1
|
Zhang X, Jiang S, Yin H, Zhang H, Yang L, Sun P, Kuai X, Chen C, Huang J. Enhanced IL36RN Expression and Its Association With Immune Microenvironment Predicts Poor Prognosis in Gastric Cancer. Cancer Med 2025; 14:e70954. [PMID: 40347070 DOI: 10.1002/cam4.70954] [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: 11/06/2024] [Revised: 03/18/2025] [Accepted: 04/29/2025] [Indexed: 05/12/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) remains a prevalent and lethal malignancy worldwide, underscoring the urgent need to identify novel therapeutic targets and elucidate the tumor microenvironment (TME) to enhance clinical outcomes. METHODS IL36RN mRNA expression in GC tissues was analyzed using The Cancer Genome Atlas (TCGA) dataset. Bioinformatics approaches, cellular models, and clinical tissue microarrays were employed to investigate the functional role of IL36RN, its interactions within the TME, and its prognostic significance. RESULTS IL36RN expression was markedly upregulated in GC tissues and associated with unfavorable survival outcomes. Functional assays demonstrated that IL36RN silencing suppressed GC cell proliferation and invasion. Elevated IL36RN expression correlated with enhanced CD8+ T cell infiltration in the TME and served as an independent prognostic indicator in GC. CONCLUSIONS IL36RN represents a potential prognostic biomarker and therapeutic target in GC, offering novel avenues for precision oncology and immunotherapeutic intervention.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Sutian Jiang
- Department of Pathology, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Hang Yin
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong University, Nantong, Jiangsu, China
| | - Hui Zhang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong University, Nantong, Jiangsu, China
| | - Lei Yang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Pingping Sun
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiaoling Kuai
- Department of Gastroenterology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Chen Chen
- Department of Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jianfei Huang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
2
|
Finucane M, Brint E, Houston A. The complex roles of IL-36 and IL-38 in cancer: friends or foes? Oncogene 2025; 44:851-861. [PMID: 40057603 PMCID: PMC11932923 DOI: 10.1038/s41388-025-03293-4] [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: 09/30/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 03/26/2025]
Abstract
The interleukin-36 (IL-36) family comprises of three pro-inflammatory receptor agonists (IL-36α, IL-36β and IL-36γ), two anti-inflammatory receptor antagonists (IL-36RA and IL-38) along with the IL-36 receptor (IL-36R). Part of the IL-1 cytokine superfamily, the IL-36 family was discovered in the early 2000s due to the homology of its member sequences to the IL-1 cytokines. As pro- and anti-inflammatory cytokines, respectively, IL-36α, IL-36β, IL-36γ and IL-38 aid in maintaining homoeostasis by reciprocally regulating the body's response to damage and disease through IL-36R-associated signalling. With the significant roles of IL-36α, IL-36β and IL-36γ in regulating the immune response realised, interest has grown in investigating their roles in cancer. While initial studies indicated solely tumour-suppressing roles, more recent work has identified tumour-promoting roles in cancer, suggesting a more complex dual functionality of the IL-36 cytokines. The activity of IL-38 in cancer is similarly complex, with the receptor antagonist displaying distinct tumour-suppressive roles, particularly in colorectal cancer (CRC), in addition to broad tumour-promoting roles in various other malignancies. This review provides a comprehensive overview of the IL-36 and IL-38 cytokines, their activation and IL-36R signalling, the physiological functions of these cytokines, and their activity in cancer.
Collapse
Affiliation(s)
- Méabh Finucane
- Department of Medicine, School of Medicine, University College Cork, Cork, Ireland
- Department of Pathology, School of Medicine, Cork University Hospital, University College Cork, Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, School of Medicine, Cork University Hospital, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Aileen Houston
- Department of Medicine, School of Medicine, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
3
|
Subhi-Issa N, Tovar Manzano D, Pereiro Rodriguez A, Sanchez Ramon S, Perez Segura P, Ocaña A. γδ T Cells: Game Changers in Immune Cell Therapy for Cancer. Cancers (Basel) 2025; 17:1063. [PMID: 40227601 PMCID: PMC11987767 DOI: 10.3390/cancers17071063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/13/2025] [Indexed: 04/15/2025] Open
Abstract
Gamma delta (γδ) T cells are a unique subset of T lymphocytes with distinctive features that make them highly promising candidates for cancer therapy. Their MHC-independent recognition of tumor antigens, ability to mediate direct cytotoxicity, and role in modulating the tumor microenvironment position them as versatile agents in cancer immunotherapy. This review integrates and synthesizes the existing data on γδ T cells, with an emphasis on the development and optimization of in vitro expansion protocols. Critical aspects are detailed such as activation strategies, co-culture systems, cytokine use, and other parameters to ensure robust cell proliferation and functionality, which may be valuable for those developing or optimizing clinical practices. Finally, we discuss current advancements in γδ T cell research, clinical experience, and highlight areas needing further exploration. Considering these data, we hypothesize and propose potential new applications such as engineering γδ T cells for enhanced resistance to immune checkpoint pathways or for localized cytokine delivery within the tumor microenvironment, which could broaden their therapeutic impact in the treatment of cancer and beyond.
Collapse
Affiliation(s)
- Nabil Subhi-Issa
- Department of Immunology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | - Daniel Tovar Manzano
- Department of Immunology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | | | - Silvia Sanchez Ramon
- Department of Immunology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
- Department of Immunology, Ophthalmology, and ORL, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Pedro Perez Segura
- Department of Oncology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain (A.O.)
| | - Alberto Ocaña
- Department of Oncology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain (A.O.)
| |
Collapse
|
4
|
Hu Y, Cai ZR, Huang RZ, Wang DS, Ju HQ, Chen DL. Circular RNA circPHLPP2 promotes tumor growth and anti-PD-1 resistance through binding ILF3 to regulate IL36γ transcription in colorectal cancer. Mol Cancer 2024; 23:272. [PMID: 39695693 DOI: 10.1186/s12943-024-02192-8] [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: 09/03/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Most Colorectal Cancer (CRC) patients exhibit limited responsiveness to anti-programmed cell death protein 1 (PD-1) therapy, with the underlying mechanisms remaining elusive. Circular RNAs (circRNAs) play a significant role in tumorigenesis and development, with potential applications in tumor screening and predicting treatment efficacy. However, there are few studies exploring the role of circRNAs in CRC immune evasion. METHODS circRNA microarrays were used to identify circPHLPP2. RT-qPCR was used to examine the associations between the expression level of circPHLPP2 and the clinical characteristics of CRC patients. MTS assay, clone formation experiment, subcutaneous tumor implantation and multicolor flow cytometry were used to confirm the biological function of circPHLPP2. RAN-seq, RT-qPCR, and WB experiments were performed to investigate the downstream signaling pathways involved in circPHLPP2. RNA pull-down, RNA immunoprecipitation (RIP) and immunofluorescence staining were performed to identify the proteins associated with circPHLPP2. RESULTS circPHLPP2 is up-regulated in CRC patients who exhibit resistance to anti-PD-1 based therapy. circPHLPP2 significantly promotes the proliferation and tumor growth of CRC cells. Knockdown of circPhlpp2 enhances the efficacy of anti-PD-1 in vivo. Mechanistically, the specific interaction between circPHLPP2 and ILF3 facilitates the nuclear accumulation of ILF3, which subsequently enhances the transcription of IL36γ. This process reduces NK cell infiltration and impairs NK cells' granzyme B and IFN-γ production, thereby promoting tumor progression. CONCLUSIONS Overall, our findings reveal a novel mechanism by which circRNA regulates CRC immune evasion. circPHLPP2 may serve as a prognostic biomarker and potential therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Yan Hu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, P. R. China
| | - Ze-Rong Cai
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, P. R. China
| | - Ren-Ze Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, P. R. China
| | - De-Shen Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, P. R. China
| | - Huai-Qiang Ju
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, P. R. China
| | - Dong-Liang Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, P. R. China.
| |
Collapse
|
5
|
Tan Y, Mu J, Chen J. IL-36 Gamma: A Novel Adjuvant Cytokine Enhancing Protective Immunity Induced by DNA Immunization with TGIST and TGNSM Against Toxoplasma gondii Infection in Mice. Microorganisms 2024; 12:2258. [PMID: 39597646 PMCID: PMC11596725 DOI: 10.3390/microorganisms12112258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Toxoplasma gondii can cause congenital infections and abortions in humans. TgIST and TgNSM play critical roles in intracellular cyst formation and chronic infection. However, no studies have explored their potential to induce protective immunity against T. gondii infection. OBJECTIVE To evaluate the immune efficacy of DNA vaccines encoding TgNSM and TgIST genes against T. gondii infection, using the acute and chronic ME49 strain (Type II). METHODS DNA vaccines, including eukaryotic plasmids pVAX-IST and pVAX-NSM, were constructed. A cocktail DNA vaccine combining these two genes was formulated. The expression and immunogenicity were determined using the indirect immunofluorescence assay (IFA). Mice were immunized with DNA vaccines encoding either TgIST or TgNSM, as well as with the cocktail DNA vaccine. Humoral and cellular immune responses were analyzed by detecting antibody levels, cytotoxic T cell (CTL) responses, cytokines, and lymphocyte surface markers. Mouse survival and brain cyst counts were assessed 1 to 2 months post-vaccination in experimental toxoplasmosis models. The adjuvant efficacy of plasmid pVAX-IL-36γ in enhancing DNA vaccine-induced protective immunity was also evaluated. RESULTS DNA immunization with pVAX-IST and pVAX-NSM elicited strong humoral and cellular immune responses, characterized by increased Toxoplasma-specific IgG2a titers, Th1 responses (including production of IFN-γ, IL-2, IL-12p40, and IL-12p70), and cell-mediated activity with elevated frequencies of CD8+ and CD4+ T cells, and CTL responses. This provided significant protective efficacy against acute and chronic T. gondii infection. Mice immunized with the two-gene cocktail (pVAX-IST + pVAX-NSM) showed greater protection than those immunized with single-gene vaccines. Co-administration of the molecular adjuvant pVAX-IL-36γ further enhanced the protective immunity induced by the cocktail DNA vaccine. CONCLUSIONS TgIST and TgNSM induce effective immunity against T. gondii infection, making them promising vaccine candidates against toxoplasmosis. Additionally, IL-36γ is a promising genetic adjuvant that enhances protective immunity in a vaccine setting against T. gondii, and it should be evaluated in strategies against other apicomplexan parasites.
Collapse
Affiliation(s)
| | | | - Jia Chen
- Department of Radiology, The Affiliated People’s Hospital of Ningbo University, Ningbo 315040, China; (Y.T.); (J.M.)
| |
Collapse
|
6
|
Eslami SM, Lu X. Recent advances in mRNA-based cancer vaccines encoding immunostimulants and their delivery strategies. J Control Release 2024; 376:S0168-3659(24)00708-9. [PMID: 39437963 DOI: 10.1016/j.jconrel.2024.10.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/01/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
The high prevalence of drug resistance, relapse, and unfavorable response rate of conventional cancer therapies necessitate the development of more efficient treatment modalities. Immunotherapy represents a novel therapeutic approach to cancer treatment in which the immune system's potential is harnessed to recognize and eliminate tumor cells. mRNA cancer vaccines, as a burgeoning field of immunotherapy, have recently drawn particular attention, and among mRNAs encoding tumor-associated antigens, tumor-specific antigens, and immune stimulatory factors, the latter has been relatively less explored. These immunostimulatory mRNAs encode a range of proteins, including stimulatory ligands, receptors, enzymes, pro-inflammatory cytokines, and inhibitory binding proteins, which collectively augment the host immune system's ability against cancerous cells. In this review, we aimed to provide a comprehensive account of mRNA-based cancer vaccines encoding immune stimulants, encompassing their current status, mechanisms of action, delivery strategies employed, as well as recent advances in preclinical and clinical studies. The potential challenges, strategies and future perspectives have also been discussed.
