with B7-H3 positive tumors (by TPS) or TIM-3 positive tumors (by TPS) had significantly worse recurrence-free survival (RFS) and overall survival (OS) (log-rank). Using CPS, patients with TIM-3 positive tumors showed significantly worse RFS (log-rank). Similarly, B7-H3 positivity (by TPS) and TIM-3 positivity (by TPS) were associated with worse RFS and OS in univariate analysis. TIM-3 positivity (by CPS) was associated with worse RFS in univariate analysis and the final Cox multivariate analysis. In conclusion, our results show that (1) B7-H3 and TIM-3 are frequently expressed in GEA and their expression overlaps incompletely with PD-L1; and (2) both B7-H3 and TIM-3 are independent negative prognostic markers in GEA.
Collapse
Affiliation(s)
- Yao Sun
- Department of PathologyZhejiang University School of Medicine Women's HospitalHangzhouZhejiang ProvincePR China
| | - Xin Zhou
- Department of PathologyZhejiang University School of Medicine Women's HospitalHangzhouZhejiang ProvincePR China
- Department of PathologyInternational Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Elena Lucas
- Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of PathologyParkland HospitalDallasTXUSA
| | - Lili Chen
- Department of GynecologyZhejiang University School of Medicine Women's HospitalHangzhouZhejiang ProvincePR China
| | - Huijuan Zhang
- Department of PathologyInternational Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Hao Chen
- Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of PathologyParkland HospitalDallasTXUSA
| | - Feng Zhou
- Department of PathologyZhejiang University School of Medicine Women's HospitalHangzhouZhejiang ProvincePR China
- Department of PathologyInternational Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPR China
| |
Collapse
|
11
|
Zhang L, Pakmehr SA, Shahhosseini R, Hariri M, Fakhrioliaei A, Karkon Shayan F, Xiang W, Karkon Shayan S. Oncolytic viruses improve cancer immunotherapy by reprogramming solid tumor microenvironment. Med Oncol 2023; 41:8. [PMID: 38062315 DOI: 10.1007/s12032-023-02233-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/21/2023] [Indexed: 12/18/2023]
Abstract
Immunotherapies using immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T-cell therapy have achieved successful results against several types of human tumors, particularly hematological malignancies. However, their clinical results for the treatment of solid tumors remain poor and unsatisfactory. The immunosuppressive tumor microenvironment (TME) plays an important role by interfering with intratumoral T-cell infiltration, promoting effector T-cell exhaustion, upregulating inhibitory molecules, inducing hypoxia, and so on. Oncolytic viruses are an encouraging biocarrier that could be used in both natural and genetically engineered platforms to induce oncolysis in a targeted manner. Oncolytic virotherapy (OV) contributes to the reprogramming of the TME, thus synergizing the functional effects of current ICIs and CAR T-cell therapy to overcome resistant barriers in solid tumors. Here, we summarize the TME-related inhibitory factors affecting the therapeutic outcomes of ICIs and CAR T cells and discuss the potential of OV-based approaches to alleviate these barriers and improve future therapies for advanced solid tumors.
Collapse
Affiliation(s)
- Ling Zhang
- The Second People's Hospital of Lianyungang, Jiangsu, 222000, China
| | | | | | - Maryam Hariri
- Department of Pathobiology, Auburn University, Auburn, AL, 36832, USA
| | | | - Farid Karkon Shayan
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Wenxue Xiang
- The Second People's Hospital of Lianyungang, Jiangsu, 222000, China.
| | - Sepideh Karkon Shayan
- Student Research Committee, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
- Clinical Research Development Unit, Bohlool Hospital, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
12
|
He X, Peng Y, He G, Ye H, Liu L, Zhou Q, Shi J, Fu S, Wang J, Zhou Z, Li W. Increased co-expression of PD1 and TIM3 is associated with poor prognosis and immune microenvironment heterogeneity in gallbladder cancer. J Transl Med 2023; 21:717. [PMID: 37828574 PMCID: PMC10571407 DOI: 10.1186/s12967-023-04589-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND The effectiveness of immune checkpoint inhibitors in treating gallbladder cancer (GBC) remains unsatisfactory. Recently, several new immune checkpoints have been identified. However, investigations exploring these immune checkpoints in GBC are limited. In this study, we aim to investigate the expression patterns and clinical implications of various immune checkpoints, and further characterize the spatial and quantitative heterogeneity of immune components in GBC. METHODS We employed single and multiplex immunohistochemistry to evaluate the expression of five immune checkpoint markers and four immune cell markers in the primary tumor core, hepatic invasion margin, and liver metastasis. Subsequently, we analyzed their interrelationships and their prognostic significance. RESULTS We observed a robust positive correlation between PD1/TIM3 expression in GBC (R = 0.614, P < 0.001). The co-expression of PD1/TIM3 exhibited a synergistic effect in predicting poor prognosis among postoperative GBC patients. Further analysis revealed that the prognostic significance of PD1/TIM3 was prominent in the subgroup with high infiltration of CD8 + T cells (P < 0.001). Multiplex immunohistochemistry reveals that PD1 + TIM3 + FOXP3 + cells constitute a significant proportion of FOXP3 + TILs in GBC tissue. Moreover, the co-high expression of PD1 and TIM3 is positively correlated with the accumulation of CD8 + TILs at the hepatic invasion margin. Lastly, our findings indicated reduced expression levels of immune checkpoints and diminished immune cell infiltration in liver metastases compared to primary tumors. CONCLUSIONS Increased co-expression of PD1/TIM3 is associated with poor prognosis in GBC patients and is related to the heterogeneity of immune microenvironment between GBC primary tumor and its hepatic invasion margin or liver metastases, which may be a potential target for future immunotherapy of GBC.
Collapse
Affiliation(s)
- Xing He
- Department of Biliary and Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Yaorong Peng
- Department of Biliary and Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Gui He
- Cellular & Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Huilin Ye
- Department of Biliary and Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Liqiang Liu
- Department of Biliary and Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Qixian Zhou
- Department of Biliary and Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Juanyi Shi
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Sha Fu
- Cellular & Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Jie Wang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Zhenyu Zhou
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
| | - Wenbin Li
- Department of Biliary and Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
13
|
Wang L, Geng H, Liu Y, Liu L, Chen Y, Wu F, Liu Z, Ling S, Wang Y, Zhou L. Hot and cold tumors: Immunological features and the therapeutic strategies. MedComm (Beijing) 2023; 4:e343. [PMID: 37638340 PMCID: PMC10458686 DOI: 10.1002/mco2.343] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
The "hotness" or "coldness" of the tumors are determined by the information of the cancer cells themselves, tumor immune characteristics, tumor microenvironment, and signaling mechanisms, which are key factors affecting cancer patients' clinical efficacy. The switch mechanism of "hotness" and "coldness" and its corresponding pathological characteristics and treatment strategies are the frontier and hot spot of tumor treatment. How to distinguish the "hotness" or "coldness" effectively and clarify the causes, microenvironment state, and characteristics are very important for the tumor response and efficacy treatments. Starting from the concept of hot and cold tumor, this review systematically summarized the molecular characteristics, influencing factors, and therapeutic strategies of "hot and cold tumors," and analyzed the immunophenotypes, the tumor microenvironment, the signaling pathways, and the molecular markers that contribute to "hot and cold tumors" in details. Different therapeutic strategies for "cold and hot tumors" based on clinical efficacy were analyzed with drug targets and proteins for "cold and hot tumors." Furthermore, this review combines the therapeutic strategies of different "hot and cold tumors" with traditional medicine and modern medicine, to provide a basis and guidance for clinical decision-making of cancer treatment.
