1
|
Wang X, Wang X, Wang D, Zhou C, Lv K, Ma Y, Chang W, Wang B, Hu J, Ji Y, Dai Z, Ma Y. Interleukin-10 overexpression in 4T1 cells: A gateway to suppressing mammary carcinoma growth. Int Immunopharmacol 2024; 142:113089. [PMID: 39244897 DOI: 10.1016/j.intimp.2024.113089] [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: 04/09/2024] [Revised: 08/04/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
Interleukin-10 (IL-10) exerts complex effects on tumor growth, exhibiting both pro- and anti-tumor properties. Recent focus on the anti-inflammatory properties of IL-10 has highlighted its potential anti-tumor properties, particularly through the enhancement of CD8+ T cell activity. However, further research is needed to fully elucidate its other anti-tumor mechanisms. Our study investigates novel anti-tumor mechanisms of IL-10 in a murine mammary carcinoma model (4T1). We found that IL-10 overexpression in mouse 4T1 cells suppressed tumor growth in vivo. This suppression was accompanied by an increase in IFN-γ-secreting CD8+ T cells and a decrease in myeloid-derived suppressor cells (MDSCs) in tumor tissue. In vitro experiments showed that IL-10-rich tumor cell-derived supernatants inhibited myeloid cell differentiation into monocytic and granulocytic MDSCs while reducing MDSCs migration. In addition, IL-10 overexpression downregulated CXCL5 expression in 4T1 cells, resulting in decreased CXCR2+ MDSCs infiltration. Using RAG1-deficient mice and CXCL5 knockdown tumor models, we demonstrated that the anti-tumor effects of IL-10 depend on both CD8+ T cells and reduced MDSC infiltration. IL-10 attenuated the immunosuppressive tumor microenvironment by enhancing CD8+ T cell activity and inhibiting MDSCs infiltration. In human breast cancer, we observed a positive correlation between CXCL5 expression and MDSC infiltration. Our findings reveal a dual mechanism of IL-10-mediated tumor suppression: (1) direct enhancement of CD8+ T cell activity and (2) indirect reduction of immunosuppressive MDSCs through CXCL5 downregulation and inhibition of myeloid cell differentiation. This study provides new insights into the role of IL-10 in anti-tumor immunity and suggests potential strategies for breast cancer immunotherapy by modulating the IL-10-CXCL5-MDSCs axis.
Collapse
Affiliation(s)
- Xiaoqin Wang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoqian Wang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Dan Wang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Can Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kaige Lv
- Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanfen Ma
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Wenjing Chang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Baofeng Wang
- Department of Radiotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jian Hu
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yanhong Ji
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Yunfeng Ma
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| |
Collapse
|
2
|
Robinson CJ, Moeller CE, Quick CN, Goodermuth CE, Carossino M, Withers SS. Macrophage Colony Stimulating Factor (M-CSF) and Interleukin-34 (IL-34) Expression in Canine Osteosarcoma in the Context of the Tumour Immune Microenvironment. Vet Comp Oncol 2024; 22:480-489. [PMID: 39164469 DOI: 10.1111/vco.12993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 08/22/2024]
Abstract
Canine osteosarcoma (OSA) is a malignancy that has been shown to modulate the host immune system. Macrophage colony-stimulating factor (M-CSF; CSF1) and interleukin-34 (IL-34; IL34) are both ligands of colony stimulating factor 1 receptor (CSF-1R), and may play a role in the pathogenesis of a variety of human cancers, including OSA. This study aimed to, (1) assess M-CSF and IL-34 expression in canine OSA cell lines and tissue samples, and (2) determine any correlations between M-CSF and IL-34 expression and immune cell infiltrates within canine OSA tissues. Four canine OSA cell lines and canine osteoblasts were treated with control media, TNFα (10 ng/mL) or IL-1β (10 ng/mL) and analysed with RT-qPCR and ELISA. IL-34 and M-CSF mRNA and protein were detectable in all cell lines, however upregulation following TNFα or IL-1β exposure was only consistently observed for transcript expression. Baseline expression of CSF1 and IL34 mRNA in OSA cell lines was equal to or higher than that of canine osteoblasts. All 10 OSA tissue samples expressed IL34 and CSF1 transcripts to varying degrees. Furthermore, CSF1 and IL34 expression both showed a moderate to high degree of correlation with M1 macrophage lineage-associated transcripts (CD80 and IL15RA). There was a moderate degree of correlation between CSF1 and CD163, but no correlation between IL34 and either M2 macrophage-associated transcripts (CD163 and CCL24). In summary, IL-34 and M-CSF are expressed in canine OSA cell lines and tissues, and expression positively correlates with a wide range of immune-related transcripts.