Collapse
Affiliation(s)
- Seyyed Majid Eslami
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA.
| | - Xiuling Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA.
| |
Collapse
|
7
|
Sugiura K, Fujita H, Komine M, Yamanaka K, Akiyama M. The role of interleukin-36 in health and disease states. J Eur Acad Dermatol Venereol 2024; 38:1910-1925. [PMID: 38779986 DOI: 10.1111/jdv.19935] [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: 11/03/2023] [Accepted: 01/29/2024] [Indexed: 05/25/2024]
Abstract
The interleukin (IL)-1 superfamily upregulates immune responses and maintains homeostasis between the innate and adaptive immune systems. Within the IL-1 superfamily, IL-36 plays a pivotal role in both innate and adaptive immune responses. Of the four IL-36 isoforms, three have agonist activity (IL-36α, IL-36β, IL-36γ) and the fourth has antagonist activity (IL-36 receptor antagonist [IL-36Ra]). All IL-36 isoforms bind to the IL-36 receptor (IL-36R). Binding of IL-36α/β/γ to the IL-36R recruits the IL-1 receptor accessory protein (IL-1RAcP) and activates downstream signalling pathways mediated by nuclear transcription factor kappa B and mitogen-activated protein kinase signalling pathways. Antagonist binding of IL-36Ra to IL-36R inhibits recruitment of IL-1RAcP, blocking downstream signalling pathways. Changes in the balance within the IL-36 cytokine family can lead to uncontrolled inflammatory responses throughout the body. As such, IL-36 has been implicated in numerous inflammatory diseases, notably a type of pustular psoriasis called generalized pustular psoriasis (GPP), a chronic, rare, potentially life-threatening, multisystemic skin disease characterised by recurrent fever and extensive sterile pustules. In GPP, IL-36 is central to disease pathogenesis, and the prevention of IL-36-mediated signalling can improve clinical outcomes. In this review, we summarize the literature describing the biological functions of the IL-36 pathway. We also consider the evidence for uncontrolled activation of the IL-36 pathway in a wide range of skin (e.g., plaque psoriasis, pustular psoriasis, hidradenitis suppurativa, acne, Netherton syndrome, atopic dermatitis and pyoderma gangrenosum), lung (e.g., idiopathic pulmonary fibrosis), gut (e.g., intestinal fibrosis, inflammatory bowel disease and Hirschsprung's disease), kidney (e.g., renal tubulointerstitial lesions) and infectious diseases caused by a variety of pathogens (e.g., COVID-19; Mycobacterium tuberculosis, Pseudomonas aeruginosa, Streptococcus pneumoniae infections), as well as in cancer. We also consider how targeting the IL-36 signalling pathway could be used in treating inflammatory disease states.
Collapse
Affiliation(s)
- Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hideki Fujita
- Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Mayumi Komine
- Department of Dermatology, Faculty of Medicine, Jichi Medical University, Tochigi, Japan
| | - Keiichi Yamanaka
- Department of Dermatology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Masashi Akiyama
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
8
|
Keller J, O' Siorain JR, Kündig TM, Mellett M. Molecular aspects of Interleukin-36 cytokine activation and regulation. Biochem Soc Trans 2024; 52:1591-1604. [PMID: 38940747 DOI: 10.1042/bst20230548] [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: 04/05/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024]
Abstract
Interleukin-36 (IL-36) cytokines are structurally similar to other Interleukin-1 superfamily members and are essential to convey inflammatory responses at epithelial barriers including the skin, lung, and gut. Due to their potent effects on immune cells, IL-36 cytokine activation is regulated on multiple levels, from expression and activation to receptor binding. Different IL-36 isoforms convey specific responses as a consequence of particular danger- or pathogen-associated molecular patterns. IL-36 expression and activation are regulated by exogenous pathogens, including fungi, viruses and bacteria but also by endogenous factors such as antimicrobial peptides or cytokines. Processing of IL-36 into potent bioactive forms is necessary for host protection but can elevate tissue damage. Indeed, exacerbated IL-36 signalling and hyperactivation are linked to the pathogenesis of diseases such as plaque and pustular psoriasis, emphasising the importance of understanding the molecular aspects regulating IL-36 activation. Here, we summarise facets of the electrochemical properties, regulation of extracellular cleavage by various proteases and receptor signalling of the pro-inflammatory and anti-inflammatory IL-36 family members. Additionally, this intriguing cytokine subfamily displays many characteristics that are unique from prototypical members of the IL-1 family and these key distinctions are outlined here.
Collapse
Affiliation(s)
- Jennifer Keller
- Department of Dermatology, University Hospital Zürich (USZ), University of Zürich (UZH), Raemistrasse 100, 8091 Zürich, Switzerland
- Faculty of Science, University of Zürich, 8091 Zürich, Switzerland
| | - James R O' Siorain
- Department of Dermatology, University Hospital Zürich (USZ), University of Zürich (UZH), Raemistrasse 100, 8091 Zürich, Switzerland
- Faculty of Medicine, University of Zürich, 8091 Zürich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zürich (USZ), University of Zürich (UZH), Raemistrasse 100, 8091 Zürich, Switzerland
- Faculty of Medicine, University of Zürich, 8091 Zürich, Switzerland
| | - Mark Mellett
- Department of Dermatology, University Hospital Zürich (USZ), University of Zürich (UZH), Raemistrasse 100, 8091 Zürich, Switzerland
- Faculty of Medicine, University of Zürich, 8091 Zürich, Switzerland
| |
Collapse
|
9
|
Lecoultre M, Walker PR, El Helali A. Oncolytic virus and tumor-associated macrophage interactions in cancer immunotherapy. Clin Exp Med 2024; 24:202. [PMID: 39196415 PMCID: PMC11358230 DOI: 10.1007/s10238-024-01443-8] [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: 05/30/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024]
Abstract
Oncolytic viruses (OV) are a promising strategy in cancer immunotherapy. Their capacity to promote anti-tumoral immunity locally raises hope that cancers unresponsive to current immunotherapy approaches could be tackled more efficiently. In this context, tumor-associated macrophages (TAM) must be considered because of their pivotal role in cancer immunity. Even though TAM tend to inhibit anti-tumoral responses, their ability to secrete pro-inflammatory cytokines and phagocytose cancer cells can be harnessed to promote therapeutic cancer immunity. OVs have the potential to promote TAM pro-inflammatory functions that favor anti-tumoral immunity. But in parallel, TAM pro-inflammatory functions induce OV clearance in the tumor, thereby limiting OV efficacy and highlighting that the interaction between OV and TAM is a double edge sword. Moreover, engineered OVs were recently developed to modulate specific TAM functions such as phagocytic activity. The potential of circulating monocytes to deliver OV into the tumor after intravenous administration is also emerging. In this review, we will present the interaction between OV and TAM, the potential of engineered OV to modulate specific TAM functions, and the promising role of circulating monocytes in OV delivery to the tumor.
Collapse
Affiliation(s)
- Marc Lecoultre
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, China
- Division of General Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Paul R Walker
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Immunobiology of Brain Tumours Laboratory, Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland
| | - Aya El Helali
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, China.
| |
Collapse
|
10
|
Tongmuang N, Krishnan M, Connor V, Crump C, Jensen LE. UL56 Is Essential for Herpes Simplex Virus-1 Virulence In Vivo but Is Dispensable for Induction of Host-Protective Immunity. Vaccines (Basel) 2024; 12:837. [PMID: 39203963 PMCID: PMC11359923 DOI: 10.3390/vaccines12080837] [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: 06/18/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Herpes simplex virus-1 (HSV-1) is common and can cause significant disease in humans. Unfortunately, efforts to develop effective vaccines against HSV-1 have so far failed. A detailed understanding of how the virus infects its host and how the host mounts potent immune responses against the virus may inform new vaccine approaches. Here, using a zosteriform mouse model, we examined how the HSV-1 gene UL56 affects the ability of the virus to cause morbidity and generate protective immunity. A UL56 deletion mutant, ΔUL56, was derived from the wild-type HSV-1 strain SC16, alongside a revertant strain in which UL56 was reintroduced in ΔUL56. In vitro, the three virus strains replicated in a similar manner; however, in vivo, only the wild type and the revertant strains caused shingles-like skin lesions and death. Mice previously infected with ΔUL56 became resistant to a lethal challenge with the wild-type SC16. The protective immunity induced by ΔUL56 was independent of IL-1, IL-33, and IL-36 signaling through IL-1RAP. Both skin and intramuscular ΔUL56 inoculation generated protective immunity against a lethal SC16 challenge. After 6 months, female mice remained resistant to infection, while male mice exhibited signs of declining protection. Our data demonstrate that UL56 is important for the ability of HSV-1 to spread within the infected host and that a ∆UL56 strain elicits an effective immune response against HSV-1 despite this loss of virulence. These findings may guide further HSV-1 vaccine development.
Collapse
Affiliation(s)
- Nopprarat Tongmuang
- Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19104, USA
- Center for Inflammation and Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19104, USA
| | - Meera Krishnan
- Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19104, USA
- Center for Inflammation and Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19104, USA
| | - Viv Connor
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Colin Crump
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Liselotte E. Jensen
- Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19104, USA
- Center for Inflammation and Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19104, USA
- Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Temple Health, Philadelphia, PA 19111, USA
| |
Collapse
|
11
|
Xu MY, Zeng N, Liu CQ, Sun JX, An Y, Zhang SH, Xu JZ, Zhong XY, Ma SY, He HD, Hu J, Xia QD, Wang SG. Enhanced cellular therapy: revolutionizing adoptive cellular therapy. Exp Hematol Oncol 2024; 13:47. [PMID: 38664743 PMCID: PMC11046957 DOI: 10.1186/s40164-024-00506-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 03/31/2024] [Indexed: 04/28/2024] Open
Abstract
Enhanced cellular therapy has emerged as a novel concept following the basis of cellular therapy. This treatment modality applied drugs or biotechnology to directly enhance or genetically modify cells to enhance the efficacy of adoptive cellular therapy (ACT). Drugs or biotechnology that enhance the killing ability of immune cells include immune checkpoint inhibitors (ICIs) / antibody drugs, small molecule inhibitors, immunomodulatory factors, proteolysis targeting chimera (PROTAC), oncolytic virus (OV), etc. Firstly, overcoming the inhibitory tumor microenvironment (TME) can enhance the efficacy of ACT, which can be achieved by blocking the immune checkpoint. Secondly, cytokines or cytokine receptors can be expressed by genetic engineering or added directly to adoptive cells to enhance the migration and infiltration of adoptive cells to tumor cells. Moreover, multi-antigen chimeric antigen receptors (CARs) can be designed to enhance the specific recognition of tumor cell-related antigens, and OVs can also stimulate antigen release. In addition to inserting suicide genes into adoptive cells, PROTAC technology can be used as a safety switch or degradation agent of immunosuppressive factors to enhance the safety and efficacy of adoptive cells. This article comprehensively summarizes the mechanism, current situation, and clinical application of enhanced cellular therapy, describing potential improvements to adoptive cellular therapy.
Collapse
Affiliation(s)
- Meng-Yao Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Na Zeng
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Si-Han Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Xing-Yu Zhong
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Si-Yang Ma
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Hao-Dong He
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jia Hu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
12
|
Mirbahari SN, Da Silva M, Zúñiga AIM, Kooshki Zamani N, St-Laurent G, Totonchi M, Azad T. Recent progress in combination therapy of oncolytic vaccinia virus. Front Immunol 2024; 15:1272351. [PMID: 38558795 PMCID: PMC10979700 DOI: 10.3389/fimmu.2024.1272351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
In recent years, oncolytic viruses have emerged as promising agents for treating various cancers. An oncolytic virus is a non-pathogenic virus that, due to genetic manipulation, tends to replicate in and cause lysis of cancerous cells while leaving healthy cells unaffected. Among these viruses, vaccinia virus is an attractive platform for use as an oncolytic platform due to its 190 Kb genome with a high capacity for encoding therapeutic payloads. Combining oncolytic VV therapy with other conventional cancer treatments has been shown to be synergistic and more effective than monotherapies. Additionally, OVV can be used as a vector to deliver therapeutic payloads, alone or in combination with other treatments, to increase overall efficacy. Here, we present a comprehensive analysis of preclinical and clinical studies that have evaluated the efficacy of oncolytic vaccinia viruses in cancer immunotherapy. We discuss the outcomes of these studies, including tumor regression rates, overall survival benefits, and long-term responses. Moreover, we provide insights into the challenges and limitations associated with oncolytic vaccinia virus- based therapies, including immune evasion mechanisms, potential toxicities, and the development of resistance.