Collapse
Affiliation(s)
- Lianjie Wang
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hui Geng
- Department of Internal MedicineShanghai International Medical CenterShanghaiChina
| | - Yujie Liu
- Department of NephrologyShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Lei Liu
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yanhua Chen
- Department of the Tumor Research Center, Academy of Integrative MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Fanchen Wu
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhiyi Liu
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Shiliang Ling
- Department of Medical OncologyNingbo Hospital of Traditional Chinese Medicine, Zhejiang ProvinceNingboChina
| | - Yan Wang
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Lihong Zhou
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
14
|
Wu R, Tong S, Yin J, Zhu Z, Mao Z, Xu L. Oncolytic vaccinia virus acts synergistically with anti-PD-L1 antibody to enhance the killing of colon cancer cells by CD8 + T cells. Pathol Res Pract 2023; 247:154535. [PMID: 37257241 DOI: 10.1016/j.prp.2023.154535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 04/11/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023]
Abstract
Both oncolytic vaccinia virus (OVV) and anti-PD-L1 antibody hold promise in cancer immunotherapy. Herein, we aimed to explore the possible synergistic effects of OVV and anti-PD-L1 on the growth and metastasis of colon cancer (CC) in mouse models. Microarray profiling of CC-related genes was first conducted. Expression of PD-L1 in CC tissues was predicted by TCGA and verified by flow cytometry and RT-qPCR. Then, mouse CC cell lines stably carrying luciferase MC38-luc and CT26-luc were infected with recombinant double-deleted vaccinia virus (vvDD) to evaluate the effect of vvDD on cell viability. The data indicated that PD-L1 was highly expressed in CC tissues and cells following vvDD infection. MC38-luc cells were inoculated into mice to construct CC-bearing mouse models, which were treated with vvDD or combined with anti-PD-L1, with tumor growth, metastasis, survival, and the immune environment analyzed. It was found that OVV combined with anti-PD-L1 antibody led to lower tumor burden and growth and higher survival rates than individual treatment in CC-bearing mice. In addition, this combination exerted a remote effect on the untreated subcutaneous tumors in the lateral abdomen, thus suppressing the tumor metastasis. Furthermore, combined therapy of OVV with anti-PD-L1 antibody activated CD8+ T cells, reduced exhaustion of CD8+ T cells, and enhanced their immune response, strengthening the killing of CC cells and inhibiting tumor growth and metastasis. In conclusion, our findings provide mechanistic insights into the action and efficacy of OVV as an immunomodulatory agent combined with the anti-PD-L1 antibody for the treatment of CC.
Collapse
Affiliation(s)
- Runda Wu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, PR China
| | - Shan Tong
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, PR China
| | - Jun Yin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, PR China
| | - Zheng Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, PR China
| | - Zhongqi Mao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, PR China.
| | - Lu Xu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, PR China.
| |
Collapse
|
15
|
Lin D, Shen Y, Liang T. Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther 2023; 8:156. [PMID: 37041165 PMCID: PMC10090134 DOI: 10.1038/s41392-023-01407-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 04/13/2023] Open
Abstract
Oncolytic viruses (OVs) have attracted growing awareness in the twenty-first century, as they are generally considered to have direct oncolysis and cancer immune effects. With the progress in genetic engineering technology, OVs have been adopted as versatile platforms for developing novel antitumor strategies, used alone or in combination with other therapies. Recent studies have yielded eye-catching results that delineate the promising clinical outcomes that OVs would bring about in the future. In this review, we summarized the basic principles of OVs in terms of their classifications, as well as the recent advances in OV-modification strategies based on their characteristics, biofunctions, and cancer hallmarks. Candidate OVs are expected to be designed as "qualified soldiers" first by improving target fidelity and safety, and then equipped with "cold weapons" for a proper cytocidal effect, "hot weapons" capable of activating cancer immunotherapy, or "auxiliary weapons" by harnessing tactics such as anti-angiogenesis, reversed metabolic reprogramming and decomposing extracellular matrix around tumors. Combinations with other cancer therapeutic agents have also been elaborated to show encouraging antitumor effects. Robust results from clinical trials using OV as a treatment congruously suggested its significance in future application directions and challenges in developing OVs as novel weapons for tactical decisions in cancer treatment.
Collapse
Affiliation(s)
- Danni Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yinan Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
16
|
Wang T, Zhang H, Han Y, Zheng Q, Liu H, Han M, Li Z. Reversing T Cell Dysfunction to Boost Glioblastoma Immunotherapy by Paroxetine-Mediated GRK2 Inhibition and Blockade of Multiple Checkpoints through Biomimetic Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204961. [PMID: 36698265 PMCID: PMC10037995 DOI: 10.1002/advs.202204961] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/02/2022] [Indexed: 05/19/2023]
Abstract
T cell dysfunction-induced tumor immune escape is particularly severe in glioblastoma (GBM), and significantly affects the efficacy of immunotherapy. It is crucial to innovatively reverse the T cell dysfunction for improving GBM immunotherapy. Herein, T cell dysfunction is remarkably reversed and immunotherapy of GBM is boosted by repurposing the U. S. Food and Drug Administration-approved antidepressant paroxetine (PX) with biomimetic nanoparticles (CS-J@CM/6 NPs). The PX is successfully applied to abrogate T cell sequestration in the bone marrow of GBM-bearing mice and increase their infiltration in tumor. The biomimetic NPs are composed of ultrasmall Cu2- x Se NPs, JQ1, and tumor cell membrane modified with CD6, and are efficiently delivered into tumor through the specific interactions between CD6 and activated leukocyte cell adhesion molecule. They ameliorate the T cell dysfunction through the double roles of loaded JQ1, which simultaneously decreases the expression of PD-1 and TIM-3 on T cells, and the expression of PD-L1 on tumor cells. The NP also induces the immunogenic cell death of tumor cells to activate immune response. The synergistic roles of PX and biomimetic CS-J@CM/6 NPs notably enhance the survival of GBM-bearing mice. This work provides new insights into tumor immunotherapy by repurposing "old drugs" with advanced NPs.