Collapse
Affiliation(s)
- Christopher J Robinson
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Cambri E Moeller
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Cally N Quick
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Christine E Goodermuth
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
- Louisiana Animal Disease Diagnostic Laboratory (LADDL), Baton Rouge, Louisiana, USA
| | - Sita S Withers
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, South Australia, Australia
| |
Collapse
|
3
|
Mueller BR, Mehta M, Campbell M, Neupane N, Cedillo G, Lee G, Coyle K, Qi J, Chen Z, George MC, Robinson-Papp J. Autonomic and Immune Stress Response Networks in Patients Living With HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618447. [PMID: 39464041 PMCID: PMC11507734 DOI: 10.1101/2024.10.15.618447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Background and Objectives Stress response systems are frequently dysregulated in patients with chronic inflammatory disorders. Pre-clinical studies have demonstrated direct influences of the sympathetic and vagal/parasympathetic branches of the autonomic nervous system (ANS) on the immune system. However, these connections have not been examined in humans. We hypothesized that the subtype and severity of autonomic neuropathy (AN) would predict immune phenotypes with distinct clinical and demographic characteristics in people living with HIV. Methods This is a cross-sectional study of 79 adult people with a history of well-controlled HIV on stable combination antiretroviral treatment (CART) recruited from a primary care clinic network within the Mount Sinai Health System in New York City. All participants underwent a standardized battery of autonomic function tests summarized as the Composite Autonomic Severity Score (CASS) and vagal and adrenergic baroreflex sensitivity (BRS-V and BRS-A). Immune profiling included: 1) measurement of interleukin-6 (IL-6) as part of the Olink assay Target 96 Inflammation Panel, 2) non-negative matrix factorization (NMF) clustering analyses on Olink immune biomarkers, and 3) mass cytometry (CyTOF) on a subset of participants with and without autonomic neuropathy (N = 10). Results Reduced activity of caudal vagal circuitry involved in the cholinergic anti-inflammatory pathway (CAP) predicted higher levels of IL-6 (Spearman's rho = -0.352, p=0.002). The comprehensive assessment of the ANS-immune network showed four immunotypes defined by NMF analyses. A pro-inflammatory immunotype defined by elevations in type 1 cytokines (IL-6, IL-17) and increased numbers of CD8+ T-cells was associated with autonomic neuropathy (AN). This association was driven by deficits in the cardiovascular sympathetic nervous system and remained strongly significant after controlling for the older age and greater burden of co-morbid illness among participants with this immunotype (aOR=4.7, p=0.017). Discussion Our results provide novel support for the clinical relevance of the CAP in patients with chronic inflammatory AN. These data also provide insight regarding the role of the sympathetic nervous system and aging in the progression and development of co-morbidities in patients with chronic HIV and support future research aimed at developing therapies focused on modulation of the sympathetic and parasympathetic/vagal nervous system.
Collapse
|
4
|
Branchett WJ, Saraiva M, O'Garra A. Regulation of inflammation by Interleukin-10 in the intestinal and respiratory mucosa. Curr Opin Immunol 2024; 91:102495. [PMID: 39357078 DOI: 10.1016/j.coi.2024.102495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
Intricate immune regulation is required at mucosal surfaces to allow tolerance to microbiota and harmless allergens and to prevent overexuberant inflammatory responses to pathogens. The cytokine Interleukin-10 (IL-10) is a key mediator of mucosal immune regulation. While IL-10 can be produced by virtually all cells of the immune system, many of its in vivo functions depend upon its production by regulatory or effector T cell populations and its signalling to macrophages, dendritic cells and specific T cell subsets. In this review, we discuss our current understanding of the role of IL-10 in regulation of immune responses, with a focus on its context-specific roles in intestinal homeostasis, respiratory infection and asthma. We highlight the importance of appropriate production and function of IL-10 for balancing pathogen clearance, control of microbiota and host tissue damage, and that precise modulation of IL-10 functions in vivo could present therapeutic opportunities.
Collapse
Affiliation(s)
- William J Branchett
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, United Kingdom.
| | - Margarida Saraiva
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Anne O'Garra
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, United Kingdom; National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
5
|
Horikawa M, Masuda K, Takahashi H, Tada H, Tomidokoro Y, Motegi M, Oyama T, Takeda S, Chikamatsu K. Tumor antigen‑specific interleukin‑10‑producing T‑cell response in patients with head and neck squamous cell carcinoma. Oncol Lett 2024; 28:456. [PMID: 39100998 PMCID: PMC11294976 DOI: 10.3892/ol.2024.14589] [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: 06/10/2024] [Accepted: 07/09/2024] [Indexed: 08/06/2024] Open
Abstract
Interleukin-10 (IL-10) is a highly pleiotropic cytokine that regulates immunological homeostasis through anti-inflammatory and/or immunostimulatory functions. Moreover, IL-10 is well known to exert diverse roles in tumor immunology and immunotherapy. The present study investigated the presence of circulating tumor antigen-specific IL-10-producing T cells in patients with head and neck squamous cell carcinoma (HNSCC), and determined factors that may influence the immunodynamics of IL-10-producing T cells. In vitro, peripheral blood mononuclear cells (PBMCs) stimulated with the tumor antigens p53 and MAGE-A4 were evaluated for interferon (IFN)-γ/IL-10 production using the IFN-γ/IL-10 double-color enzyme-linked immunosorbent spot assay. The proportion of T cells expressing immune checkpoint molecules in PBMCs was analyzed using flow cytometry. Of the 18 patients with HNSCC, 2 (11.1%) and 9 (50.0%) exhibited p53-specific IFN-γ and IL-10 production, respectively. Meanwhile, MAGE-A4-specific IFN-γ and IL-10 production was detected in 4 (28.6%) and 7 (50.0%) of 14 patients. In the p53-specific responses, IL-10-producing T cells were observed in significantly more patients than IFN-γ producing T cells (P=0.0275). In both CD4+ and CD8+ T cells, the proportion of T cells expressing lymphocyte activation gene-3 (Lag-3) was significantly lower in patients with p53-specific IL-10 production than in those without. In certain patients, Lag-3 blockade enhanced tumor antigen-specific IL-10. Taken together, the present study successfully demonstrated that tumor antigen-specific IL-10-producing T cells exist in the peripheral blood of patients with HNSCC and that Lag-3+ T cells may serve an important role in modulating IL-10-producing T cells. These findings provide novel insights into the roles of IL-10 and Lag-3 in mediating antitumor immune responses.