Collapse
Affiliation(s)
- Seyedeh Nasim Mirbahari
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Miles Da Silva
- Department of Microbiology and Immunology, University of British Colombia, Vancouver, BC, Canada
- Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
| | - Abril Ixchel Muñoz Zúñiga
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Nika Kooshki Zamani
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Gabriel St-Laurent
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| | - Mehdi Totonchi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Taha Azad
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC, Canada
| |
Collapse
|
13
|
Zhou Y, Chen J, Bai S, Yang F, Yan R, Song Y, Yang B, Li C, Wang J. Interleukin-36gamma Mediates the In Vitro Activation of CD8 + T Cells from Patients Living with Chronic Human Immunodeficiency Virus-1 Infection. Viral Immunol 2024; 37:24-35. [PMID: 38301135 DOI: 10.1089/vim.2023.0080] [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] [Indexed: 02/03/2024] Open
Abstract
Interleukin-36 (IL-36) signaling plays an important role in promoting CD8+ T cell-mediated antitumor immune responses. The role of IL-36 signaling in CD8+ T cells that are involved in host immune responses during human immunodeficiency virus-1 (HIV-1) infection has not been characterized. Sixty-one patients living with chronic HIV-1 infection and 23 controls were enrolled in this study. The levels of IL-36 cytokine family members were measured by enzyme-linked immunosorbent assay. Purified CD8+ T cells were stimulated with recombinant IL-36gamma (1 or 10 ng/mL). The expression of inhibitory receptors, the secretion of cytotoxic molecules and interferon-gamma, and the mRNA levels of apoptosis-related ligands were assessed to evaluate the effect of IL-36gamma on CD8+ T cell function in vitro. There were no significant differences in IL-36alpha, IL-36beta, or IL-36 receptor antagonist levels between patients living with chronic HIV-1 infection and controls. Plasma IL-36gamma levels were reduced in patients living with chronic HIV-1 infection. Perforin, granzyme B, and granulysin secretion, as well as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and Fas ligand (FasL) mRNA expression, but not programmed death-1 (PD-1) or cytotoxic T lymphocyte-associated protein-4 (CTLA-4) expression was downregulated in CD8+ T cells from patients living with chronic HIV-1 infection. The addition of both 1 and 10 ng/mL IL-36gamma enhanced perforin, granzyme B, granulysin, and interferon-gamma secretion by CD8+ T cells without affecting PD-1/CTLA-4 or TRAIL/FasL mRNA expression in CD8+ T cells from patients living with chronic HIV-1 infection. The addition of 1 ng/mL IL-36gamma also promoted perforin and granzyme B secretion by HIV-1-specific CD8+ T cells from patients living with chronic HIV-1 infection. The reduced IL-36gamma levels in patients living with chronic HIV-1 infection might be insufficient for the activation of CD8+ T cells, leading to CD8+ T cell exhaustion.
Collapse
Affiliation(s)
- Yingquan Zhou
- Department of Infectious Diseases, Lanzhou Pulmonary Hospital, Lanzhou, China
| | - Jijun Chen
- Institute for STD and AIDS Prevention and Control, Lanzhou Center for Disease Control and Prevention, Lanzhou, China
| | - Shaoli Bai
- Department of Infectious Diseases, Lanzhou Pulmonary Hospital, Lanzhou, China
- Department of Internal Medicine, Gansu Province Hospital Rehabilitation Center, Lanzhou, China
| | - Fan Yang
- Department of Infectious Diseases, Lanzhou Pulmonary Hospital, Lanzhou, China
| | - Ruqing Yan
- Department of Infectious Diseases, Lanzhou Pulmonary Hospital, Lanzhou, China
| | - Yanjun Song
- Department of Infectious Diseases, Lanzhou Pulmonary Hospital, Lanzhou, China
| | - Binfa Yang
- Department of Infectious Diseases, Lanzhou Pulmonary Hospital, Lanzhou, China
| | - Chao Li
- Department of Infectious Diseases, Lanzhou Pulmonary Hospital, Lanzhou, China
| | - Jianyun Wang
- Department of Infectious Diseases, Gansu Province Hospital Rehabilitation Center, Lanzhou, China
| |
Collapse
|
14
|
Fischer B, Kübelbeck T, Kolb A, Ringen J, Waisman A, Wittmann M, Karbach S, Kölsch SM, Kramer D. IL-17A-driven psoriasis is critically dependent on IL-36 signaling. Front Immunol 2023; 14:1256133. [PMID: 38162658 PMCID: PMC10754973 DOI: 10.3389/fimmu.2023.1256133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Plaque psoriasis is an autoinflammatory and autoimmune skin disease, affecting 1-3% of the population worldwide. Previously, high levels of IL-36 family cytokines were found in psoriatic skin lesions, thereby contributing to keratinocyte hyperproliferation and infiltration of immune cells such as neutrophils. While treatment with anti-IL36 receptor (IL36R) antibodies was recently approved for generalized pustular psoriasis (GPP), it remains unclear, if targeting the IL36R might also inhibit plaque psoriasis. Here we show that antibody-mediated inhibition of IL36R is sufficient to suppress imiquimod-induced psoriasis-like skin inflammation and represses the disease's development in a model that depends on IL-17A overexpression in the skin. Importantly, treatment with anti-IL36R antibodies inhibited skin inflammation and attenuated psoriasis-associated, systemic inflammation. This is possibly due to a widespread effect of IL36R inhibition, which not only suppresses pro-inflammatory gene expression in keratinocytes, but also the activation of other immune cells such as T-cells or dendritic cells. In conclusion, we propose that inhibition of the IL-36 signaling pathway might constitute an attractive, alternative approach for treating IL-17A-driven psoriasis and psoriasis-linked comorbidities.
Collapse
Affiliation(s)
- Berenice Fischer
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Tanja Kübelbeck
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Antonia Kolb
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Julia Ringen
- Center for Cardiology- Cardiology I, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Ari Waisman
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Miriam Wittmann
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Susanne Karbach
- Center for Cardiology- Cardiology I, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK) – Partner Site Rhine-Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Stephan Marcus Kölsch
- Boehringer Ingelheim Pharma GmbH & Co. KG, Medical Affairs, Ingelheim am Rhein, Germany
| | - Daniela Kramer
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| |
Collapse
|
15
|
Tang L, Pan S, Wei X, Xu X, Wei Q. Arming CAR-T cells with cytokines and more: Innovations in the fourth-generation CAR-T development. Mol Ther 2023; 31:3146-3162. [PMID: 37803832 PMCID: PMC10638038 DOI: 10.1016/j.ymthe.2023.09.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/11/2023] [Accepted: 09/29/2023] [Indexed: 10/08/2023] Open
Abstract
Chimeric antigen receptor T cells (CAR-T) therapy has shown great potential in tumor treatment. However, many factors impair the efficacy of CAR-T therapy, such as antigenic heterogeneity and loss, limited potency and persistence, poor infiltration capacity, and a suppressive tumor microenvironment. To overcome these obstacles, recent studies have reported a new generation of CAR-T cells expressing cytokines called armored CAR-T, TRUCK-T, or the fourth-generation CAR-T. Here we summarize the strategies of arming CAR-T cells with natural or synthetic cytokine signals to enhance their anti-tumor capacity. Moreover, we summarize the advances in CAR-T cells expressing non-cytokine proteins, such as membrane receptors, antibodies, enzymes, co-stimulatory molecules, and transcriptional factors. Furthermore, we discuss several prospective strategies for armored CAR-T therapy development. Altogether, these ideas may provide new insights for the innovations of the next-generation CAR-T therapy.
Collapse
Affiliation(s)
- Lin Tang
- Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Sheng Pan
- Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xuyong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Qiang Wei
- Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
16
|
Song X, Guo L, Zhang Q, Chen W, Fan W, Lv C, Tang P, Dong Z, Ye X, Ding Q. The diagnostic value of interleukin-36 cytokines in pleural effusions of varying etiologies. Clin Chim Acta 2023; 549:117533. [PMID: 37660939 DOI: 10.1016/j.cca.2023.117533] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND The clinical management of pleural effusion (PE) poses challenges due to its diverse etiologies. The objective of this research was to investigate the concentrations of interleukin-36 (IL-36) cytokines in pleural fluid (PF) from different etiologies and assess their diagnostic efficacy in distinguishing the causes of PE. METHODS This study enrolled 89 patients with confirmed PE, comprising 11 cases classified as transudate, 24 cases as malignant pleural effusion (MPE), 24 cases as tuberculous pleural effusion (TPE), and 30 cases as parapneumonic pleural effusion (PPE). The PPE group was further subdivided into 20 cases of uncomplicated parapneumonic effusion (UPPE) and 10 cases of complicated parapneumonic effusion (CPPE)/empyema. The concentrations of IL-36 cytokines in the PF of all 89 patients were quantified by the enzyme-linked immunosorbent assay (ELISA). RESULTS IL-36α exhibited excellent diagnostic accuracy in TPE, achieving a sensitivity of 91.7 % and specificity of 83.1 %, along with a cut-off value of 435.3 pg/ml. IL-36Ra also demonstrated relatively favorable diagnostic performance in PPE, with a sensitivity of 80.0 % and specificity of 76.3 %, along with a cut-off value of 390.8 pg/ml. Multivariable logistic regression models were successfully developed for both TPE and PPE, confirming their diagnostic utility. Furthermore, the levels of IL-36Ra were notably elevated in CPPE/empyema in comparison to UPPE. Moreover, in PF, IL-36γ exhibited positive associations with both IL-36α and IL-36Ra. CONCLUSION IL-36α and IL-36Ra may serve as novel biomarkers for diagnosing TPE and PPE, respectively. The multivariate models established significantly enhance the diagnostic efficacy of both TPE and PPE. Furthermore, IL-36Ra can function as an indicator for assessing the extent of pleural inflammation. Additionally, the interaction among IL-36 cytokines in PF may contribute to their expression modulation.
Collapse
Affiliation(s)
- Xuxiang Song
- Health Science Center, Ningbo University, Ningbo 315211, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Lun Guo
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Qipan Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Weili Chen
- Health Science Center, Ningbo University, Ningbo 315211, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Wei Fan
- Health Science Center, Ningbo University, Ningbo 315211, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Chengna Lv
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Pan Tang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo 315020, China
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo Second Hospital, Ningbo 315010, China
| | - Xudeng Ye
- Department of Respiratory and Critical Care Medicine, The Cixi Integration of Traditional Chinese and Western Medicine Medical & Health Group, Ningbo 315302, China.
| | - Qunli Ding
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo 315020, China.
| |
Collapse
|
17
|
Roy S, Fitzgerald K, Lalani A, Lai CW, Kim A, Kim J, Ou P, Mirsoian A, Liu X, Ramrakhiani A, Zhao H, Zhou H, Xu H, Meisen H, Li CM, Lugt BV, Thibault S, Tinberg CE, DeVoss J, Egen J, Wu LC, Noubade R. Autonomous IL-36R signaling in neutrophils activates potent antitumor effector functions. J Clin Invest 2023; 133:e162088. [PMID: 37317970 DOI: 10.1172/jci162088] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 04/19/2023] [Indexed: 06/16/2023] Open
Abstract
While the rapid advancement of immunotherapies has revolutionized cancer treatment, only a small fraction of patients derive clinical benefit. Eradication of large, established tumors appears to depend on engaging and activating both innate and adaptive immune system components to mount a rigorous and comprehensive immune response. Identifying such agents is a high unmet medical need, because they are sparse in the therapeutic landscape of cancer treatment. Here, we report that IL-36 cytokine can engage both innate and adaptive immunity to remodel an immune-suppressive tumor microenvironment (TME) and mediate potent antitumor immune responses via signaling in host hematopoietic cells. Mechanistically, IL-36 signaling modulates neutrophils in a cell-intrinsic manner to greatly enhance not only their ability to directly kill tumor cells but also promote T and NK cell responses. Thus, while poor prognostic outcomes are typically associated with neutrophil enrichment in the TME, our results highlight the pleiotropic effects of IL-36 and its therapeutic potential to modify tumor-infiltrating neutrophils into potent effector cells and engage both the innate and adaptive immune system to achieve durable antitumor responses in solid tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Haoda Xu
- Therapeutic Discovery, Amgen, South San Francisco, California, USA
| | | | | | | | - Steve Thibault
- Therapeutic Discovery, Amgen, South San Francisco, California, USA
| | | | | | | | | | | |
Collapse
|
18
|
Li M, Jiang W, Wang Z, Lu Y, Zhang J. New insights on IL‑36 in intestinal inflammation and colorectal cancer (Review). Exp Ther Med 2023; 25:275. [PMID: 37206554 PMCID: PMC10189745 DOI: 10.3892/etm.2023.11974] [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: 12/26/2022] [Accepted: 03/21/2023] [Indexed: 05/21/2023] Open
Abstract
Interleukin (IL)-36 is a member of the IL-1 superfamily, which includes three receptor agonists and one antagonist and exhibits a familial feature of inflammatory regulation. Distributed among various tissues, such as the skin, lung, gut and joints, the mechanism of IL-36 has been most completely investigated in the skin and has been used in clinical treatment of generalized pustular psoriasis. Meanwhile, the role of IL-36 in the intestine has also been under scrutiny and has been shown to be involved in the regulation of various intestinal diseases. Inflammatory bowel disease and colorectal cancer are the most predominant inflammatory and neoplastic diseases of the intestine, and multiple studies have identified a complex role for IL-36 in both of them. Indeed, inhibiting IL-36 signaling is currently regarded as a promising therapeutic approach. Therefore, the present review briefly describes the composition and expression of IL-36 and focuses on the role of IL-36 in intestinal inflammation and colorectal cancer. The targeted therapies that are currently being developed for the IL-36 receptor are also discussed.