Collapse
Affiliation(s)
- Tingting Wang
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Hao Zhang
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Qing Zheng
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Hanghang Liu
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Mengxiao Han
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| |
Collapse
|
17
|
Zhou K, Li S, Zhao Y, Cheng K. Mechanisms of drug resistance to immune checkpoint inhibitors in non-small cell lung cancer. Front Immunol 2023; 14:1127071. [PMID: 36845142 PMCID: PMC9944349 DOI: 10.3389/fimmu.2023.1127071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) in the form of anti-CTLA-4 and anti-PD-1/PD-L1 have become the frontier of cancer treatment and successfully prolonged the survival of patients with advanced non-small cell lung cancer (NSCLC). But the efficacy varies among different patient population, and many patients succumb to disease progression after an initial response to ICIs. Current research highlights the heterogeneity of resistance mechanisms and the critical role of tumor microenvironment (TME) in ICIs resistance. In this review, we discussed the mechanisms of ICIs resistance in NSCLC, and proposed strategies to overcome resistance.
Collapse
Affiliation(s)
- Kexun Zhou
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
- Abdominal Oncology Ward, Division of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shuo Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center, West China Hospital Sichuan University, Chengdu, China
| | - Yi Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Ke Cheng
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
- Abdominal Oncology Ward, Division of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Gowan CC, Bartee MY, Flores E, Aksoy BA, Templeton C, Baillie K, Happe M, Bartee E. The Combination of TIM3-Based Checkpoint Blockade and Oncolytic Virotherapy Regresses Established Solid Tumors. J Immunother 2023; 46:1-4. [PMID: 36472581 PMCID: PMC9783015 DOI: 10.1097/cji.0000000000000444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/08/2022] [Indexed: 12/12/2022]
Abstract
T-cell immunoglobulin and mucin domain 3 (TIM3) is emerging as a potential target for antibody-based checkpoint blockade. However, the efficacy of TIM3 blockade in combination with other treatment modalities, has not been extensively studied. In the current work we combined TIM3 blockade with myxoma virus-based oncolytic virotherapy (OV). Our results demonstrate that myxoma virus's ability to initiate an immense antitumor immune response complements the ability of TIM3 blockade to shift the tumor microenvironment to a more proinflammatory state. As a result, the combination of TIM3 blockade and OV is able to completely eradicate established disease, while neither monotherapy is effective. These data represent the first demonstration that OV can enhance the efficacy of TIM3 blockade and suggest that this treatment may need to be incorporated into more aggressive, combinatorial regimens in order to fulfill its potential as an immunotherapeutic.
Collapse
Affiliation(s)
- Cody C Gowan
- Division of Nephrology and Hypertension, Mayo Clinical, Jacksonville, FL
| | - Mee Y Bartee
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Erica Flores
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM
| | | | - Conor Templeton
- Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC
| | | | | | - Eric Bartee
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM
| |
Collapse
|
19
|
Wen Y, Wang X, Meng W, Guo W, Duan C, Cao J, Kang L, Guo N, Lin Q, Lv P, Zhang R, Xing L, Zhang X, Shen H. TNF-α-dependent lung inflammation upregulates PD-L1 in monocyte-derived macrophages to contribute to lung tumorigenesis. FASEB J 2022; 36:e22595. [PMID: 36205325 DOI: 10.1096/fj.202200434rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/20/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022]
Abstract
Chronic inflammation, which is dominated by macrophage-involved inflammatory responses, is an instigator of cancer initiation. Macrophages are the most abundant immune cells in healthy lungs, and associated with lung tumor development and promotion. PD-L1 is a negative molecule in macrophages and correlated with an immunosuppressive function in tumor environment. Macrophages expressing PD-L1, rather than tumor cells, exhibits a critical role in tumor growth and progression. However, whether and how PD-L1 in macrophages contributes to inflammation-induced lung tumorigenesis requires further elucidation. Here, we found that higher expression of PD-L1 in CD11b+ CD206+ macrophages was positively correlated with tumor progression and PD-1+ CD8+ T cells population in human adenocarcinoma patients. In the urethane-induced inflammation-driven lung adenocarcinoma (IDLA) mouse model, the infiltration of circulating CD11bhigh F4/80+ monocyte-derived macrophages (MoMs) was increased in pro-tumor inflamed lung tissues and lung adenocarcinoma. PD-L1 was mainly upregulated in MoMs associated with enhanced T cells exhaustion in lung tissues. Anti-PD-L1 treatment can reduce T cells exhaustion at pro-tumor inflammatory stage, and then inhibit tumorigenesis in IDLA. The pro-tumor lung inflammation depended on TNF-α to upregulate PD-L1 and CSN6 expression in MoMs, and induced cytokines production by alveolar type-II cells (AT-II). Furthermore, inflammatory AT-II cells could secret TNF-α to upregulate PD-L1 expression in bone-marrow driven macrophages (BM-M0). Inhibition of CSN6 decreased PD-L1 expression in TNF-α-activated macrophage in vitro, suggesting a critical role of CSN6 in PD-L1 upregulation. Thus, pro-tumor inflammation can depend on TNF-α to upregulate PD-L1 in recruited MoMs, which may be essential for lung tumorigenesis.
Collapse
Affiliation(s)
- Yue Wen
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Department of Ultrasound, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiuqing Wang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Wei Meng
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Wenli Guo
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Chenyang Duan
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Jingjing Cao
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Lifei Kang
- Department of Pathology, Hebei Chest Hospital, Shijiazhuang, China
| | - Ningfei Guo
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Qiang Lin
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, Renqiu, China
| | - Ping Lv
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Lingxiao Xing
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Xianghong Zhang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China.,Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
Optimal timing of PD-1 blockade in combination with oncolytic virus therapy. Semin Cancer Biol 2022; 86:971-980. [PMID: 34033895 DOI: 10.1016/j.semcancer.2021.05.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 01/27/2023]
Abstract
Anti-PD-1 and oncolytic viruses (OVs) have non-overlapping anti-tumor mechanisms, since each agent works at different steps of the cancer-immunity cycle. Evidence suggests that OVs improve therapeutic responses to anti-PD-1 therapy by reversing immunosuppressive factors, increasing the number and diversity of infiltrating lymphocytes, and promoting PD-L1 expression in both injected and non-injected tumors. Many studies in preclinical models suggest that the timing of anti-PD-1 administration influences the therapeutic success of the combination therapy (anti-PD-1 + OV). Therefore, determining the appropriate sequencing of agents is of critical importance to designing a rationale OV-based combinational clinical trial. Currently, the combination of anti-PD-1 and OVs are being delivered using various schedules, and we have classified the timing of administration of anti-PD-1 and OVs into five categories: (i) anti-PD-1 lead-in → OV; (ii) concurrent administration; (iii) OV lead-in → anti-PD-1; (iv) concurrent therapy lead-in → anti-PD-1; and (v) OV lead-in → concurrent therapy. Based on the reported preclinical and clinical literature, the most promising treatment strategy to date is hypothesized to be OV lead-in → concurrent therapy. In the OV lead-in → concurrent therapy approach, initial OV treatment results in T cell priming and infiltration into tumors and an immunologically hot tumor microenvironment (TME), which can be counterbalanced by engagement of PD-L1 to PD-1 receptor on immune cells, leading to T cell exhaustion. Therefore, after initial OV therapy, concurrent use of both OV and anti-PD-1 is critical through which OV maintains T cell priming and an immunologically hot TME, whereas PD-1 blockade helps to overcome PD-L1/PD-1-mediated T cell exhaustion. It is important to note that the hypothetical conclusion drawn in this review is based on thorough literature review on current understanding of OV + anti-PD-1 combination therapies and rhythm of treatment-induced cancer-immunity cycle. A variety of confounding factors such as tumor types, OV types, presence or absence of cytokine transgenes carried by an OV, timing of treatment initiation, varying dosages and treatment frequencies/duration of OV and anti-PD-1, etc. may affect the validity of our conclusion that will need to be further examined by future research (such as side-by-side comparative studies using all five treatment schedules in a given tumor model).