Collapse
Affiliation(s)
- Momoka Horikawa
- Faculty of Science and Technology, Division of Molecular Science, Gunma University, Kiryu, Gunma 376-8515, Japan
| | - Kei Masuda
- Department of Pathology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hideyuki Takahashi
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hiroe Tada
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Yuichi Tomidokoro
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Masaomi Motegi
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Tetsunari Oyama
- Department of Pathology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Shigeki Takeda
- Faculty of Science and Technology, Division of Molecular Science, Gunma University, Kiryu, Gunma 376-8515, Japan
| | - Kazuaki Chikamatsu
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
6
|
Boersma B, Poinot H, Pommier A. Stimulating the Antitumor Immune Response Using Immunocytokines: A Preclinical and Clinical Overview. Pharmaceutics 2024; 16:974. [PMID: 39204319 PMCID: PMC11357675 DOI: 10.3390/pharmaceutics16080974] [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: 06/03/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Cytokines are immune modulators which can enhance the immune response and have been proven to be an effective class of immunotherapy. Nevertheless, the clinical use of cytokines in cancer treatment has faced several challenges associated with poor pharmacokinetic properties and the occurrence of adverse effects. Immunocytokines (ICKs) have emerged as a promising approach to overcome the pharmacological limitations observed with cytokines. ICKs are fusion proteins designed to deliver cytokines in the tumor microenvironment by taking advantage of the stability and specificity of immunoglobulin-based scaffolds. Several technological approaches have been developed. This review focuses on ICKs designed with the most impactful cytokines in the cancer field: IL-2, TNFα, IL-10, IL-12, IL-15, IL-21, IFNγ, GM-CSF, and IFNα. An overview of the pharmacological effects of the naked cytokines and ICKs tested for cancer therapy is detailed. A particular emphasis is given on the immunomodulatory effects of ICKs associated with their technological design. In conclusion, this review highlights active ways of development of ICKs. Their already promising results observed in clinical trials are likely to be improved with the advances in targeting technologies such as cytokine/linker engineering and the design of multispecific antibodies with tumor targeting and immunostimulatory functional properties.
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
| | - Hélène Poinot
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Translational Research Centre in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Aurélien Pommier
- UMR1240 Imagerie Moléculaire et Stratégies Théranostiques INSERM, Université Clermont Auvergne, BP 184, F-63005 Clermont-Ferrand, France
| |
Collapse
|
7
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
8
|
Minnie SA, Waltner OG, Zhang P, Takahashi S, Nemychenkov NS, Ensbey KS, Schmidt CR, Legg SRW, Comstock M, Boiko JR, Nelson E, Bhise SS, Wilkens AB, Koyama M, Dhodapkar MV, Chesi M, Riddell SR, Green DJ, Spencer A, Furlan SN, Hill GR. TIM-3 + CD8 T cells with a terminally exhausted phenotype retain functional capacity in hematological malignancies. Sci Immunol 2024; 9:eadg1094. [PMID: 38640253 PMCID: PMC11093588 DOI: 10.1126/sciimmunol.adg1094] [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: 12/02/2022] [Accepted: 03/27/2024] [Indexed: 04/21/2024]
Abstract
Chronic antigen stimulation is thought to generate dysfunctional CD8 T cells. Here, we identify a CD8 T cell subset in the bone marrow tumor microenvironment that, despite an apparent terminally exhausted phenotype (TPHEX), expressed granzymes, perforin, and IFN-γ. Concurrent gene expression and DNA accessibility revealed that genes encoding these functional proteins correlated with BATF expression and motif accessibility. IFN-γ+ TPHEX effectively killed myeloma with comparable efficacy to transitory effectors, and disease progression correlated with numerical deficits in IFN-γ+ TPHEX. We also observed IFN-γ+ TPHEX within CD19-targeted chimeric antigen receptor T cells, which killed CD19+ leukemia cells. An IFN-γ+ TPHEX gene signature was recapitulated in TEX cells from human cancers, including myeloma and lymphoma. Here, we characterize a TEX subset in hematological malignancies that paradoxically retains function and is distinct from dysfunctional TEX found in chronic viral infections. Thus, IFN-γ+ TPHEX represent a potential target for immunotherapy of blood cancers.