Collapse
Affiliation(s)
- Minghui Li
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Wei Jiang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Zehui Wang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yihan Lu
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Jun Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
- Correspondence to: Dr Jun Zhang, Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, 8th Floor, 8th Building, 68 Changle Road, Qinhuai, Nanjing, Jiangsu 210006, P.R. China
| |
Collapse
|
19
|
Liu L, Ju M, Hu Y, Luan C, Zhang J, Chen K. Genome-wide DNA methylation and transcription analysis in psoriatic epidermis. Epigenomics 2023; 15:209-226. [PMID: 37158398 DOI: 10.2217/epi-2022-0458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
Aim: To identify DNA methylation and transcription biomarkers in the psoriatic epidermis. Materials & methods: Gene transcription and DNA methylation datasets of psoriatic epidermal tissue were obtained from the Gene Expression Omnibus. Machine learning algorithm analysis and weighted gene coexpression network analysis were carried out to screen hub genes. Results: Differentially methylated and expressed genes were identified in the psoriatic epidermis. Six hub genes were selected - GZMB, CRIP1, S100A12, ISG15, CRABP2 and VNN1 - whose transcript levels showed a significant correlation with Psoriasis Area and Severity Index scores and immune infiltration. Conclusion: Psoriatic epidermis is primarily in a hypermethylated status. Epidermis-specific hub differentially methylated and expressed genes are potential biomarkers to help judge the condition of psoriasis.
Collapse
Affiliation(s)
- Lingxi Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Mei Ju
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Yu Hu
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Chao Luan
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Jiaan Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Kun Chen
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| |
Collapse
|
20
|
Zeng F, Wang X, Hu Y, Wang Z, Li Y, Hu J, Yu J, Zhou P, Teng X, Zhou H, Zheng H, Zhao F, Gu L, Yue C, Chen S, Cheng J, Hao Y, Zhao Q, Zhang C, Zou S, Hu Z, Wei X, Liu X, Li G, Huang N, Wu W, Zhou Y, Li W, Cui K, Li J. Interleukin-37 promotes DMBA/TPA skin cancer through SIGIRR-mediated inhibition of glycolysis in CD103 +DC cells. MedComm (Beijing) 2023; 4:e229. [PMID: 36891351 PMCID: PMC9986080 DOI: 10.1002/mco2.229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 03/07/2023] Open
Abstract
Interleukin 37 (IL-37), a member of the IL-1 family, is considered a suppressor of innate and adaptive immunity and, hence is a regulator of tumor immunity. However, the specific molecular mechanism and role of IL-37 in skin cancer remain unclear. Here, we report that IL-37b-transgenic mice (IL-37tg) treated with the carcinogenic 7,12-dimethylbenzoanthracene (DMBA)/12-o-tetradecylphorbol-13-acetate (TPA) exhibited enhanced skin cancer and increased tumor burden in the skin by inhibiting the function of CD103+ dendritic cells (DCs). Notably, IL-37 induced rapid phosphorylation of adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), and via single immunoglobulin IL-1-related receptor (SIGIRR), inhibited the long-term Akt activation. Specifically, by affecting the SIGIRR-AMPK-Akt signaling axis, which is related to the regulation of glycolysis in CD103+DCs, IL-37 inhibited their anti-tumor function. Our results show that a marked correlation between the CD103+DC signature (IRF8, FMS-like tyrosine kinase 3 ligand, CLEC9A, CLNK, XCR1, BATF3, and ZBTB46) and chemokines C-X-C motif chemokine ligand 9, CXCL10, and CD8A in a mouse model with DMBA/TPA-induced skin cancer. In a word, our results highlight that IL-37 as an inhibitor of tumor immune surveillance through modulating CD103+DCs and establishing an important link between metabolism and immunity as a therapeutic target for skin cancer.
Collapse
Affiliation(s)
- Fan‐lian Zeng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Xiao‐yan Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Ya‐wen Hu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Zhen Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
- Department of Liver Surgery and Liver TransplantationWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
- Laboratory of Liver SurgeryWest China HospitalSichuan UniversityChengduChina
| | - Ya Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Jing Hu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Jia‐dong Yu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Pei Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Xiu Teng
- Laboratory of Human Disease and ImmunotherapiesWest China HospitalSichuan UniversityChengduChina
| | - Hong Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Hua‐ping Zheng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Fu‐lei Zhao
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Lin‐na Gu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Cheng‐cheng Yue
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Shu‐wen Chen
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Juan Cheng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Yan Hao
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Qi‐xiang Zhao
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Chen Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Song Zou
- Department of CardiologyWest China HospitalSichuan UniversityChengduChina
| | - Zhong‐lan Hu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Xiao‐qiong Wei
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Xiao Liu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Guo‐lin Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Nong‐yu Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Wen‐ling Wu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Yi‐fan Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Wei Li
- Department of DermatovenereologyWest China HospitalSichuan UniversityChengduChina
| | - Kaijun Cui
- Department of CardiologyWest China HospitalSichuan UniversityChengduChina
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalWest China Medical SchoolSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| |
Collapse
|
21
|
Baker KJ, Buskiewicz E, Finucane M, Chelliah A, Burke L, Houston A, Brint E. IL-36 expression is increased in NSCLC with IL-36 stimulation of lung cancer cells promoting a pro-tumorigenic phenotype. Cytokine 2023; 165:156170. [PMID: 36931148 DOI: 10.1016/j.cyto.2023.156170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/17/2023]
Abstract
The IL-36 cytokines are a recently described subset of the IL-1 family of cytokines, and have been shown to play a role in the pathogenesis of respiratory diseases such as asthma and COPD. Given the common aetiological links between COPD and lung cancer development, as well as the involvement of other IL-1 family members in lung tumorigenesis, the aim of this work was to investigate the role of IL-36 cytokines in the pathogenesis of lung cancer. In this study we demonstrate that expression of IL-36 cytokines and receptor mRNA and protein are significantly increased in lung cancer tissue compared to adjacent non-tumour tissue. In vitro assays showed that stimulation of two lung cancer cell lines, SKMES-1 human squamous cell and LLC murine lung cancer, with IL-36R agonists resulted in increased cellular migration and proliferation. All IL-36 cytokines induced the expression of pro-inflammatory chemokines in both lung cancer cell lines with synergistic effects identified upon co-stimulation of cells with IL-17, IL-22 and TNFα. Furthermore, we report that IL-36 cytokines induce protein expression of the immune checkpoint inhibitor protein PD-L1 on lung cancer cells. Taken together, this data indicates that targeting IL-36R signalling may be a useful targeted therapy for lung cancer patients with IL-36R+ cancer cells.
Collapse
Affiliation(s)
- Kevin James Baker
- Department of Pathology, UCC, Cork, Ireland; Department of Medicine, UCC, Cork, Ireland
| | | | - Méabh Finucane
- Department of Pathology, UCC, Cork, Ireland; Department of Medicine, UCC, Cork, Ireland
| | | | - Louise Burke
- Department of Pathology, UCC, Cork, Ireland; Dept. Of Pathology CUH, Ireland
| | - Aileen Houston
- Department of Medicine, UCC, Cork, Ireland; APC Microbiome Ireland, UCC, Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, UCC, Cork, Ireland; APC Microbiome Ireland, UCC, Cork, Ireland.
| |
Collapse
|
22
|
Baker KJ, Brint E, Houston A. Transcriptomic and functional analyses reveal a tumour-promoting role for the IL-36 receptor in colon cancer and crosstalk between IL-36 signalling and the IL-17/ IL-23 axis. Br J Cancer 2023; 128:735-747. [PMID: 36482185 PMCID: PMC9977920 DOI: 10.1038/s41416-022-02083-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The interleukin (IL)-36 cytokines are a sub-family of the IL-1 family which are becoming increasingly implicated in the pathogenesis of inflammatory diseases and malignancies. Initial studies of IL-36 signalling in tumorigenesis identified an immune-mediated anti-tumorigenic function for these cytokines. However, more recent studies have shown IL-36 cytokines also contribute to the pathogenesis of lung and colorectal cancer (CRC). METHODS The aim of this study was to investigate IL-36 expression in CRC using transcriptomic datasets and software such as several R packages, Cytoscape, GEO2R and AnalyzeR. Validation of results was completed by qRT-PCR on both cell lines and a patient cohort. Cellular proliferation was assessed by flow cytometry and resazurin reduction. RESULTS We demonstrate that IL-36 gene expression increases with CRC development. Decreased tumoral IL-36 receptor expression was shown to be associated with improved patient outcome. Our differential gene expression analysis revealed a novel role for the IL-36/IL-17/IL-23 axis, with these findings validated using patient-derived samples and cell lines. IL-36γ, together with either IL-17a or IL-22, was able to synergistically induce different genes involved in the IL-17/IL-23 axis in CRC cells and additively induce colon cancer cell proliferation. CONCLUSIONS Collectively, this data support a pro-tumorigenic role for IL-36 signalling in colon cancer, with the IL-17/IL-23 axis influential in IL-36-mediated colon tumorigenesis.
Collapse
Affiliation(s)
- Kevin James Baker
- Department of Pathology, University College Cork, Cork, Ireland.,Department of Medicine, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, University College Cork, Cork, Ireland. .,APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Aileen Houston
- Department of Medicine, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
23
|
Mahmoudvand S, Shokri S, Nakhaie M, Jalilian FA, Mehri-Ghahfarrokhi A, Yarani R, Shojaeian A. Small extracellular vesicles as key players in cancer development caused by human oncogenic viruses. Infect Agent Cancer 2022; 17:58. [DOI: 10.1186/s13027-022-00471-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Abstract
Background
Exosomes are the smallest group of extracellular vesicles in size from 30 to 150 nm, surrounded by a lipid bilayer membrane, and originate from multivesicular bodies secreted by different types of cells, such as virus-infected cells. The critical role of exosomes is information transfer among cells, representing a unique way for intercellular communication via a load of many kinds of molecules, including various signaling proteins and nucleic acids. In this review, we aimed to comprehensively investigate the role of exosomes in promoting human oncogenic viruses-associated cancers.
Methods
Our search was conducted for published researches between 2000 and 2022 by using several international databases includeing Scopus, PubMed, and Web of Science as well as Google scholar. We also reviewed additional evidence from relevant published articles.
Results
It has been shown that exosomes can create the conditions for viral spread in viral infections. Exosome secretion in a human tumor virus can switch on the cell signaling pathways by transferring exosome-encapsulated molecules, including viral oncoproteins, signal transduction molecules, and virus-encoded miRNAs, into various cells.
Conclusion
Given the role of exosomes in viruses-associated cancers, they can also be considered as molecular targets in diagnosis and treatment.
Collapse
|
24
|
Tong Y, Cao Y, Jin T, Huang Z, He Q, Mao M. Role of Interleukin-1 family in bone metastasis of prostate cancer. Front Oncol 2022; 12:951167. [PMID: 36237303 PMCID: PMC9552844 DOI: 10.3389/fonc.2022.951167] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022] Open
Abstract
Prostate cancer (PCa) is one of the most fatal diseases in male patients with high bone metastatic potential. Bone metastasis severely shortens overall survival and brings skeletal-related events (SREs) which reduces the life quality of patients, and this situation is currently regarded as irreversible and incurable. The progression and metastasis of PCa are found to be closely associated with inflammatory cytokines and chemokines. As pivotal members of inflammatory cytokines, Interleukin-1 (IL-1) family plays a crucial role in this process. Elevated expression of IL-1 family was detected in PCa patients with bone metastasis, and accumulating evidences proved that IL-1 family could exert vital effects on the progression and bone metastasis of many cancers, while some members have dual effects. In this review, we discuss the role of IL-1 family in the bone metastasis of PCa. Furthermore, we demonstrate that many members of IL-1 family could act as pivotal biomarkers to predict the clinical stage and prognosis of PCa patients. More importantly, we have elucidated the role of IL-1 family in the bone metastasis of PCa, which could provide potential targets for the treatment of PCa bone metastasis and probable directions for future research.