Collapse
|
21
|
Li Z, Feiyue Z, Gaofeng L, Haifeng L. Lung cancer and oncolytic virotherapy--enemy's enemy. Transl Oncol 2022; 27:101563. [PMID: 36244134 PMCID: PMC9561464 DOI: 10.1016/j.tranon.2022.101563] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/27/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022] Open
Abstract
Lung cancer is one of the malignant tumors that seriously threaten human health worldwide, while the covid-19 virus has become people's nightmare after the coronavirus pandemic. There are too many similarities between cancer cells and viruses, one of the most significant is that both of them are our enemies. The strategy to take the advantage of the virus to beat cancer cells is called Oncolytic virotherapy. When immunotherapy represented by immune checkpoint inhibitors has made remarkable breakthroughs in the clinical practice of lung cancer, the induction of antitumor immunity from immune cells gradually becomes a rapidly developing and promising strategy of cancer therapy. Oncolytic virotherapy is based on the same mechanisms that selectively kill tumor cells and induce systemic anti-tumor immunity, but still has a long way to go before it becomes a standard treatment for lung cancer. This article provides a comprehensive review of the latest progress in oncolytic virotherapy for lung cancer, including the specific mechanism of oncolytic virus therapy and the main types of oncolytic viruses, and the combination of oncolytic virotherapy and existing standard treatments. It aims to provide new insights and ideas on oncolytic virotherapy for lung cancer.
Collapse
Affiliation(s)
- Zhang Li
- Department of Oncology, Gejiu People's Hospital, The Fifth Affiliated Hospital of Kunming Medical University, China
| | - Zhang Feiyue
- Department of Oncology, Yuxi People's Hospital, The Sixth Affiliated Hospital of Kunming Medical University, China
| | - Li Gaofeng
- Department of Thoracic Surgery, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, China
| | - Liang Haifeng
- Department of Oncology, Gejiu People's Hospital, The Fifth Affiliated Hospital of Kunming Medical University, China,Corresponding author.
| |
Collapse
|
22
|
Repáraz D, Ruiz M, Silva L, Aparicio B, Egea J, Guruceaga E, Ajona D, Senent Y, Conde E, Navarro F, Barace S, Alignani D, Hervás-Stubbs S, Lasarte JJ, Llopiz D, Sarobe P. Gemcitabine-mediated depletion of immunosuppressive dendritic cells enhances the efficacy of therapeutic vaccination. Front Immunol 2022; 13:991311. [PMID: 36300124 PMCID: PMC9589451 DOI: 10.3389/fimmu.2022.991311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Vaccination using optimized strategies may increase response rates to immune checkpoint inhibitors (ICI) in some tumors. To enhance vaccine potency and improve thus responses to ICI, we analyzed the gene expression profile of an immunosuppressive dendritic cell (DC) population induced during vaccination, with the goal of identifying druggable inhibitory mechanisms. RNAseq studies revealed targetable genes, but their inhibition did not result in improved vaccines. However, we proved that immunosuppressive DC had a monocytic origin. Thus, monocyte depletion by gemcitabine administration reduced the generation of these DC and increased vaccine-induced immunity, which rejected about 20% of LLC-OVA and B16-OVA tumors, which are non-responders to anti-PD-1. This improved efficacy was associated with higher tumor T-cell infiltration and overexpression of PD-1/PD-L1. Therefore, the combination of vaccine + gemcitabine with anti-PD-1 was superior to anti-PD-1 monotherapy in both models. B16-OVA tumors benefited from a synergistic effect, reaching 75% of tumor rejection, but higher levels of exhausted T-cells in LLC-OVA tumors co-expressing PD-1, LAG3 and TIM3 precluded similar levels of efficacy. Our results indicate that gemcitabine is a suitable combination therapy with vaccines aimed at enhancing PD-1 therapies by targeting vaccine-induced immunosuppressive DC.
Collapse
Affiliation(s)
- David Repáraz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
| | - Marta Ruiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
| | - Leyre Silva
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
| | - Belén Aparicio
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
| | - Josune Egea
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
| | - Elizabeth Guruceaga
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Daniel Ajona
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Yaiza Senent
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Enrique Conde
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Flor Navarro
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Sergio Barace
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Diego Alignani
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Sandra Hervás-Stubbs
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
| | - Juan José Lasarte
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Diana Llopiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
| | - Pablo Sarobe
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- *Correspondence: Pablo Sarobe,
| |
Collapse
|
23
|
Lak S, Janelle V, Djedid A, Boudreau G, Brasey A, Lisi V, Smaani A, Carli C, Busque L, Lavallée VP, Delisle JS. Combined PD-L1 and TIM3 blockade improves expansion of fit human CD8 + antigen-specific T cells for adoptive immunotherapy. Mol Ther Methods Clin Dev 2022; 27:230-245. [PMID: 36320412 PMCID: PMC9593254 DOI: 10.1016/j.omtm.2022.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 09/29/2022] [Indexed: 11/27/2022]
Abstract
Antigen-specific T cell expansion ex vivo followed by adoptive transfer enables targeting of a multitude of microbial and cancer antigens. However, clinical-scale T cell expansion from rare precursors requires repeated stimulation, which may lead to T cell dysfunction and limited therapeutic potential. We used a clinically compliant protocol to expand Epstein-Barr virus (EBV) and Wilms tumor 1 (WT1) antigen-specific CD8+ T cells, and leveraged T cell exhaustion-associated inhibitory receptor blockade to improve T cell expansion. Several inhibitory receptors were expressed early by ex vivo-expanded antigen-specific CD8+ T cells, including PD-1 and TIM3, with co-expression matching evidence of T cell dysfunction as the cultures progressed. Introduction of anti-PD-L1 and anti-TIM3 blockade in combination (but not individually) to the culture led to markedly improved antigen-specific T cell expansion without inducing T cell dysfunction. Single-cell RNA sequencing (RNA-seq) and T cell receptor (TCR) repertoire profiling revealed that double blockade does not impart specific transcriptional programs in T cells or alterations in TCR repertoires. However, combined blockade may affect gene expression in a minority of clonotypes in a donor-specific fashion. We conclude that antigen-specific CD8+ T cell manufacturing can be improved by using TIM3 and PD-L1/PD-1 axis blockade in combination. This approach is readily applicable to several adoptive immunotherapy strategies.