Collapse
Affiliation(s)
- Simone A. Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Olivia G. Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Ping Zhang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Shuichiro Takahashi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Nicole S. Nemychenkov
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Kathleen S. Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Christine R. Schmidt
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Samuel RW. Legg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Melissa Comstock
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Julie R. Boiko
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Department of Pediatrics, University of Washington; WA, UNITED STATES
| | - Ethan Nelson
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Shruti S. Bhise
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Alec B. Wilkens
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
| | - Madhav V. Dhodapkar
- Department of Hematology/Medical Oncology, Atlanta, GA, UNITED STATES
- Winship Cancer Institute, Emory University, Atlanta, GA, UNITED STATES
| | - Marta Chesi
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, UNITED STATES
| | - Stanley R. Riddell
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Division of Medical Oncology, University of Washington; Seattle, WA, UNITED STATES
| | - Damian J. Green
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Division of Medical Oncology, University of Washington; Seattle, WA, UNITED STATES
| | - Andrew Spencer
- Australian Center for Blood Diseases, Monash University/The Alfred Hospital, Melbourne, VIC, AUSTRALIA
- Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital, Melbourne, VIC, AUSTRALIA
- Department of Clinical Haematology, Monash University, Melbourne, VIC
| | - Scott N. Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Department of Pediatrics, University of Washington; WA, UNITED STATES
| | - Geoffrey R. Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center; Seattle, WA, UNITED STATES
- Division of Medical Oncology, University of Washington; Seattle, WA, UNITED STATES
| |
Collapse
|
9
|
Flippot R, Teixeira M, Rey-Cardenas M, Carril-Ajuria L, Rainho L, Naoun N, Jouniaux JM, Boselli L, Naigeon M, Danlos FX, Escudier B, Scoazec JY, Cassard L, Albiges L, Chaput N. B cells and the coordination of immune checkpoint inhibitor response in patients with solid tumors. J Immunother Cancer 2024; 12:e008636. [PMID: 38631710 PMCID: PMC11029261 DOI: 10.1136/jitc-2023-008636] [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] [Accepted: 03/31/2024] [Indexed: 04/19/2024] Open
Abstract
Immunotherapy profoundly changed the landscape of cancer therapy by providing long-lasting responses in subsets of patients and is now the standard of care in several solid tumor types. However, immunotherapy activity beyond conventional immune checkpoint inhibition is plateauing, and biomarkers are overall lacking to guide treatment selection. Most studies have focused on T cell engagement and response, but there is a growing evidence that B cells may be key players in the establishment of an organized immune response, notably through tertiary lymphoid structures. Mechanisms of B cell response include antibody-dependent cellular cytotoxicity and phagocytosis, promotion of CD4+ and CD8+ T cell activation, maintenance of antitumor immune memory. In several solid tumor types, higher levels of B cells, specific B cell subpopulations, or the presence of tertiary lymphoid structures have been associated with improved outcomes on immune checkpoint inhibitors. The fate of B cell subpopulations may be widely influenced by the cytokine milieu, with versatile roles for B-specific cytokines B cell activating factor and B cell attracting chemokine-1/CXCL13, and a master regulatory role for IL-10. Roles of B cell-specific immune checkpoints such as TIM-1 are emerging and could represent potential therapeutic targets. Overall, the expanding field of B cells in solid tumors of holds promise for the improvement of current immunotherapy strategies and patient selection.