Collapse
Affiliation(s)
- Yuanhao Tong
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Yinghao Cao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianzhe Jin
- Department of Gynecologic Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhengwei Huang
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Qinyuan He
- Organization Department, Suzhou Traditional Chinese Medicine Hospital, Suzhou, China
| | - Min Mao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Min Mao,
| |
Collapse
|
25
|
Chen L, Huang H, Zheng X, Li Y, Chen J, Tan B, Liu Y, Sun R, Xu B, Yang M, Li B, Wu C, Lu B, Jiang J. IL1R2 increases regulatory T cell population in the tumor microenvironment by enhancing MHC-II expression on cancer-associated fibroblasts. J Immunother Cancer 2022. [PMCID: PMC9438093 DOI: 10.1136/jitc-2022-004585] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Regulatory T cells (Treg) are an integral part of the tumor immune tolerance. Carcinoma-associated fibroblasts (CAFs) is a pivotal driver for accumulation of Treg cells in the tumor microenvironment (TME). The molecular nature underpinning Treg cells and CAFs coupling needs to be further defined. Methods The Il1r2flox/floxFoxp3Cre mice were generated to establish the conditional knock-out of Il1r2 in Foxp3+ Tregs in vivo. Using the MC38 tumor model, we evaluated the antitumor efficacy of immune checkpoint inhibitors (ICIs) and further analyzed the immune profiling of the TME by multicolor flow cytometry. Single-cell RNA sequencing of the whole tumor tissues, TCR repertoire analysis of sorted CD3+ TILs were also performed. Results We showed that IL1 receptor 2 (IL1R2), a decoy receptor that neutralizes IL1, was highly expressed in Treg cells in the TME. In addition, we found that Il1r1 was largely expressed in the CAFs, suggesting IL1R2 plays a role in modulating crosstalk between Tregs and CAFs. We further demonstrated that Il1r2 deficiency in Treg cells led to greater antitumor efficacy of ICI, decreased Tregs and increased CD8+ T cells in the TME, as well as reduced levels of T cell dysfunction. Mechanistically, we showed that IL1 inhibited major histocompatibility complex class II (MHC-II) expression on fibroblasts and Treg-specific Il1r2 deletion led to a decrease in genes associated with MHC-II antigen presentation in CAFs. Conclusions Our study established a critical role of IL1 signaling in inhibiting Treg-mediated tumor immune suppression through downregulating MHC-II antigen presentation in CAFs.
Collapse
Affiliation(s)
- Lujun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Hao Huang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Yuan Li
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Junjun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Bo Tan
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Yingting Liu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Runzi Sun
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Bin Xu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Min Yang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Department of Nephrology, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changping Wu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jingting Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
- Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, China
| |
Collapse
|
26
|
Okuma A, Ishida Y, Kawara T, Hisada S, Araki S. Secretory co-factors in next-generation cellular therapies for cancer. Front Immunol 2022; 13:907022. [PMID: 36059449 PMCID: PMC9433659 DOI: 10.3389/fimmu.2022.907022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Since chimeric antigen receptor (CAR) T-cell therapies for hematologic malignancies were approved by the U.S. Food and Drug Administration, numerous "next-generation" CAR T cells have been developed to improve their safety, efficacy, and applicability. Although some of these novel therapeutic strategies are promising, it remains difficult to apply these therapies to solid tumors and to control adverse effects, such as cytokine release syndrome and neurotoxicity. CAR T cells are generated using highly scalable genetic engineering techniques. One of the major strategies for producing next-generation CAR T cells involves the integration of useful co-factor(s) into the artificial genetic design of the CAR gene, resulting in next-generation CAR T cells that express both CAR and the co-factor(s). Many soluble co-factors have been reported for CAR T cells and their therapeutic effects and toxicity have been tested by systemic injection; therefore, CAR T cells harnessing secretory co-factors could be close to clinical application. Here, we review the various secretory co-factors that have been reported to improve the therapeutic efficacy of CAR T cells and ameliorate adverse events. In addition, we discuss the different co-factor expression systems that have been used to optimize their beneficial effects. Altogether, we demonstrate that combining CAR T cells with secretory co-factors will lead to next-generation CAR T-cell therapies that can be used against broader types of cancers and might provide advanced tools for more complicated synthetic immunotherapies.
Collapse
Affiliation(s)
- Atsushi Okuma
- Center for Exploratory Research, Research and Development Group, Hitachi Ltd., Kobe, Japan
| | | | | | | | | |
Collapse
|
27
|
Poudel M, Bhattarai PY, Shrestha P, Choi HS. Regulation of Interleukin-36γ/IL-36R Signaling Axis by PIN1 in Epithelial Cell Transformation and Breast Tumorigenesis. Cancers (Basel) 2022; 14:cancers14153654. [PMID: 35954317 PMCID: PMC9367291 DOI: 10.3390/cancers14153654] [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: 06/13/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Members of the interleukin (IL)-1 cytokine family exhibit dual functions in the regulation of inflammation and cancer. Recent studies have shown the critical role of IL-36γ, the newly identified IL-1 family member, in the regulation of cellular processes implicated in the progression of cancer. Therefore, the underlying mechanism of IL-36γ in tumor development is of considerable interest. Here, we identified the pivotal role of IL-36γ in the proliferation of breast cancer cells. Consistently, IL-36γ was found to promote epithelial cell transformation via the activation of c-Fos, c-Jun, and AP-1 transcription factors, followed by the IL36R-mediated MEK/ERK and JNK/c-Jun cascades. Furthermore, our findings demonstrate the critical role of PIN1 in the regulation of IL-36γ-induced mammary gland tumorigenesis. Abstract Given the increasing recognition of the relationship between IL-1 cytokines, inflammation, and cancer, the significance of distinct members of the IL-1 cytokine family in the etiology of cancer has been widely researched. In the present study, we investigated the underlying mechanism of the IL-36γ/IL-36R axis during breast cancer progression, which has not yet been elucidated. Initially, we determined the effects of IL-36γ on the proliferation and epithelial cell transformation of JB6 Cl41 mouse epidermal and MCF7 human breast cancer cells using BrdU incorporation and anchorage-independent growth assays. We found that treatment with IL-36γ increased the proliferation and colony formation of JB6 Cl41 and MCF7 cells. Analysis of the mechanism underlying the neoplastic cell transformation revealed that IL-36γ induced IL-36R-mediated phosphorylation of MEK1/2, ERK1/2, JNK1/2, and c-Jun, resulting in increased c-Fos, c-Jun, and AP-1 activities in JB6 Cl41 and MCF7 cells. Furthermore, the IL-36γ-induced tumorigenic capacity of MCF7 cells was considerably enhanced by PIN1, following MEK/ERK and JNK/c-Jun signaling. Interestingly, blocking PIN1 activity using juglone suppressed the IL-36γ-induced increase in the anchorage-independent growth of 4T1 metastatic mouse breast cancer cells. Finally, in a syngeneic mouse model, IL-36γ-induced tumor growth in the breast mammary gland was significantly inhibited following PIN1 knockout.
Collapse
Affiliation(s)
| | | | | | - Hong Seok Choi
- Correspondence: ; Tel.: +82-622306379; Fax: +82-622225414
| |
Collapse
|
28
|
Safarzadeh Kozani P, Safarzadeh Kozani P, Ahmadi Najafabadi M, Yousefi F, Mirarefin SMJ, Rahbarizadeh F. Recent Advances in Solid Tumor CAR-T Cell Therapy: Driving Tumor Cells From Hero to Zero? Front Immunol 2022; 13:795164. [PMID: 35634281 PMCID: PMC9130586 DOI: 10.3389/fimmu.2022.795164] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/04/2022] [Indexed: 12/21/2022] Open
Abstract
Chimeric antigen receptor T-cells (CAR-Ts) are known as revolutionary living drugs that have turned the tables of conventional cancer treatments in certain hematologic malignancies such as B-cell acute lymphoblastic leukemia (B-ALL) and diffuse large B-cell lymphoma (DLBCL) by achieving US Food and Drug Administration (FDA) approval based on their successful clinical outcomes. However, this type of therapy has not seen the light of victory in the fight against solid tumors because of various restricting caveats including heterogeneous tumor antigen expression and the immunosuppressive tumor microenvironments (TME) that negatively affect the tumor-site accessibility, infiltration, stimulation, activation, and persistence of CAR-Ts. In this review, we explore strategic twists including boosting vaccines and designing implementations that can support CAR-T expansion, proliferation, and tumoricidal capacity. We also step further by underscoring novel strategies for triggering endogenous antitumor responses and overcoming the limitation of poor CAR-T tumor-tissue infiltration and the lack of definitive tumor-specific antigens. Ultimately, we highlight how these approaches can address the mentioned arduous hurdles.
Collapse
Affiliation(s)
- Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Milad Ahmadi Najafabadi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Yousefi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
29
|
Wu KS, Jian TY, Sung SY, Hsieh CL, Huang MH, Fang CL, Wong TT, Lin YL. Enrichment of Tumor-Infiltrating B Cells in Group 4 Medulloblastoma in Children. Int J Mol Sci 2022; 23:ijms23095287. [PMID: 35563678 PMCID: PMC9101625 DOI: 10.3390/ijms23095287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 12/21/2022] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children. It is classified into core molecular subgroups (wingless activated (WNT), sonic hedgehog activated (SHH), Group 3 (G3), and Group 4 (G4)). In this study, we analyzed the tumor-infiltrating immune cells and cytokine profiles of 70 MB patients in Taiwan using transcriptome data. In parallel, immune cell composition in tumors from the SickKids cohort dataset was also analyzed to confirm the findings. The clinical cohort data showed the WNT and G4 MB patients had lower recurrence rates and better 5-year relapse-free survival (RFP) compared with the SHH and G3 MB patients, among the four subgroups of MB. We found tumor-infiltrating B cells (TIL-Bs) enriched in the G4 subgroups in the Taiwanese MB patients and the SickKids cohort dataset. In the G4 subgroups, the patients with a high level of TIL-Bs had better 5-year overall survival. Mast cells presented in G4 MB tumors were positively correlated with TIL-Bs. Higher levels of CXCL13, IL-36γ, and CCL27 were found compared to other subgroups or normal brains. These three cytokines, B cells and mast cells contributed to the unique immune microenvironment in G4 MB tumors. Therefore, B-cell enrichment is a G4-subgroup-specific immune signature and the presence of B cells may be an indicator of a better prognosis in G4 MB patients.
Collapse
Affiliation(s)
- Kuo-Sheng Wu
- Graduate Institute of Clinical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (K.-S.W.); (S.-Y.S.)
| | - Ting-Yan Jian
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Shian-Ying Sung
- Graduate Institute of Clinical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (K.-S.W.); (S.-Y.S.)
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Office of Human Research, Taipei Medical University, Taipei 110, Taiwan
- TMU-Research Center of Urology and Kidney, Taipei Medical University, Taipei 110, Taiwan
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
- Laboratory of Translational Medicine, Development Center for Biotechnology, Taipei 115, Taiwan
| | - Man-Hsu Huang
- Department of Pathology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan;
| | - Chia-Lang Fang
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
| | - Tai-Tong Wong
- Graduate Institute of Clinical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (K.-S.W.); (S.-Y.S.)
- Pediatric Brain Tumor Program, Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan
- Neuroscience Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Taipei Neuroscience Institute, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (T.-T.W.); (Y.-L.L.)
| | - Yu-Ling Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan;
- Correspondence: (T.-T.W.); (Y.-L.L.)
| |
Collapse
|
30
|
Le N, Luk I, Chisanga D, Shi W, Pang L, Scholz G, Mariadason J, Ernst M, Huynh J. IL-36G promotes cancer-cell intrinsic hallmarks in human gastric cancer cells. Cytokine 2022; 155:155887. [PMID: 35512531 DOI: 10.1016/j.cyto.2022.155887] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/24/2022] [Accepted: 04/07/2022] [Indexed: 11/28/2022]
Abstract
Interleukin-36 gamma (IL-36G) is a member of the IL-36 subfamily of cytokines and acts as a potent driver of inflammation. IL-36G has been extensively characterized in the pathogenesis of psoriasis and has been recently described to play roles in wound healing particularly in the gastrointestinal tract. However, the effects of IL-36G during cancer development including gastric cancer remain unexplored. Here, we show that IL-36G induced ERK1/2 activation in AGS, MKN1 and MKN45 human gastric cancer cell lines. Moreover, IL-36G induced colony formation, migration and invasion of these gastric cancer cell lines that was inhibited by the natural antagonist, IL-36 receptor antagonist (RA). Interrogation of TCGA stomach adenocarcinoma patient datasets revealed highly elevated IL-36G gene expression in human gastric cancer compared to normal tissue independent of tumor stage, and high IL-36G expression corresponded with poorer patient survival. Collectively, our results indicate for the first time that IL-36G supports a neoplastic phenotype in human gastric cancer cells.