Collapse
Affiliation(s)
- Shirin Lak
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Valérie Janelle
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Anissa Djedid
- Centre de Recherche Du CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Gabrielle Boudreau
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Ann Brasey
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Biomarker Unit, Centre C3i, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Véronique Lisi
- Centre de Recherche Du CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Ali Smaani
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Cédric Carli
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada
| | - Lambert Busque
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Biomarker Unit, Centre C3i, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Department of Medicine, Université de Montréal, CP 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada,Hematology-Oncology and Cell Therapy Division, Hôpital Maisonneuve-Rosemont, Montréal, QC Canada
| | - Vincent-Philippe Lavallée
- Centre de Recherche Du CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada,Department of Pediatrics, Université de Montréal, Montréal, QC, Canada,Hematology-Oncology Division, CHU Sainte-Justine, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Jean-Sébastien Delisle
- Centre de Recherche de L’Hôpital Maisonneuve-Rosemont, 5415 Boul. de L’Assomption, Montréal, QC H1T 2M4, Canada,Department of Medicine, Université de Montréal, CP 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada,Hematology-Oncology and Cell Therapy Division, Hôpital Maisonneuve-Rosemont, Montréal, QC Canada,Corresponding author Jean-Sébastien Delisle, MD, FRCPC, PhD, Centre de recherche de l’Hôpital Maisonneuve-Rosemont 5415, Boul de L’Assomption, Montréal, QC, H1T 2M4, Canada.
| |
Collapse
|
24
|
Wu H, Chen C, Gu L, Li J, Yue Y, Lyu M, Cui Y, Zhang X, Liu Y, Zhu H, Liao X, Zhang T, Sun F, Hu W. B cell deficiency promotes the initiation and progression of lung cancer. Front Oncol 2022; 12:1006477. [PMID: 36249034 PMCID: PMC9556970 DOI: 10.3389/fonc.2022.1006477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Currently commercialized CAR-T cell therapies targeting CD19 and BCMA show great efficacy to cure B cell malignancies. However, intravenous infusion of these CAR-T cells severely destroys both transformed and normal B cells in most tissues and organs, in particular lung, leading to a critical question that what the impact of normal B cell depletion on pulmonary diseases and lung cancer is. Herein, we find that B cell frequency is remarkably reduced in both smoking carcinogen-treated lung tissues and lung tumors, which is associated with advanced cancer progression and worse patient survival. B cell depletion by anti-CD20 antibody significantly accelerates the initiation and progression of lung tumors, which is mediated by repressed tumor infiltration of T cells and macrophage elimination of tumor cells. These findings unveil the overall antitumor activity of B cells in lung cancer, providing novel insights into both mechanisms underlying lung cancer pathogenesis and clinical prevention post CAR-T cell therapy.
Collapse
Affiliation(s)
- Han Wu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Chen Chen
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lixing Gu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
- College of Science, Wuhan University of Science and Technology, Wuhan, China
| | - Jiapeng Li
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
- College of Science, Wuhan University of Science and Technology, Wuhan, China
| | - Yunqiang Yue
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Mengqing Lyu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yeting Cui
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaoyu Zhang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yu Liu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Haichuan Zhu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xinghua Liao
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Tongcun Zhang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
- *Correspondence: Tongcun Zhang, ; Fan Sun, ; Weidong Hu,
| | - Fan Sun
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, China
- *Correspondence: Tongcun Zhang, ; Fan Sun, ; Weidong Hu,
| | - Weidong Hu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Tongcun Zhang, ; Fan Sun, ; Weidong Hu,
| |
Collapse
|
25
|
Dynamic Expression of EpCAM in Primary and Metastatic Lung Cancer Is Controlled by Both Genetic and Epigenetic Mechanisms. Cancers (Basel) 2022; 14:cancers14174121. [PMID: 36077658 PMCID: PMC9454530 DOI: 10.3390/cancers14174121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Epithelial cell adhesion molecule (EpCAM) is a tumor marker widely used in both basic studies and clinics. However, our study demonstrates that EpCAM expression is strongly upregulated by gene amplification and promoter hypomethylation in primary lung tumors, but severely downregulated by epigenetic repression (including promoter hypermethylation and histone deacetylation), tumor-associated macrophages (TAMs), and TAMs-derived TGFβ in metastatic lung tumors. DNMT inhibitor 5-aza-dC, HDAC inhibitor MS-275, and TGFβ neutralizing antibody are able to restore EpCAM expression in highly metastatic lung cancer cells. These findings disclose that multiple mechanisms contribute to the dynamic expression patterns of EpCAM in primary and metastatic lung tumors, redefining the application of EpCAM as a biomarker in tumor cell identification and isolation in specific cancers and clinical stages. Abstract Although great progress has been achieved in cancer treatment in the past decades, lung cancer remains the leading cause of cancer death, which is partially caused by the fact that most lung cancers are diagnosed at advanced stages. To improve the sensitivity and specificity of lung cancer diagnosis, the underlying mechanisms of current diagnosis methods are in urgent need to be explored. Herein, we find that the expression of EpCAM, the widely used molecular marker for tumor cell characterization and isolation, is strongly upregulated in primary lung tumors, which is caused by both gene amplification and promoter hypomethylation. In contrast, EpCAM expression is severely repressed in metastatic lung tumors, which can be reversed by epigenetic drugs, DNMT inhibitor 5-aza-dC and HDAC inhibitor MS-275. Moreover, tumor-associated macrophages (TAMs) impede EpCAM expression probably through TGFβ-induced EMT signaling. These findings unveil the dynamic expression patterns of EpCAM and differential roles of epigenetic modification in EpCAM expression in primary and metastatic lung tumors, providing novel insights into tumor cell isolation and lung cancer diagnosis.