Collapse
Affiliation(s)
- Ronan Flippot
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Marcus Teixeira
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Macarena Rey-Cardenas
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Lucia Carril-Ajuria
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
- Medical Oncology, CHU Brugmann, Brussels, Belgium
| | - Larissa Rainho
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Natacha Naoun
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Jean-Mehdi Jouniaux
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Lisa Boselli
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Marie Naigeon
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Francois-Xavier Danlos
- LRTI, INSERM U1015, Gustave Roussy, Villejuif, France
- Drug Development Department, Gustave Roussy, Villejuif, France
| | - Bernard Escudier
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | | | - Lydie Cassard
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Nathalie Chaput
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
10
|
Wang M, Chen S, He X, Yuan Y, Wei X. Targeting inflammation as cancer therapy. J Hematol Oncol 2024; 17:13. [PMID: 38520006 PMCID: PMC10960486 DOI: 10.1186/s13045-024-01528-7] [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: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 03/25/2024] Open
Abstract
Inflammation has accompanied human beings since the emergence of wounds and infections. In the past decades, numerous efforts have been undertaken to explore the potential role of inflammation in cancer, from tumor development, invasion, and metastasis to the resistance of tumors to treatment. Inflammation-targeted agents not only demonstrate the potential to suppress cancer development, but also to improve the efficacy of other therapeutic modalities. In this review, we describe the highly dynamic and complex inflammatory tumor microenvironment, with discussion on key inflammation mediators in cancer including inflammatory cells, inflammatory cytokines, and their downstream intracellular pathways. In addition, we especially address the role of inflammation in cancer development and highlight the action mechanisms of inflammation-targeted therapies in antitumor response. Finally, we summarize the results from both preclinical and clinical studies up to date to illustrate the translation potential of inflammation-targeted therapies.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Siyuan Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
11
|
Pu K, Feng Y, Tang Q, Yang G, Xu C. Review of dietary patterns and gastric cancer risk: epidemiology and biological evidence. Front Oncol 2024; 14:1333623. [PMID: 38444674 PMCID: PMC10912593 DOI: 10.3389/fonc.2024.1333623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Due to rapid research expansion on dietary factors and development of cancer prevention guidelines, the field of dietary pattern and its relationship to cancer risk has gained more focus. Numerous epidemiology studies have reported associations between Gastric Cancer (GC) and both data-driven posteriori dietary pattern and priori dietary pattern defined by predetermined dietary indexes. As dietary patterns have evolved, a series of patterns based on biological markers has advanced, offering deeper insights into the relationship between diet and the risk of cancer. Although researches on dietary patterns and cancer risk are booming, there is limited body of literature focusing specifically on GC. In this study, we compare the similarities and differences among the specific components of dietary patterns and indices, summarize current state of knowledge regarding dietary patterns related to GC and illustrate their potential mechanisms for GC prevention. In conclusion, we offer suggestions for future research based on the emerging themes within this rapidly evolving field.
Collapse
Affiliation(s)
- Ke Pu
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yang Feng
- Department of Neurosurgery, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
| | - Qian Tang
- Statesboro Office, Southeast Medical Group, Atlanta, GA, United States
| | - Guodong Yang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Chuan Xu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Pei Y, Cui X, Wang Y. Regulation of IL-10 expression and function by JAK-STAT in CD8 + T cells. Int Immunopharmacol 2024; 128:111563. [PMID: 38246002 DOI: 10.1016/j.intimp.2024.111563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024]
Abstract
IL-10 is a pleiotropic cytokine that plays a significant role in antiviral and antitumor immunity. Potent CD8+ T cells express IL-10 after stimulation by strong TCR signaling, which promotes the killing effect of CD8+ T cells. However, the regulation of IL-10 expression in CD8+ T cells and its signaling pathway to enhance CD8+ T cell function are largely unknown. In this study, we investigated the JAK-STAT signaling molecules that regulate IL-10 expression in CD8+ T cells and the JAK-STAT signaling pathway that IL-10 enhances the function of CD8+ T cells through its receptor, using small molecule inhibitors and CRISPR-Cas9 gene editing. Our findings provide new insights and a theoretical basis for the immunotherapy of tumors.
Collapse
Affiliation(s)
- Yu Pei
- Life Science Institute, Jinzhou Medical University, Jinzhou, China; Department of Clinical Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiuping Cui
- Life Science Institute, Jinzhou Medical University, Jinzhou, China
| | - Yu Wang
- Life Science Institute, Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
13
|
Filosto S, Vardhanabhuti S, Canales MA, Poiré X, Lekakis LJ, de Vos S, Portell CA, Wang Z, To C, Schupp M, Poddar S, Trinh T, Warren CM, Aguilar EG, Budka J, Cheng P, Chou J, Bot A, Shen RR, Westin JR. Product Attributes of CAR T-cell Therapy Differentially Associate with Efficacy and Toxicity in Second-line Large B-cell Lymphoma (ZUMA-7). Blood Cancer Discov 2024; 5:21-33. [PMID: 37983485 PMCID: PMC10772511 DOI: 10.1158/2643-3230.bcd-23-0112] [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: 06/30/2023] [Revised: 10/05/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023] Open
Abstract
Treatment resistance and toxicities remain a risk following chimeric antigen receptor (CAR) T-cell therapy. Herein, we report pharmacokinetics, pharmacodynamics, and product and apheresis attributes associated with outcomes among patients with relapsed/refractory large B-cell lymphoma (LBCL) treated with axicabtagene ciloleucel (axi-cel) in ZUMA-7. Axi-cel peak expansion associated with clinical response and toxicity, but not response durability. In apheresis material and final product, a naive T-cell phenotype (CCR7+CD45RA+) expressing CD27 and CD28 associated with improved response durability, event-free survival, progression-free survival, and a lower number of prior therapies. This phenotype was not associated with high-grade cytokine release syndrome (CRS) or neurologic events. Higher baseline and postinfusion levels of serum inflammatory markers associated with differentiated/effector products, reduced efficacy, and increased CRS and neurologic events, thus suggesting targets for intervention. These data support better outcomes with earlier CAR T-cell intervention and may improve patient care by informing on predictive biomarkers and development of next-generation products. SIGNIFICANCE In ZUMA-7, the largest randomized CAR T-cell trial in LBCL, a naive T-cell product phenotype (CCR7+CD45RA+) expressing CD27 and CD28 associated with improved efficacy, decreased toxicity, and a lower number of prior therapies, supporting earlier intervention with CAR T-cell therapy. In addition, targets for improvement of therapeutic index are proposed. This article is featured in Selected Articles from This Issue, p. 4.