Collapse
Affiliation(s)
- Ngan Le
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - Ian Luk
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia; School of Computing and Information Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia; School of Computing and Information Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Lokman Pang
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - Glen Scholz
- Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, VIC, Australia
| | - John Mariadason
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| |
Collapse
|
31
|
Sun R, Gao DS, Shoush J, Lu B. The IL-1 family in tumorigenesis and antitumor immunity. Semin Cancer Biol 2022; 86:280-295. [DOI: 10.1016/j.semcancer.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
|
32
|
Yang W, Dong H, Wang P, Xu Z, Xian J, Chen J, Wu H, Lou Y, Lin D, Zhong B. IL-36γ and IL-36Ra Reciprocally Regulate Colon Inflammation and Tumorigenesis by Modulating the Cell-Matrix Adhesion Network and Wnt Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103035. [PMID: 35119210 PMCID: PMC8981487 DOI: 10.1002/advs.202103035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/14/2021] [Indexed: 05/07/2023]
Abstract
Inflammatory bowel disease and colorectal cancer are associated with dysregulation of cytokine networks. However, it is challenging to target cytokines for effective intervention because of the overlapping functions and unpredictable interactions of cytokines in such diverse networks. Here, it is shown that IL-36γ and IL-36Ra, an agonist and an antagonist for IL-36R signaling respectively, reciprocally regulate the experimental colitis and the colon cancer development in mice. Knockout or neutralization of IL-36γ alleviates dextran sulfate sodium (DSS)-induced colitis and inhibits colon cancer development, whereas knockout of IL-36Ra exacerbates DSS-induced colitis and promotes colonic tumorigenesis in multiple colon cancer models in mice. Mechanistically, IL-36γ upregulates extracellular matrix and cell-matrix adhesion molecules and facilitates Wnt signaling, which is mitigated by IL-36Ra or IL-36γ neutralizing antibody. Consistently, IL-36γ levels are positively correlated with extracellular matrix levels and β-catenin levels in human colorectal tumor biopsies. These findings suggest the critical role of IL-36γ and IL-36Ra in gut inflammation and tumorigenesis and indicate that targeting the IL-36γ/IL-36Ra signal balance provides potential therapeutic strategy for inflammatory bowel disease and gastrointestinal cancers.
Collapse
Affiliation(s)
- Wei Yang
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Department of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
- Wuhan Research Center for Infectious Diseases and CancerChinese Academy of Medical SciencesWuhan430071China
| | - Hong‐Peng Dong
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Department of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Peng Wang
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Department of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Zhi‐Gao Xu
- Institute of Hepatobiliary Diseases and Transplant CenterZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Jiahuan Xian
- Yurogen Biosystems LLC (Wuhan)666 Gaoxin Avenue, Building C6, Donghu DistrictWuhan430064China
| | - Jiachen Chen
- Yurogen Biosystems LLC (Wuhan)666 Gaoxin Avenue, Building C6, Donghu DistrictWuhan430064China
| | - Hai Wu
- Yurogen Biosystems LLC (Wuhan)666 Gaoxin Avenue, Building C6, Donghu DistrictWuhan430064China
| | - Yang Lou
- Yurogen Biosystems LLC (Wuhan)666 Gaoxin Avenue, Building C6, Donghu DistrictWuhan430064China
| | - Dandan Lin
- Cancer CenterRenmin Hospital of Wuhan UniversityWuhan430061China
| | - Bo Zhong
- Department of Gastrointestinal SurgeryMedical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Department of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
- Wuhan Research Center for Infectious Diseases and CancerChinese Academy of Medical SciencesWuhan430071China
| |
Collapse
|
33
|
IL-36 signalling enhances a pro-tumorigenic phenotype in colon cancer cells with cancer cell growth restricted by administration of the IL-36R antagonist. Oncogene 2022; 41:2672-2684. [PMID: 35365751 PMCID: PMC9076531 DOI: 10.1038/s41388-022-02281-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 11/08/2022]
Abstract
The IL-36 cytokines are a recently described subset of the IL-1 family of cytokines, shown to play a role in the pathogenesis of intestinal diseases such as Inflammatory Bowel Disease (IBD). Given the link between IBD and colitis -associated cancer, as well as the involvement of other IL-1 family members in intestinal tumorigenesis, the aim of this work was to investigate whether IL-36 cytokines play a role in the pathogenesis of colon cancer. Whilst research to date has focused on the role of IL-36 family members in augmenting the immune response to induce tumour rejection, very little remains known about IL-36R signalling in tumour cells in this context. In this study we demonstrate that expression of IL-36 family member mRNA and protein are significantly increased in colorectal cancer tissue compared to adjacent non-tumour. In vitro assays showed stimulation of colon cancer cell lines with IL-36R agonists resulted in the activation of the pro-tumorigenic phenotypes of increased cellular migration, invasion and proliferation in both 2D and 3D models. In addition, the IL-36 cytokines induced strong expression of pro-inflammatory chemokines in both human and murine cell lines. Intraperitoneal injection of IL-36Ra significantly reduced tumour burden using the subcutaneous CT26 tumour model in syngeneic Balb/mice, and this was associated with a decrease in Ki-67 expression by tumour cells in the IL-36Ra- treated group relative to untreated, suggesting the inhibition of the pro-proliferative signalling of IL-36 agonists resulted in the decreased tumour size. Moreover, colon cancer cells lacking the IL-36R also showed reduced tumour growth and reduced Ki-67 expression in vivo. Taken together, this data suggests that targeting IL-36R signalling may be a useful targeted therapy for colorectal cancer patients with IL-36R+ tumour cells.
Collapse
|
34
|
Wang X, Liang Y, Wang H, Zhang B, Soong L, Cai J, Yi P, Fan X, Sun J. The Protective Role of IL-36/IL-36R Signal in Con A-Induced Acute Hepatitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:861-869. [PMID: 35046104 PMCID: PMC8830780 DOI: 10.4049/jimmunol.2100481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 12/07/2021] [Indexed: 12/23/2022]
Abstract
The IL-36 family, including IL-36α, IL-36β, IL-36γ, and IL-36R antagonist, belong to the IL-1 superfamily. It was reported that IL-36 plays a role in immune diseases. However, it remains unclear how IL-36 regulates inflammation. To determine the role of IL-36/IL-36R signaling pathways, we established an acute hepatitis mouse model (C57BL/6) by i.v. injection of the plant lectin Con A. We found that the levels of IL-36 were increased in the liver after Con A injection. Our results demonstrated the infiltrated neutrophils, but not the hepatocytes, were the main source of IL-36 in the liver. Using the IL-36R-/- mouse model (H-2b), we surprisingly found that the absence of IL-36 signals led to aggravated liver injury, as evidenced by increased mortality, elevated serum alanine aminotransferase and aspartate aminotransferase levels, and severe liver pathological changes. Further investigations demonstrated that a lack of IL-36 signaling induced intrahepatic activation of CD4+ and CD8+ T lymphocytes and increased the production of inflammatory cytokines. In addition, IL-36R-/- mice had reduced T regulatory cell numbers and chemokines in the liver. Together, our results from the mouse model suggested a vital role of IL-36 in regulating T cell function and homeostasis during liver inflammation.
Collapse
Affiliation(s)
- Xiaofang Wang
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX
| | - Hui Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX
| | - Biao Zhang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
- Department of Histology and Embryology, Guangdong Medical University, Zhanjiang, Guangdong, China; and
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX
- Department of Pathology, University of Texas Medical Branch, Galveston, TX
| | - Jiyang Cai
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX
| | - Panpan Yi
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, Hunan, China;
| | - Xuegong Fan
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, Hunan, China;
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX;
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX
- Department of Pathology, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
35
|
The role of IL-36 subfamily in intestinal disease. Biochem Soc Trans 2022; 50:223-230. [PMID: 35166319 DOI: 10.1042/bst20211264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/25/2021] [Accepted: 01/06/2022] [Indexed: 11/17/2022]
Abstract
Interleukin (IL)-36 is a subfamily, of the IL-1 super-family and includes IL-36α, IL-36β, IL-36γ, IL-38 and IL-36Ra. IL-36 cytokines are involved in the pathology of multiple tissues, including skin, lung, oral cavity, intestine, kidneys and joints. Recent studies suggest that IL-36 signaling regulates autoimmune disease in addition to antibacterial and antiviral responses. Most research has focused on IL-36 in skin diseases such as psoriasis, however, studies on intestinal diseases are also underway. This review outlines what is known about the bioactivity of the IL-36 subfamily and its role in the pathogenesis of intestinal diseases such as inflammatory bowel disease, colorectal cancer, gut dysbacteriosis and infection, and proposes that IL-36 may be a target for novel therapeutic strategies to prevent or treat intestinal diseases.
Collapse
|
36
|
Song P, Wusiman D, Li F, Wu X, Guo L, Li W, Gao S, He J. Pan-cancer analysis combined with experiments explores the oncogenic role of spindle apparatus coiled-coil protein 1 (SPDL1). Cancer Cell Int 2022; 22:49. [PMID: 35093072 PMCID: PMC8801078 DOI: 10.1186/s12935-022-02461-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/10/2022] [Indexed: 11/10/2022] Open
Abstract
Background The function of spindle apparatus coiled-coil protein 1 (SPDL1) as a cancer-promoting gene has been reported in a number of studies. However, the pan-cancer analysis of SPDL1 is still lacking. Here, we performed this pan-cancer analysis to evaluate the expression and prognostic value of SPDL1 and gain insights into the association between SPDL1 and immune infiltration. Methods In this study, based on the datasets of The cancer genome atlas (TCGA), Gene expression omnibus (GEO), The Genotype-Tissue Expression (GTEx) and Clinical Proteomic Tumor Analysis Consortium (CPTAC), we used R4.1.0 software and the online tools, including TIMER2.0, GEPIA2, cBioPortal, Modbase, UALCAN, MEXPRESS, STRING, Ensembl, NCBI, HPA, Oncomine, PhosphoNET and the Kaplan-Meier plotter, to explore the potential oncogenic roles of SPDL1. The expression of SPDL1 was also further verified by immunohistochemistry (IHC) in lung adenocarcinoma (LUAD) tissues. Results SPDL1 was overexpressed in most tumors compared with adjacent normal tissues, and SPDL1 expression was significantly correlated with the prognosis in most tumor types. The main type of genetic mutation of SPDL1 was missense mutation and the frequency of R318Q/W mutation was highest (4/119). The expression of SPDL1 was closely associated with genomic instability. The SPDL1 phosphorylation levels in S555 was enhanced in ovarian cancer. The SPDL1 expression was positively correlated with the immune infiltration of CD8+ T-cells and cancer-associated fibroblasts (CAFs) in most of the tumor types. Nuclear division, organelle fission and chromosome segregation were involved in the functional mechanisms of SPDL1. Conclusions These findings suggested that SPDL1 might serve as a biomarker for poor prognosis and immune infiltration in cancers, shedding new light on therapeutics of cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02461-w.
Collapse
|
37
|
Peñaloza HF, van der Geest R, Ybe JA, Standiford TJ, Lee JS. Interleukin-36 Cytokines in Infectious and Non-Infectious Lung Diseases. Front Immunol 2021; 12:754702. [PMID: 34887860 PMCID: PMC8651476 DOI: 10.3389/fimmu.2021.754702] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/14/2021] [Indexed: 12/26/2022] Open
Abstract
The IL-36 family of cytokines were identified in the early 2000’s as a new subfamily of the IL-1 cytokine family, and since then, the role of IL-36 cytokines during various inflammatory processes has been characterized. While most of the research has focused on the role of these cytokines in autoimmune skin diseases such as psoriasis and dermatitis, recent studies have also shown the importance of IL-36 cytokines in the lung inflammatory response during infectious and non-infectious diseases. In this review, we discuss the biology of IL-36 cytokines in terms of how they are produced and activated, as well as their effects on myeloid and lymphoid cells during inflammation. We also discuss the role of these cytokines during lung infectious diseases caused by bacteria and influenza virus, as well as other inflammatory conditions in the lungs such as allergic asthma, lung fibrosis, chronic obstructive pulmonary disease, cystic fibrosis and cancer. Finally, we discuss the current therapeutic advances that target the IL-36 pathway and the possibility to extend these tools to treat lung inflammatory diseases.
Collapse
Affiliation(s)
- Hernán F Peñaloza
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rick van der Geest
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joel A Ybe
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN, United States
| | - Theodore J Standiford
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Janet S Lee
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
38
|
Guo HZ, Guo ZH, Yu SH, Niu LT, Qiang WT, Huang MM, Tian YY, Chen J, Yang H, Weng XQ, Zhang Y, Zhang W, Hu SY, Shi J, Zhu J. Leukemic progenitor cells enable immunosuppression and post-chemotherapy relapse via IL-36-inflammatory monocyte axis. SCIENCE ADVANCES 2021; 7:eabg4167. [PMID: 34623912 PMCID: PMC8500518 DOI: 10.1126/sciadv.abg4167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 08/17/2021] [Indexed: 06/13/2023]
Abstract
Chemotherapy can effectively reduce the leukemic burden and restore immune cell production in most acute myeloid leukemia (AML) cases. Nevertheless, endogenous immunosurveillance usually fails to recover after chemotherapy, permitting relapse. The underlying mechanisms of this therapeutic failure have remained poorly understood. Here, we show that abnormal IL-36 production activated by NF-κB is an essential feature of mouse and human leukemic progenitor cells (LPs). Mechanistically, IL-36 directly activates inflammatory monocytes (IMs) in bone marrow, which then precludes clearance of leukemia mediated by CD8+ T cells and facilitates LP growth. While sparing IMs, common chemotherapeutic agents stimulate IL-36 production from residual LPs via caspase-1 activation, thereby enabling the persistence of this immunosuppressive IL-36–IM axis after chemotherapy. Furthermore, IM depletion by trabectedin, with chemotherapy and PD-1 blockade, can synergistically restrict AML progression and relapse. Collectively, these results suggest inhibition of the IL-36–IM axis as a potential strategy for improving AML treatment.