Collapse
|
26
|
Mackenzie NJ, Nicholls C, Templeton AR, Perera MPJ, Jeffery PL, Zimmermann K, Kulasinghe A, Kenna TJ, Vela I, Williams ED, Thomas PB. Modelling the tumor immune microenvironment for precision immunotherapy. CLINICAL & TRANSLATIONAL IMMUNOLOGY 2022; 11:e1400. [PMID: 35782339 PMCID: PMC9234475 DOI: 10.1002/cti2.1400] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/14/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Nathan J Mackenzie
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
| | - Clarissa Nicholls
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
| | - Abby R Templeton
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
| | - Mahasha PJ Perera
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
- Department of Urology Princess Alexandra Hospital Woolloongabba QLD Australia
| | - Penny L Jeffery
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
| | - Kate Zimmermann
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Centre for Immunology and Infection Control School of Biomedical Sciences Queensland University of Technology (QUT) Brisbane QLD Australia
- Centre for Microbiome Research School of Biomedical Sciences Queensland University of Technology (QUT) Brisbane QLD Australia
| | - Arutha Kulasinghe
- University of Queensland Diamantina Institute The University of Queensland Brisbane QLD Australia
| | - Tony J Kenna
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Centre for Immunology and Infection Control School of Biomedical Sciences Queensland University of Technology (QUT) Brisbane QLD Australia
- Centre for Microbiome Research School of Biomedical Sciences Queensland University of Technology (QUT) Brisbane QLD Australia
| | - Ian Vela
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
- Department of Urology Princess Alexandra Hospital Woolloongabba QLD Australia
| | - Elizabeth D Williams
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
| | - Patrick B Thomas
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
| |
Collapse
|
27
|
Guo ZS, Qu Z. PDLIM2: Signaling pathways and functions in cancer suppression and host immunity. Biochim Biophys Acta Rev Cancer 2021; 1876:188630. [PMID: 34571051 DOI: 10.1016/j.bbcan.2021.188630] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/30/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022]
Abstract
PDZ and LIM domains-containing proteins play pivotal functions in cell cytoskeleton organization, cell polarization and differentiation. As a key member of the family, PDLIM2 regulates stability and activity of transcription factors such as NF-κB, STATs and β-catenin, and thus exert it functions in inflammation, immunity, and cancer. PDLIM2 functions as a tumor suppressor in multiple tissues and it is often genetically mutated or epigenetically silenced in human cancers derived from lung, breast, ovarian and other histologies. However, in certain types of cancers, PDLIM2 may promote cancer cell proliferation and metastases. Therefore, PDLIM2 is added to a long list of genes that can function as tumor suppressor or oncogenic protein. During tumorigenesis induced by oncogenic viruses, PDLIM2 is a key target. Through promotion of NF-κB/RelA and STAT3 degradation, PDLIM2 enhances expression of proteins involved in antigen presentation and promotes T-cell activation while repressing multidrug resistance genes, thereby rendering mutated cells susceptible to immune surveillance and cytotoxicity mediated by immune cells and chemotherapeutic drugs. Intriguingly, PDLIM2 in alveolar macrophages (AMs) plays key roles in monitoring lung tumorigenesis, as its selective genetic deletion leads to constitutive activation of STAT3, driving monocyte differentiation to AMs with pro-tumorigenic polarization and activation. PDLIM2 has also been explored as a therapeutic target for cancer therapy. At the end of this review, we provide perspectives on this important molecule and discuss the future directions of both basic and translational studies.
Collapse
Affiliation(s)
- Zong Sheng Guo
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Zhaoxia Qu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
Sun F, Li L, Xiao Y, Gregory AD, Shapiro SD, Xiao G, Qu Z. Alveolar Macrophages Inherently Express Programmed Death-1 Ligand 1 for Optimal Protective Immunity and Tolerance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:110-114. [PMID: 34135059 PMCID: PMC8674373 DOI: 10.4049/jimmunol.2100046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/28/2021] [Indexed: 01/12/2023]
Abstract
Macrophages play a central role in lung physiology and pathology. In this study, we show in mice that alveolar macrophages (AMs), unlike other macrophage types (interstitial, peritoneal, and splenic macrophages), constitutively express programmed death-1 ligand 1 (PD-L1), thereby possessing a superior phagocytic ability and the capacity to repress CTLs by cis- and trans-interacting with CD80 and programmed death-1 (PD-1), respectively. This extraordinary ability of AMs assures optimal protective immunity and tolerance within the lung. These findings uncover a unique characteristic of AMs and an innate immune function of PD-L1 and CD80 and therefore help in the understanding of lung physiology, diseases, and PD-L1/PD-1-based immunotherapy.
Collapse
Affiliation(s)
- Fan Sun
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA; and
| | - Liwen Li
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA; and
| | - Yadong Xiao
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA
- Department of Medicine, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA
| | - Alyssa D Gregory
- Department of Medicine, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA
| | - Steven D Shapiro
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA;
- Department of Medicine, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA
| | - Gutian Xiao
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA;
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA; and
| | - Zhaoxia Qu
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA;
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA; and
| |
Collapse
|
29
|
Chi A, He X, Hou L, Nguyen NP, Zhu G, Cameron RB, Lee JM. Classification of Non-Small Cell Lung Cancer's Tumor Immune Micro-Environment and Strategies to Augment Its Response to Immune Checkpoint Blockade. Cancers (Basel) 2021; 13:cancers13122924. [PMID: 34208113 PMCID: PMC8230820 DOI: 10.3390/cancers13122924] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Immune checkpoint blockade (ICB) has become a major treatment for lung cancer. Better understanding of the tumor immune micro-environment (TIME) in non-small cell lung cancer (NSCLC) is urgently needed to better treat it with this type of therapy. In this review, we describe and explore how NSCLC’s TIME relates to response to ICB, as well as how to treat those with unresponsive types of TIME, which will significantly impact future research in lung cancer immunotherapy. Abstract Immune checkpoint blockade (ICB) with checkpoint inhibitors has led to significant and durable response in a subset of patients with advanced stage EGFR and ALK wild-type non-small cell lung cancer (NSCLC). This has been consistently shown to be correlated with the unique characteristics of each patient’s tumor immune micro-environment (TIME), including the composition and distribution of the tumor immune cell infiltrate; the expression of various checkpoints by tumor and immune cells, such as PD-L1; and the presence of various cytokines and chemokines. In this review, the classification of various types of TIME that are present in NSCLC and their correlation with response to ICB in NSCLC are discussed. This is conducted with a focus on the characteristics and identifiable biomarkers of different TIME subtypes that may also be used to predict NSCLC’s clinical response to ICB. Finally, treatment strategies to augment response to ICB in NSCLC with unresponsive types of TIME are explored.
Collapse
Affiliation(s)
- Alexander Chi
- Department of Radiation Oncology, Beijing Chest Hospital, Capital Medical University, Beijing 101100, China
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing 210009, China
- Correspondence: (A.C.); (X.H.)
| | - Xia He
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing 210009, China
- Correspondence: (A.C.); (X.H.)
| | - Lin Hou
- Center for Statistical Science, Tsinghua University, Beijing 100084, China;
| | - Nam P. Nguyen
- Department of Radiation Oncology, Howard University, Washington, DC 20060, USA;
| | - Guangying Zhu
- Department of Radiation Oncology, China-Japan Friendship Hospital, Beijing 100029, China;
| | - Robert B. Cameron
- Division of Thoracic Surgery, Department of Surgery, University of California at Los Angeles, Los Angeles, CA 90095, USA; (R.B.C.); (J.M.L.)
| | - Jay M. Lee
- Division of Thoracic Surgery, Department of Surgery, University of California at Los Angeles, Los Angeles, CA 90095, USA; (R.B.C.); (J.M.L.)
| |
Collapse
|
30
|
Cao Y, Li Q, Liu H, He X, Huang F, Wang Y. Role of Tim-3 in regulating tumorigenesis, inflammation, and antitumor immunity therapy. Cancer Biomark 2021; 32:237-248. [PMID: 34092621 DOI: 10.3233/cbm-210114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Over the past decade, cancer immunotherapy, such as immune checkpoint inhibitors (ICRs), has attained considerable progresses in clinical practice. T-cell immunoglobulin and mucin domain-containing protein 3 (Tim-3) act as next ICRs, and originally function as a co-inhibitory receptor expressed on interferon (IFN)-γ producing CD4+ and CD8+ T-cells. Furthermore, Tim-3 has also been found to express on innate immune cells and several types of tumors, signifying the pivotal role that Tim-3 plays in chronic viral infections and cancer. In addition, Tim-3 and multiple ICRs are concurrently expressed and regulated on dysfunctional or exhausted T-cells, leading to improved antitumor immune responses in preclinical or clinical cancer therapy through co-blockade of Tim-3 and other ICRs such as programmed cell death-1 (PD-1). In this review, the biological characteristics of Tim-3 and the function of Tim-3 in regulating tumorigenesis and inflammation have been summarized. The usage of a single blockade of Tim-3 or in combination with multiple immunotherapy regimens have drawn attention to antitumor potential as a target for immunotherapy.