Collapse
Affiliation(s)
| | | | | | - Xavier Poiré
- Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Lazaros J. Lekakis
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, Florida
| | - Sven de Vos
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | | | - Zixing Wang
- Kite, a Gilead Company, Santa Monica, California
| | - Christina To
- Kite, a Gilead Company, Santa Monica, California
| | - Marco Schupp
- Kite, a Gilead Company, Santa Monica, California
| | | | - Tan Trinh
- Kite, a Gilead Company, Santa Monica, California
| | | | | | - Justin Budka
- Kite, a Gilead Company, Santa Monica, California
| | - Paul Cheng
- Kite, a Gilead Company, Santa Monica, California
| | - Justin Chou
- Kite, a Gilead Company, Santa Monica, California
| | - Adrian Bot
- Kite, a Gilead Company, Santa Monica, California
| | | | - Jason R. Westin
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
14
|
Chang CC, Yang CH, Chuang CH, Jiang SJ, Hwang YM, Liou JW, Hsu HJ. A peptide derived from interleukin-10 exhibits potential anticancer activity and can facilitate cell targeting of gold nanoparticles loaded with anticancer therapeutics. Commun Chem 2023; 6:278. [PMID: 38102207 PMCID: PMC10724200 DOI: 10.1038/s42004-023-01079-x] [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: 05/22/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023] Open
Abstract
Human interleukin-10 (IL-10) is an immunosuppressive and anti-inflammatory cytokine, and its expression is upregulated in tumor tissues and serum samples of patients with various cancers. Because of its immunosuppressive nature, IL-10 has also been suggested to be a factor leading to tumor cells' evasion of immune surveillance and clearance by the host immune system. In this study, we refined a peptide with 20 amino acids, named NK20a, derived from the binding region of IL-10 on the basis of in silico analysis of the complex structure of IL-10 with IL-10Ra, the ligand binding subunit of the IL-10 receptor. The binding ability of the peptide was confirmed through in vitro biophysical biolayer interferometry and cellular experiments. The IL-10 inhibitory peptide exerted anticancer effects on lymphoma B cells and could abolish the suppression effect of IL-10 on macrophages. NK20a was also conjugated with gold nanoparticles to target the chemotherapeutic 5-fluorouracil (5-FU)-loaded nanoparticles to enhance the anticancer efficacy of 5-FU against the breast cancer cell line BT-474. Our study demonstrated that NK20a designed in silico with improved binding affinity to the IL-10 receptor can be used as a tool in developing anticancer strategies.
Collapse
Affiliation(s)
- Chun-Chun Chang
- Department of Laboratory Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan, ROC
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Chin-Hao Yang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Chin-Hsien Chuang
- Department of Biomedical Sciences and Engineering, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Shinn-Jong Jiang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Yin-Min Hwang
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Je-Wen Liou
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC.
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC.
| | - Hao-Jen Hsu
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC.
- Department of Biomedical Sciences and Engineering, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC.
| |
Collapse
|
15
|
Chen P, Paraiso WKD, Cabral H. Revitalizing Cytokine-Based Cancer Immunotherapy through Advanced Delivery Systems. Macromol Biosci 2023; 23:e2300275. [PMID: 37565723 DOI: 10.1002/mabi.202300275] [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: 06/14/2023] [Revised: 08/04/2023] [Indexed: 08/12/2023]
Abstract
Cytokines can coordinate robust immune responses, holding great promise as therapeutics against infections, autoimmune diseases, and cancers. In cancer treatment, numerous pro-inflammatory cytokines have displayed promising efficacy in preclinical studies. However, their clinical application is hindered by poor pharmacokinetics, significant toxicity and unsatisfactory anticancer efficacy. Thus, while IFN-α and IL-2 are approved for specific cancer treatments, other cytokines still remain subject of intense investigation. To accelerate the application of cytokines as cancer immunotherapeutics, strategies need to be directed to improve their safety and anticancer performance. In this regard, delivery systems could be used to generate innovative therapies by targeting the cytokines or nucleic acids, such as DNA and mRNA, encoding the cytokines to tumor tissues. This review centers on these innovative delivery strategies for cytokines, summarizing key approaches, such as gene delivery and protein delivery, and critically examining their potential and challenges for clinical translation.