Collapse
Affiliation(s)
- He-Zhou Guo
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
- Department of Hematology, Shanghai Ninth People’s Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200011, China
| | - Zi-Hua Guo
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Shan-He Yu
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Li-Ting Niu
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Wan-Ting Qiang
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Meng-Meng Huang
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Yuan-Yuan Tian
- Fels Institute, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Juan Chen
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Hui Yang
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Xiang-Qin Weng
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Yi Zhang
- Fels Institute, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Wu Zhang
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Shao-Yan Hu
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, No 92, Zhongnan Street, Suzhou 215025, China
| | - Jun Shi
- Department of Hematology, Shanghai Ninth People’s Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200011, China
| | - Jiang Zhu
- Shanghai Institute of Hematology and State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated with Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
- School of Life Sciences and Biotechnology, Shanghai Jiao-Tong University, Shanghai 200025, China
| |
Collapse
|
39
|
Wang P, Yang W, Guo H, Dong H, Guo Y, Gan H, Wang Z, Cheng Y, Deng Y, Xie S, Yang X, Lin D, Zhong B. IL-36γ and IL-36Ra Reciprocally Regulate NSCLC Progression by Modulating GSH Homeostasis and Oxidative Stress-Induced Cell Death. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101501. [PMID: 34369094 PMCID: PMC8498882 DOI: 10.1002/advs.202101501] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/31/2021] [Indexed: 05/05/2023]
Abstract
The balance between antioxidants and reactive oxygen species (ROS) critically regulates tumor initiation and progression. However, whether and how the tumor-favoring redox status is controlled by cytokine networks remain poorly defined. Here, it is shown that IL-36γ and IL-36Ra reciprocally regulate the progression of non-small cell lung cancer (NSCLC) by modulating glutathione metabolism and ROS resolution. Knockout, inhibition, or neutralization of IL-36γ significantly inhibits NSCLC progression and prolongs survival of the KrasLSL-G12D/+ Tp53fl/fl and KrasLSL-G12D/+ Lkb1fl/fl mice after tumor induction, whereas knockout of IL-36Ra exacerbates tumorigenesis in these NSCLC mouse models and accelerates death of mice. Mechanistically, IL-36γ directly upregulates an array of genes involved in glutathione homeostasis to reduce ROS and prevent oxidative stress-induced cell death, which is mitigated by IL-36Ra or IL-36γ neutralizing antibody. Consistently, IL-36γ staining is positively and negatively correlated with glutathione biosynthesis and ROS in human NSCLC tumor biopsies, respectively. These findings highlight essential roles of cytokine networks in redox for tumorigenesis and provide potential therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Peng Wang
- Department of Gastrointestinal SurgeryCollege of Life SciencesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of ImmunologyMedical Research Institute and Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Wuhan Research Center for Infectious Diseases and CancerChinese Academy of Medical SciencesWuhan430071China
| | - Wei Yang
- Department of Gastrointestinal SurgeryCollege of Life SciencesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of ImmunologyMedical Research Institute and Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
| | - Hao Guo
- Department of Gastrointestinal SurgeryCollege of Life SciencesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of ImmunologyMedical Research Institute and Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
| | - Hong‐Peng Dong
- Department of Gastrointestinal SurgeryCollege of Life SciencesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of ImmunologyMedical Research Institute and Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
| | - Yu‐Yao Guo
- Department of Gastrointestinal SurgeryCollege of Life SciencesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of ImmunologyMedical Research Institute and Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
| | - Hu Gan
- Department of Gastrointestinal SurgeryCollege of Life SciencesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of ImmunologyMedical Research Institute and Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
| | - Zou Wang
- Wuhan Biobank Co., Ltd, WuhanWuhan430075China
| | | | - Yu Deng
- Department of Thoracic SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Shizhe Xie
- CAS Key Laboratory of Special PathogensWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xinglou Yang
- CAS Key Laboratory of Special PathogensWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of Chinese Academy of SciencesBeijing100049China
| | - Dandan Lin
- Cancer CenterRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Bo Zhong
- Department of Gastrointestinal SurgeryCollege of Life SciencesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of Pulmonary and Critical Care MedicineZhongnan Hospital of Wuhan UniversityWuhan430071China
- Department of ImmunologyMedical Research Institute and Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071China
- Wuhan Research Center for Infectious Diseases and CancerChinese Academy of Medical SciencesWuhan430071China
| |
Collapse
|
40
|
Boersma B, Jiskoot W, Lowe P, Bourquin C. The interleukin-1 cytokine family members: Role in cancer pathogenesis and potential therapeutic applications in cancer immunotherapy. Cytokine Growth Factor Rev 2021; 62:1-14. [PMID: 34620560 DOI: 10.1016/j.cytogfr.2021.09.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023]
Abstract
The interleukin-1 (IL-1) family is one of the first described cytokine families and consists of eight cytokines (IL-1β, IL-1α, IL-18, IL-33, IL-36α, IL-36β, IL-36γ and IL-37) and three receptor antagonists (IL-1Ra, IL-36Ra and IL-38). The family members are known to play an essential role in inflammation. The importance of inflammation in cancer has been well established in the past decades. This review sets out to give an overview of the role of each IL-1 family member in cancer pathogenesis and show their potential as potential anticancer drug candidates. First, the molecular structure is described. Next, both the pro- and anti-tumoral properties are highlighted. Additionally, a critical interpretation of current literature is given. To conclude, the IL-1 family is a toolbox with a collection of powerful tools that can be considered as potential drugs or drug targets.
Collapse
Affiliation(s)
- Bart Boersma
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland.
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| | - Peter Lowe
- Department of Biomolecule Generation and Optimization, Institut de Recherche Pierre Fabre, Centre d'Immunologie Pierre Fabre, Saint-Julien-en-Genevois, France.
| | - Carole Bourquin
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Department of Anesthesiology, Pharmacology and Intensive Care, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
41
|
Elias M, Zhao S, Le HT, Wang J, Neurath MF, Neufert C, Fiocchi C, Rieder F. IL-36 in chronic inflammation and fibrosis - bridging the gap? J Clin Invest 2021; 131:144336. [PMID: 33463541 DOI: 10.1172/jci144336] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IL-36 is a member of the IL-1 superfamily and consists of three agonists and one receptor antagonist (IL-36Ra). The three endogenous agonists, IL-36α, -β, and -γ, act primarily as proinflammatory cytokines, and their signaling through the IL-36 receptor (IL-36R) promotes immune cell infiltration and secretion of inflammatory and chemotactic molecules. However, IL-36 signaling also fosters secretion of profibrotic soluble mediators, suggesting a role in fibrotic disorders. IL-36 isoforms and IL-36 have been implicated in inflammatory diseases including psoriasis, arthritis, inflammatory bowel diseases, and allergic rhinitis. Moreover, IL-36 has been connected to fibrotic disorders affecting the kidney, lung, and intestines. This review summarizes the expression, cellular source, and function of IL-36 in inflammation and fibrosis in various organs, and proposes that IL-36 modulation may prove valuable in preventing or treating inflammatory and fibrotic diseases and may reveal a mechanistic link between inflammation and fibrosis.
Collapse
Affiliation(s)
- Michael Elias
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Shuai Zhao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hongnga T Le
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jie Wang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Markus F Neurath
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie DZI, Universitaetsklinikum Erlangen, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Clemens Neufert
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie DZI, Universitaetsklinikum Erlangen, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
42
|
Byrne J, Baker K, Houston A, Brint E. IL-36 cytokines in inflammatory and malignant diseases: not the new kid on the block anymore. Cell Mol Life Sci 2021; 78:6215-6227. [PMID: 34365521 PMCID: PMC8429149 DOI: 10.1007/s00018-021-03909-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 11/02/2022]
Abstract
The IL-36 family of cytokines were first identified in 2000 based on their sequence homology to IL-1 cytokines. Over subsequent years, the ability of these cytokines to either agonise or antagonise an IL-1R homologue, now known as the IL-36 Receptor (IL-36R), was identified and these cytokines went through several cycles of renaming with the current nomenclature being proposed in 2010. Despite being identified over 20 years ago, it is only during the last decade that the function of these cytokines in health and disease has really begun to be appreciated, with both homeostatic functions in wound healing and response to infection, as well as pathological functions now ascribed. In the disease context, over activation of IL-36 has now been associated with many inflammatory diseases including Psoriasis and inflammatory bowel diseases, with roles in cancer also now being investigated. This review summarises the current knowledge of IL-36 biology, its role in inflammatory diseases and focuses on an emerging role for IL-36 in cancer.
Collapse
Affiliation(s)
- James Byrne
- Department of Pathology, Cork University Hospital, University College Cork, Clinical Sciences Building, Cork, Ireland
| | - Kevin Baker
- Department of Pathology, Cork University Hospital, University College Cork, Clinical Sciences Building, Cork, Ireland
| | - Aileen Houston
- Department of Medicine, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, Cork University Hospital, University College Cork, Clinical Sciences Building, Cork, Ireland. .,APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
43
|
Giannoudaki E, Stefanska AM, Lawler H, Leon G, Hernandez Santana YE, Hassan N, Russell SE, Horan R, Sweeney C, Preston RS, Mantovani A, Garlanda C, Fallon PG, Walsh PT. SIGIRR Negatively Regulates IL-36-Driven Psoriasiform Inflammation and Neutrophil Infiltration in the Skin. THE JOURNAL OF IMMUNOLOGY 2021; 207:651-660. [PMID: 34253575 DOI: 10.4049/jimmunol.2100237] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/17/2021] [Indexed: 11/19/2022]
Abstract
SIGIRR has been described as a negative regulator of several IL-1R/TLR family members and has been implicated in several inflammatory disease conditions. However, it is unknown whether it can suppress IL-36 family cytokines, which are members of the broader IL-1 superfamily that have emerged as critical orchestrators of psoriatic inflammation in both humans and mice. In this study, we demonstrate that SIGIRR is downregulated in psoriatic lesions in humans and mice, and this correlates with increased expression of IL-36 family cytokines. Using Sigirr -/- mice, we identify, for the first time (to our knowledge), SIGIRR as a negative regulator of IL-36 responses in the skin. Mechanistically, we identify dendritic cells and keratinocytes as the primary cell subsets in which IL-36 proinflammatory responses are regulated by SIGIRR. Both cell types displayed elevated IL-36 responsiveness in absence of SIGIRR activity, characterized by enhanced expression of neutrophil chemoattractants, leading to increased neutrophil infiltration to the inflamed skin. Blockade of IL-36R signaling ameliorated exacerbated psoriasiform inflammation in Sigirr-/- mice and inhibited neutrophil infiltration. These data identify SIGIRR activity as an important regulatory node in suppressing IL-36-dependent psoriatic inflammation in humans and mice.