Collapse
Affiliation(s)
- Yuting Cao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Qiang Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Huihui Liu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Xianglei He
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Fang Huang
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| |
Collapse
|
31
|
Zhang Y, Li Y, Chen K, Qian L, Wang P. Oncolytic virotherapy reverses the immunosuppressive tumor microenvironment and its potential in combination with immunotherapy. Cancer Cell Int 2021; 21:262. [PMID: 33985527 PMCID: PMC8120729 DOI: 10.1186/s12935-021-01972-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
It has been intensively reported that the immunosuppressive tumor microenvironment (TME) results in tumor resistance to immunotherapy, especially immune checkpoint blockade and chimeric T cell antigen therapy. As an emerging therapeutic agent, oncolytic viruses (OVs) can specifically kill malignant cells and modify immune and non-immune TME components through their intrinsic properties or genetically incorporated with TME regulators. Strategies of manipulating OVs against the immunosuppressive TME include serving as a cancer vaccine, expressing proinflammatory factors and immune checkpoint inhibitors, and regulating nonimmune stromal constituents. In this review, we summarized the mechanisms and applications of OVs against the immunosuppressive TME, and strategies of OVs in combination with immunotherapy. We also introduced future directions to achieve efficient clinical translation including optimization of preclinical models that simulate the human TME and achieving systemic delivery of OVs.
Collapse
Affiliation(s)
- Yalei Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ye Li
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
| | - Kun Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ling Qian
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peng Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
32
|
Yang C, Hua N, Xie S, Wu Y, Zhu L, Wang S, Tong X. Oncolytic viruses as a promising therapeutic strategy for hematological malignancies. Biomed Pharmacother 2021; 139:111573. [PMID: 33894623 DOI: 10.1016/j.biopha.2021.111573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/23/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
Abstract
The incidence of hematological malignancies such as multiple myeloma, leukemia, and lymphoma has increased over time. Although bone marrow transplantation, immunotherapy and chemotherapy have led to significant improvements in efficacy, poor prognosis in elderly patients, recurrence and high mortality among hematological malignancies remain major challenges, and innovative therapeutic strategies should be explored. Besides directly lyse tumor cells, oncolytic viruses can activate immune responses or be engineered to express therapeutic factors to increase antitumor efficacy, and have gradually been recognized as an appealing approach for fighting cancers. An increasing number of studies have applied oncolytic viruses in hematological malignancies and made progress. In particular, strategies combining immunotherapy and oncolytic virotherapy are emerging. Various phase I clinical trials of oncolytic reovirus with lenalidomide or programmed death 1(PD-1) immune checkpoint inhibitors in multiple myeloma are ongoing. Moreover, preclinical studies of combinations with chimeric antigen receptor T (CAR-T) cells are underway. Thus, oncolytic virotherapy is expected to be a promising approach to cure hematological malignancies. This review summarizes progress in oncolytic virus research in hematological malignancies. After briefly reviewing the development and oncolytic mechanism of oncolytic viruses, we focus on delivery methods of oncolytic viruses, especially systemic delivery that is suitable for hematological tumors. We then discuss the main types of oncolytic viruses applied for hematological malignancies and related clinical trials. In addition, we present several ways to improve the antitumor efficacy of oncolytic viruses. Finally, we discuss current challenges and provide suggestions for future studies.
Collapse
Affiliation(s)
- Chen Yang
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; Department of Clinical Medicine, Qingdao University, Qingdao, PR China
| | - Nanni Hua
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Shufang Xie
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Yi Wu
- Phase I clinical research center, Zhejiang Provincial People's Hospital,Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China
| | - Lifeng Zhu
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China
| | - Shibing Wang
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital ,Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China.
| | - Xiangmin Tong
- Molecular diagnosis laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, PR China; The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital ,Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China.
| |
Collapse
|
33
|
Kong X, Lu P, Liu C, Guo Y, Yang Y, Peng Y, Wang F, Bo Z, Dou X, Shi H, Meng J. A combination of PD‑1/PD‑L1 inhibitors: The prospect of overcoming the weakness of tumor immunotherapy (Review). Mol Med Rep 2021; 23:362. [PMID: 33760188 PMCID: PMC7985997 DOI: 10.3892/mmr.2021.12001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
Programmed cell death protein-1 (PD-1)/programmed death protein ligand-1 (PD-L1) inhibitors for treatment of a various types of cancers have revolutionized cancer immunotherapy. However, PD-1/PD-L1 inhibitors are associated with a low response rate and are only effective on a small number of patients with cancer. Development of an anti-PD-1/PD-L1 sensitizer for improving response rate and effectiveness of immunotherapy is a challenge. The present study reviews the synergistic effects of PD-1/PD-L1 inhibitor with oncolytic virus, tumor vaccine, molecular targeted drugs, immunotherapy, chemotherapy, radiotherapy, intestinal flora and traditional Chinese medicine, to provide information for development of effective combination therapies.
Collapse
Affiliation(s)
- Xianbin Kong
- Integrated Traditional Chinese and Western Medicine Laboratory, College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Peng Lu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Chuanxin Liu
- Department of Pharmaceutical Analysis, School of Chinese Materia Medical, Beijing University of Chinese Medicine, Beijing 102488, P.R. China
| | - Yuzhu Guo
- Department of Radiotherapy, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Yuying Yang
- Integrated Traditional Chinese and Western Medicine Laboratory, College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Yingying Peng
- Integrated Traditional Chinese and Western Medicine Laboratory, College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Fangyuan Wang
- Integrated Traditional Chinese and Western Medicine Laboratory, College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Zhichao Bo
- Integrated Traditional Chinese and Western Medicine Laboratory, College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Xiaoxin Dou
- Integrated Traditional Chinese and Western Medicine Laboratory, College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Haoyang Shi
- Integrated Traditional Chinese and Western Medicine Laboratory, College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Jingyan Meng
- Integrated Traditional Chinese and Western Medicine Laboratory, College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| |
Collapse
|
34
|
Li L, Sun F, Han L, Liu X, Xiao Y, Gregory AD, Shapiro SD, Xiao G, Qu Z. PDLIM2 repression by ROS in alveolar macrophages promotes lung tumorigenesis. JCI Insight 2021; 6:144394. [PMID: 33539325 PMCID: PMC8021114 DOI: 10.1172/jci.insight.144394] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/29/2021] [Indexed: 01/01/2023] Open
Abstract
One of the most fundamental and challenging questions in the field of cancer is how immunity is transformed from tumor immunosurveillance to tumor-promoting inflammation. Here, we identified the tumor suppressor PDZ-LIM domain–containing protein 2 (PDLIM2) as a checkpoint of alveolar macrophages (AMs) important for lung tumor suppression. During lung tumorigenesis, PDLIM2 expression in AMs is downregulated by ROS-activated transcription repressor BTB and CNC homology 1 (BACH1). PDLIM2 downregulation leads to constitutive activation of the transcription factor STAT3, driving AM protumorigenic polarization/activation and differentiation from monocytes attracted from the circulation to suppress cytotoxic T lymphocytes and promote lung cancer. PDLIM2 downregulation also decreases AM phagocytosis. These findings establish ROS/BACH1/PDLIM2/STAT3 as a signaling pathway driving AMs for lung tumor promotion.