Collapse
Affiliation(s)
- Pengwen Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | | | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| |
Collapse
|
16
|
de Souza Teixeira AA, Biondo L, Silveira LS, Lima EA, Diniz TA, Lira FS, Seelaender M, Rosa Neto JC. Exercise training induces alteration of clock genes and myokines expression in tumor-bearing mice. Cell Biochem Funct 2023; 41:1383-1394. [PMID: 37877577 DOI: 10.1002/cbf.3872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/23/2023] [Accepted: 10/02/2023] [Indexed: 10/26/2023]
Abstract
To investigate the impact of different exercise training schedules (following a fixed schedule or at random times of the day) on clock genes and myokine expression patterns in the skeletal muscle of tumor-bearing mice. Mice were divided into three groups: tumor (LLC), tumor + exercise training (LLC + T) always performed at the same time of the day (ZT2) and exercise training at random times of the day (ZTAlt). Mice were inoculated subcutaneously with Lewis lung carcinoma cells. The gastrocnemius muscle was dissected and the clock gene expression (Clock/Per1/Per2/Per3/Rev-Erbα/GAPDH) was investigated by quantitative reverse transcription polymerase chain reaction with SYBR® Green. Myokine content in muscle (tumour necrosis factor alpha/IL-10/IL-4) was assessed by enzyme-linked immunosorbent assay. At the end of the protocol, the trained groups showed a reduction in total weight, when compared to Lewis lung carcinoma. Tumor weight was lower in the LLC + T (ZTAlt), when compared to LLC. Clock gene mRNA expression showed a significant increase for ZT20 in the groups that performed physical exercise at LLC + T (ZTAlt), when compared with LLC. The Per family showed increased mRNA expression in ZT4 in both trained mice groups, when compared with LLC. LLC + T (ZTAlt) presented reduction of the expression of anti-inflammatory myokines (Il-10/IL-4) during the night, compared with LLC + T(ZT2). Exercise training is able to induce marked modification of clock gene expression and of the production of myokines, in a way that is dependent on schedule exercise training strategy. Taken together, the results show that exercise is a potent Zeitgeber and may thus contribute to change clock genes expression and myokines that are able to reduce the tumor weight.
Collapse
Affiliation(s)
- Alexandre Abilio de Souza Teixeira
- Department of Cell and Developmental Biology, Immunometabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Luana Biondo
- Department of Cell and Developmental Biology, Immunometabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Loreana Sanches Silveira
- Department of Cell and Developmental Biology, Immunometabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Edson A Lima
- Department of Cell and Developmental Biology, Immunometabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Tiego A Diniz
- Department of Cell and Developmental Biology, Immunometabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Fabio Santos Lira
- Department of Physical Education, Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil
- CIDAF, University of Coimbra, Coimbra, Portugal
| | - Marilia Seelaender
- Department of Surgery and LIM26 HC-USP, Cancer Metabolism Research Group, University of São Paulo, São Paulo, Brazil
| | - José Cesar Rosa Neto
- Department of Cell and Developmental Biology, Immunometabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| |
Collapse
|
17
|
Canaria DA, Rodriguez JA, Wang L, Yeo FJ, Yan B, Wang M, Campbell C, Kazemian M, Olson MR. Tox induces T cell IL-10 production in a BATF-dependent manner. Front Immunol 2023; 14:1275423. [PMID: 38054003 PMCID: PMC10694202 DOI: 10.3389/fimmu.2023.1275423] [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: 08/10/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Tox is a member of the high mobility group (HMG)-Box transcription factors and plays important roles in thymic T cell development. Outside of the thymus, however, Tox is also highly expressed by CD8 and CD4 T cells in various states of activation and in settings of cancer and autoimmune disease. In CD4 T cells, Tox has been primarily studied in T follicular helper (TFH) cells where it, along with Tox2, promotes TFH differentiation by regulating key TFH-associated genes and suppressing CD4 cytotoxic T cell differentiation. However, the role of Tox in other T helper (Th) cell subtypes is less clear. Here, we show that Tox is expressed in several physiologically-activated Th subtypes and its ectopic expression enhances the in vitro differentiation of Th2 and T regulatory (Treg) cells. Tox overexpression in unpolarized Th cells also induced the expression of several genes involved in cell activation (Pdcd1), cellular trafficking (Ccl3, Ccl4, Xcl1) and suppressing inflammation (Il10) across multiple Th subtypes. We found that Tox binds the regulatory regions of these genes along with the transcription factors BATF, IRF4, and JunB and that Tox-induced expression of IL-10, but not PD-1, is BATF-dependent. Based on these data, we propose a model where Tox regulates Th cell chemotactic genes involved in facilitating dendritic cell-T cell interactions and aids in the resolution or prevention of inflammation through the production of IL-10.