Collapse
Affiliation(s)
- Eirini Giannoudaki
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Anna M Stefanska
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Hazel Lawler
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Gemma Leon
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Yasmina E Hernandez Santana
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Najma Hassan
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Shane E Russell
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Rachel Horan
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Cheryl Sweeney
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Roger S Preston
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alberto Mantovani
- Scientific Institute for Research, Hospitalization and Healthcare, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; and.,The William Harvey Research Institute, Queen Mary University, London, United Kingdom
| | - Cecilia Garlanda
- Scientific Institute for Research, Hospitalization and Healthcare, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; and
| | - Padraic G Fallon
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland.,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Patrick T Walsh
- Trinity Translational Medicine Institute, School of Medicine, Trinity College, Dublin, Ireland; .,National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland
| |
Collapse
|
44
|
Cytokine/Chemokine Release Patterns and Transcriptomic Profiles of LPS/IFNγ-Activated Human Macrophages Differentiated with Heat-Killed Mycobacterium obuense, M-CSF, or GM-CSF. Int J Mol Sci 2021; 22:ijms22137214. [PMID: 34281268 PMCID: PMC8268300 DOI: 10.3390/ijms22137214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 01/15/2023] Open
Abstract
Macrophages (Mφs) are instrumental regulators of the immune response whereby they acquire diverse functional phenotypes following their exposure to microenvironmental cues that govern their differentiation from monocytes and their activation. The complexity and diversity of the mycobacterial cell wall have empowered mycobacteria with potent immunomodulatory capacities. A heat-killed (HK) whole-cell preparation of Mycobacterium obuense (M. obuense) has shown promise as an adjunctive immunotherapeutic agent for the treatment of cancer. Moreover, HK M. obuense has been shown to trigger the differentiation of human monocytes into a monocyte-derived macrophage (MDM) type named Mob-MDM. However, the transcriptomic profile and functional properties of Mob-MDMs remain undefined during an activation state. Here, we characterized cytokine/chemokine release patterns and transcriptomic profiles of lipopolysaccharide (LPS)/interferon γ (IFNγ)-activated human MDMs that were differentiated with HK M. obuense (Mob-MDM(LPS/IFNγ)), macrophage colony-stimulating factor M-MDM(LPS/IFNγ)), or granulocyte/macrophage colony-stimulating factor (GM-MDM(LPS/IFNγ)). Mob-MDM(LPS/IFNγ) demonstrated a unique cytokine/chemokine release pattern (interleukin (IL)-10low, IL-12/23p40low, IL-23p19/p40low, chemokine (C-x-C) motif ligand (CXCL)9low) that was distinct from those of M-MDM(LPS/IFNγ) and GM-MDM(LPS/IFNγ). Furthermore, M-MDM(LPS/IFNγ) maintained IL-10 production at significantly higher levels compared to GM-MDM(LPS/IFNγ) and Mob-MDM(LPS/IFNγ) despite being activated with M1-Mφ-activating stimuli. Comparative RNA sequencing analysis pointed to a distinct transcriptome profile for Mob-MDM(LPS/IFNγ) relative to both M-MDM(LPS/IFNγ) and GM-MDM(LPS/IFNγ) that comprised 417 transcripts. Functional gene-set enrichment analysis revealed significant overrepresentation of signaling pathways and biological processes that were uniquely related to Mob-MDM(LPS/IFNγ). Our findings lay a foundation for the potential integration of HK M. obuense in specific cell-based immunotherapeutic modalities such as adoptive transfer of Mφs (Mob-MDM(LPS/IFNγ)) for cancer treatment.
Collapse
|
45
|
Bell M, Gottschalk S. Engineered Cytokine Signaling to Improve CAR T Cell Effector Function. Front Immunol 2021; 12:684642. [PMID: 34177932 PMCID: PMC8220823 DOI: 10.3389/fimmu.2021.684642] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/11/2021] [Indexed: 12/20/2022] Open
Abstract
Adoptive immunotherapy with T cells genetically modified to express chimeric antigen receptors (CARs) is a promising approach to improve outcomes for cancer patients. While CAR T cell therapy is effective for hematological malignancies, there is a need to improve the efficacy of this therapeutic approach for patients with solid tumors and brain tumors. At present, several approaches are being pursued to improve the antitumor activity of CAR T cells including i) targeting multiple antigens, ii) improving T cell expansion/persistence, iii) enhancing homing to tumor sites, and iv) rendering CAR T cells resistant to the immunosuppressive tumor microenvironment (TME). Augmenting signal 3 of T cell activation by transgenic expression of cytokines or engineered cytokine receptors has emerged as a promising strategy since it not only improves CAR T cell expansion/persistence but also their ability to function in the immunosuppressive TME. In this review, we will provide an overview of cytokine biology and highlight genetic approaches that are actively being pursued to augment cytokine signaling in CAR T cells.
Collapse
Affiliation(s)
- Matthew Bell
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
46
|
Wang X, Yi P, Liang Y. The Role of IL-36 in Infectious Diseases: Potential Target for COVID-19? Front Immunol 2021; 12:662266. [PMID: 34054828 PMCID: PMC8155493 DOI: 10.3389/fimmu.2021.662266] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/28/2021] [Indexed: 12/24/2022] Open
Abstract
IL-36 is a member of the interleukin 1 cytokine family, which is currently experiencing a renaissance due to the growing understanding of its context-dependent roles and advances in our understanding of the inflammatory response. The immunological role of IL-36 has revealed its profound and indispensable functional roles in psoriasis, as well as in several inflammatory diseases, including inflammatory bowel disease (IBD), systemic lupus erythematosus, rheumatoid arthritis (RA) and cancer. More recently, an increasing body of evidence suggests that IL-36 plays a crucial role in viral, bacterial and fungal infections. There is a growing interest as to whether IL-36 contributes to host protective immune responses against infection as well as the potential implications of IL-36 for the development of new therapeutic strategies. In this review, we summarize the recent progress in understanding cellular expression, regulatory mechanisms and biological roles of IL-36 in infectious diseases, which suggest more specific strategies to maneuver IL-36 as a diagnostic or therapeutic target, especially in COVID-19.
Collapse
Affiliation(s)
- Xiaofang Wang
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, China
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Panpan Yi
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, China
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
47
|
Beck JD, Reidenbach D, Salomon N, Sahin U, Türeci Ö, Vormehr M, Kranz LM. mRNA therapeutics in cancer immunotherapy. Mol Cancer 2021; 20:69. [PMID: 33858437 PMCID: PMC8047518 DOI: 10.1186/s12943-021-01348-0] [Citation(s) in RCA: 220] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/15/2021] [Indexed: 02/08/2023] Open
Abstract
Synthetic mRNA provides a template for the synthesis of any given protein, protein fragment or peptide and lends itself to a broad range of pharmaceutical applications, including different modalities of cancer immunotherapy. With the ease of rapid, large scale Good Manufacturing Practice-grade mRNA production, mRNA is ideally poised not only for off-the shelf cancer vaccines but also for personalized neoantigen vaccination. The ability to stimulate pattern recognition receptors and thus an anti-viral type of innate immune response equips mRNA-based vaccines with inherent adjuvanticity. Nucleoside modification and elimination of double-stranded RNA can reduce the immunomodulatory activity of mRNA and increase and prolong protein production. In combination with nanoparticle-based formulations that increase transfection efficiency and facilitate lymphatic system targeting, nucleoside-modified mRNA enables efficient delivery of cytokines, costimulatory receptors, or therapeutic antibodies. Steady but transient production of the encoded bioactive molecule from the mRNA template can improve the pharmacokinetic, pharmacodynamic and safety properties as compared to the respective recombinant proteins. This may be harnessed for applications that benefit from a higher level of expression control, such as chimeric antigen receptor (CAR)-modified adoptive T-cell therapies. This review highlights the advancements in the field of mRNA-based cancer therapeutics, providing insights into key preclinical developments and the evolving clinical landscape.
Collapse
Affiliation(s)
- Jan D Beck
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany
| | - Daniel Reidenbach
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University gGmbH, Freiligrathstraße 12, 55131, Mainz, Germany
| | - Nadja Salomon
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University gGmbH, Freiligrathstraße 12, 55131, Mainz, Germany
| | - Ugur Sahin
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany
| | - Özlem Türeci
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany
| | | | - Lena M Kranz
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany.
| |
Collapse
|
48
|
Santos PM, Adamik J, Howes TR, Du S, Vujanovic L, Warren S, Gambotto A, Kirkwood JM, Butterfield LH. Impact of checkpoint blockade on cancer vaccine-activated CD8+ T cell responses. J Exp Med 2021; 217:151736. [PMID: 32369107 PMCID: PMC7336310 DOI: 10.1084/jem.20191369] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/04/2019] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Immune and molecular profiling of CD8 T cells of patients receiving DC vaccines expressing three full-length melanoma antigens (MAs) was performed. Antigen expression levels in DCs had no significant impact on T cell or clinical responses. Patients who received checkpoint blockade before DC vaccination had higher baseline MA-specific CD8 T cell responses but no evidence for improved functional responses to the vaccine. Patients who showed the best clinical responses had low PD-1 expression on MA-specific T cells before and after DC vaccination; however, blockade of PD-1 during antigen presentation by DC had minimal functional impact on PD-1high MA-specific T cells. Gene and protein expression analyses in lymphocytes and tumor samples identified critical immunoregulatory pathways, including CTLA-4 and PD-1. High immune checkpoint gene expression networks correlated with inferior clinical outcomes. Soluble serum PD-L2 showed suggestive positive association with improved outcome. These findings show that checkpoint molecular pathways are critical for vaccine outcomes and suggest specific sequencing of vaccine combinations.
Collapse
Affiliation(s)
- Patricia M Santos
- University of Pittsburgh Medical Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Juraj Adamik
- Parker Institute for Cancer Immunotherapy, San Francisco, CA
| | - Timothy R Howes
- Parker Institute for Cancer Immunotherapy, San Francisco, CA
| | - Samuel Du
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Lazar Vujanovic
- University of Pittsburgh Medical Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | | | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - John M Kirkwood
- University of Pittsburgh Medical Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA.,Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Lisa H Butterfield
- University of Pittsburgh Medical Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA.,Parker Institute for Cancer Immunotherapy, San Francisco, CA.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA.,Department of Surgery, University of Pittsburgh, Pittsburgh, PA.,Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
49
|
Yang M, Giehl E, Feng C, Feist M, Chen H, Dai E, Liu Z, Ma C, Ravindranathan R, Bartlett DL, Lu B, Guo ZS. IL-36γ-armed oncolytic virus exerts superior efficacy through induction of potent adaptive antitumor immunity. Cancer Immunol Immunother 2021; 70:2467-2481. [PMID: 33538860 PMCID: PMC8360872 DOI: 10.1007/s00262-021-02860-4] [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: 10/15/2020] [Accepted: 01/11/2021] [Indexed: 01/22/2023]
Abstract
In this study, we aimed to apply the cytokine IL-36γ to cancer immunotherapy by constructing new oncolytic vaccinia viruses (OV) expressing interleukin-36γ (IL-36γ-OVs), leveraging unique synergism between OV and IL-36γ’s ability to promote antitumor adaptive immunity and modulate tumor microenvironment (TME). IL-36γ-OV had dramatic therapeutic efficacies in multiple murine tumor models, frequently leading to complete cancer eradication in large fractions of mice. Mechanistically, IL-36-γ-armed OV induced infiltration of lymphocytes and dendritic cells, decreased myeloid-derived suppressor cells and M2-like tumor-associated macrophages, and T cell differentiation into effector cells. Further study showed that IL-36γ-OV increased the number of tumor antigen-specific CD4+ and CD8+ T cells and the therapeutic efficacy depended on both CD8+ and CD4+ T cells. These results demonstrate that these IL36γ-armed OVs exert potent therapeutic efficacy mainly though antitumor immunity and they may hold great potential to advance treatment in human cancer patients.
Collapse
Affiliation(s)
- Min Yang
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Esther Giehl
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany
| | - Chao Feng
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mathilde Feist
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Surgery, CCM/CVK, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Hongqi Chen
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Enyong Dai
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zuqiang Liu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Congrong Ma
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Roshni Ravindranathan
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L Bartlett
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,AHN-Cancer Institute, Pittsburgh, PA, USA
| | - Binfeng Lu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA. .,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Zong Sheng Guo
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA. .,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
50
|
Zhang F, Jiang J, Xu B, Xu Y, Wu C. Over-expression of CXCL2 is associated with poor prognosis in patients with ovarian cancer. Medicine (Baltimore) 2021; 100:e24125. [PMID: 33530204 PMCID: PMC7850676 DOI: 10.1097/md.0000000000024125] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/08/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND In the present study, we aimed to detect the expression of CXCL2 in epithelial ovarian cancer (OC) and explore its clinical significance. METHODS TCGA (The Cancer Genome Atlas) database was adopted to assess the significance of CXCL2. Tissue microarray and immunohistochemical staining were used to detect the expression of CXCL2 in epithelial OC, and its correlation with clinicopathological features and prognosis was statistically analyzed. RESULTS CXCL2 was highly expressed in epithelial OC tissues compared with the adjacent tissues. Such up-regulation of CXCL2 was significantly correlated with tumor differentiation (P = .001), tumor stage (P = .01), tumor location (unilateral or bilateral) (P = .003), and metastasis (P = .003). Kaplan-Meier and Cox proportional hazards regression analyses showed that high expression of CXCL2 was not an independent predictor of poor prognosis in epithelial OC. CONCLUSIONS Collectively, the high expression of CXCL2 might be related to the invasion and metastasis of epithelial OC.
Collapse
Affiliation(s)
- Fenghua Zhang
- Department of Tumor Biological Treatment
- Jiangsu Engineering Research Center for Tumor Immunotherapy
- Department of Gynaecology, the Third Affiliated Hospital of Soochow University
- Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment
- Jiangsu Engineering Research Center for Tumor Immunotherapy
- Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Bin Xu
- Department of Tumor Biological Treatment
- Jiangsu Engineering Research Center for Tumor Immunotherapy
- Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Yun Xu
- Department of Tumor Biological Treatment
- Jiangsu Engineering Research Center for Tumor Immunotherapy
- Department of Gynaecology, the Third Affiliated Hospital of Soochow University
- Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Changping Wu
- Department of Tumor Biological Treatment
- Jiangsu Engineering Research Center for Tumor Immunotherapy
- Institute of Cell Therapy, Soochow University, Changzhou, China
| |
Collapse
|