Collapse
Affiliation(s)
- Liwen Li
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Fan Sun
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lei Han
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Xujie Liu
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yadong Xiao
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Medicine, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alyssa D Gregory
- Department of Medicine, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Steven D Shapiro
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Medicine, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gutian Xiao
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Zhaoxia Qu
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
35
|
Lin X, Wang X, Gu Q, Lei D, Liu X, Yao C. Emerging nanotechnological strategies to reshape tumor microenvironment for enhanced therapeutic outcomes of cancer immunotherapy. Biomed Mater 2021; 16. [PMID: 33601351 DOI: 10.1088/1748-605x/abe7b3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/18/2021] [Indexed: 12/12/2022]
Abstract
Immunotherapy was emerged as a novel cancer treatment in the last decade, however, efficacious responses to mono-immunotherapy have only been achieved in a relatively small portion of patients whereas combinational immunotherapies often lead to concurrent side effects. It has been proved that the tumor microenvironment (TME) is responsible for tumor immune escape and the ultimate treatment failure. Recently, both the understanding of the TME and the applications of nanotechnological strategies have achieved remarkable progresses, and reviewing the emerging immune-regulatory nanosystems may provide valuable information for specifically modulating the TME at different immune stages. In this review, we focus on comprehending the recently proposed T-cell-based tumor classification and identifying the most promising targets for different tumor phenotypes, and then summarizing the nanotechnological strategies to best target corresponding immune-related factors. For future precise personalized immunotherapy, the tailor-made TME modulation strategies conducted by well-designed nanosystems to alleviate the suppressive TME and then promote anti-tumor immune responses will significantly benefit the clinical outcomes of cancer patients.
Collapse
Affiliation(s)
- Xinyi Lin
- Xi'an Jiaotong University School of Life Science and Technology, NO. 28 Xianning Xi Road, Xi'an, Shaanxi, 710049, CHINA
| | - Xiaoyan Wang
- Fujian Agriculture and Forestry University, NO.15 Shangdian Road, Fuzhou, 350002, CHINA
| | - Qing Gu
- Xi'an Jiaotong University School of Life Science and Technology, NO.28 Xianning Xi Road, Xi'an, 710049, CHINA
| | - Dongqin Lei
- Xi'an Jiaotong University, NO.28 Xianning Xi Road, Xi'an, 710049, CHINA
| | - Xiaolong Liu
- Mengchao Hepatobiliary Hospital of Fujian Medical University, NO.312 Xihong Road, Fuzhou, Fujian, 350025, CHINA
| | - Cuiping Yao
- Xi'an Jiaotong University School of Life Science and Technology, NO.28 Xianning Xi Road, Xi'an, Shaanxi, 710049, CHINA
| |
Collapse
|
36
|
Immunotherapy for Glioblastoma: Current State, Challenges, and Future Perspectives. Cancers (Basel) 2020; 12:cancers12092334. [PMID: 32824974 PMCID: PMC7565291 DOI: 10.3390/cancers12092334] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/10/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most lethal intracranial primary malignancy by no optimal treatment option. Cancer immunotherapy has achieved remarkable survival benefits against various advanced tumors, such as melanoma and non-small-cell lung cancer, thus triggering great interest as a new therapeutic strategy for glioblastoma. Moreover, the central nervous system has been rediscovered recently as a region for active immunosurveillance. There are vibrant investigations for successful glioblastoma immunotherapy despite the fact that initial clinical trial results are somewhat disappointing with unique challenges including T-cell dysfunction in the patients. This review will explore the potential of current immunotherapy modalities for glioblastoma treatment, especially focusing on major immune checkpoint inhibitors and the future strategies with novel targets and combo therapies. Immune-related adverse events and clinical challenges in glioblastoma immunotherapy are also summarized. Glioblastoma provides persistent difficulties for immunotherapy with a complex state of patients’ immune dysfunction and a variety of constraints in drug delivery to the central nervous system. However, rational design of combinational regimens and new focuses on myeloid cells and novel targets to circumvent current limitations hold promise to advent truly viable immunotherapy for glioblastoma.
Collapse
|
37
|
Liu W, Zhang L, Xiu Z, Guo J, Wang L, Zhou Y, Jiao Y, Sun M, Cai J. Combination of Immune Checkpoint Inhibitors with Chemotherapy in Lung Cancer. Onco Targets Ther 2020; 13:7229-7241. [PMID: 32801752 PMCID: PMC7394580 DOI: 10.2147/ott.s255491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
Tremendous progress has been achieved in the field of immune checkpoint inhibitors (ICIs) therapy in lung cancer in recent years. To generate robust, long-lasting anti-tumor immune responses in lung cancer patients, combinational ICI therapies have been explored deeply. Conventionally, chemotherapy was considered as immunosuppressive. It is now recognized that chemotherapy could also reinstate cancer cell immune-surveillance and enable the perception of cancer cells as dangerous. That is to say that chemotherapeutic drugs are not only a source of direct cytotoxic effects but also an adjuvant for anti-tumor immunity. Recently, multiple clinical studies of ICIs combined with chemotherapeutic drugs have been explored and proved effective. However, there are still crucial questions that are not well addressed, such as the optimal dose and schedule for a given combination may differ across disease indications, and the appropriate strategy of selecting patient population that can benefit from ICIs remains unclear. To facilitate more rational lung cancer ICIs therapy development, this review summarizes the immune-regulatory effects and related mechanisms of chemotherapeutic drugs and the clinical progress of ICIs and their combination with chemotherapies in lung cancer treatment.
Collapse
Affiliation(s)
- Wei Liu
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China.,Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Lei Zhang
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China.,Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Zhiming Xiu
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Jian Guo
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Liye Wang
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX 77030, USA
| | - Yue Zhou
- Department of Statistics, North Dakota State University, Fargo, ND 58102, USA
| | - Yang Jiao
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Meiyan Sun
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China
| | - Jianhui Cai
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, People's Republic of China.,Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin 132013, People's Republic of China
| |
Collapse
|