Collapse
Affiliation(s)
- D. Alejandro Canaria
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | | | - Luopin Wang
- Department of Computer Science, Purdue University, West Lafayette, IN, United States
| | - Franklin J. Yeo
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Bingyu Yan
- Department of Biochemistry, Purdue University, West Lafayette, IN, United States
| | - Mengbo Wang
- Department of Computer Science, Purdue University, West Lafayette, IN, United States
| | - Charlotte Campbell
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Majid Kazemian
- Department of Computer Science, Purdue University, West Lafayette, IN, United States
| | - Matthew R. Olson
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
18
|
Raman V, Howell LM, Bloom SMK, Hall CL, Wetherby VE, Minter LM, Kulkarni AA, Forbes NS. Intracellular Salmonella delivery of an exogenous immunization antigen refocuses CD8 T cells against cancer cells, eliminates pancreatic tumors and forms antitumor immunity. Front Immunol 2023; 14:1228532. [PMID: 37868996 PMCID: PMC10585021 DOI: 10.3389/fimmu.2023.1228532] [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: 05/24/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Immunotherapies have shown great promise, but are not effective for all tumors types and are effective in less than 3% of patients with pancreatic ductal adenocarcinomas (PDAC). To make an immune treatment that is effective for more cancer patients and those with PDAC specifically, we genetically engineered Salmonella to deliver exogenous antigens directly into the cytoplasm of tumor cells. We hypothesized that intracellular delivery of an exogenous immunization antigen would activate antigen-specific CD8 T cells and reduce tumors in immunized mice. Methods To test this hypothesis, we administered intracellular delivering (ID) Salmonella that deliver ovalbumin as a model antigen into tumor-bearing, ovalbumin-vaccinated mice. ID Salmonella delivers antigens by autonomously lysing in cells after the induction of cell invasion. Results We showed that the delivered ovalbumin disperses throughout the cytoplasm of cells in culture and in tumors. This delivery into the cytoplasm is essential for antigen cross-presentation. We showed that co-culture of ovalbumin-recipient cancer cells with ovalbumin-specific CD8 T cells triggered a cytotoxic T cell response. After the adoptive transfer of OT-I CD8 T cells, intracellular delivery of ovalbumin reduced tumor growth and eliminated tumors. This effect was dependent on the presence of the ovalbumin-specific T cells. Following vaccination with the exogenous antigen in mice, intracellular delivery of the antigen cleared 43% of established KPC pancreatic tumors, increased survival, and prevented tumor re-implantation. Discussion This response in the immunosuppressive KPC model demonstrates the potential to treat tumors that do not respond to checkpoint inhibitors, and the response to re-challenge indicates that new immunity was established against intrinsic tumor antigens. In the clinic, ID Salmonella could be used to deliver a protein antigen from a childhood immunization to refocus pre-existing T cell immunity against tumors. As an off-the-shelf immunotherapy, this bacterial system has the potential to be effective in a broad range of cancer patients.
Collapse
Affiliation(s)
- Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
- Ernest Pharmaceuticals, LLC, Hadley, MA, United States
| | - Lars M. Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
| | - Shoshana M. K. Bloom
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
| | - Christopher L. Hall
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
- Ernest Pharmaceuticals, LLC, Hadley, MA, United States
| | | | - Lisa M. Minter
- Molecular and Cell Biology Program, University of Massachusetts, Amherst, MA, United States
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
- Institute for Applied Life Science, University of Massachusetts, Amherst, MA, United States
| | - Ashish A. Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
- Institute for Applied Life Science, University of Massachusetts, Amherst, MA, United States
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
- Molecular and Cell Biology Program, University of Massachusetts, Amherst, MA, United States
- Institute for Applied Life Science, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
19
|
Zhang C, Cao K, Yang M, Wang Y, He M, Lu J, Huang Y, Zhang G, Liu H. C5aR1 blockade reshapes immunosuppressive tumor microenvironment and synergizes with immune checkpoint blockade therapy in high-grade serous ovarian cancer. Oncoimmunology 2023; 12:2261242. [PMID: 37791232 PMCID: PMC10543342 DOI: 10.1080/2162402x.2023.2261242] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/15/2023] [Indexed: 10/05/2023] Open
Abstract
High-grade serous ovarian cancer (HGSC), with a modest response to immune checkpoint blockade (ICB) targeting PD-1/PD-L1 monotherapy, is densely infiltrated by M2-polarized tumor-associated macrophages (TAMs) and regulatory T (Treg) cells. The complement C5a/C5aR1 axis contributes to the programming of the immunosuppressive phenotype of TAMs in solid tumors and represents a promising immunomodulatory target for treating HGSCs. Here, we aimed to identify the relevance of C5aR1 in prognosis, immune microenvironment, and immunotherapy response in HGSCs. The expression and relationship of C5aR1 with tumor-infiltrating immune cells were assessed by immunohistochemistry and flow cytometry in the training cohort (n = 120) and fresh HGSC tissues (n = 36). Transcriptomic analyses of the xenografts delineated the mechanisms driving the immunomodulatory activity of PMX53, an orally bioavailable C5aR1 inhibitor. Therapeutic relevance was confirmed in ex vivo tumor cultures and The Cancer Genome Atlas (TCGA) datasets. C5aR1 expression independently predicted dismal prognosis and was linked to the immunoevasive subtype of HGSC, characterized by increased infiltration of pro-tumor cells (Treg cells, M2-polarized macrophages, and neutrophils) and impaired CD8+T functions. PMX53 antagonized subcutaneous tumor growth, modulated immunosuppressive mechanisms and synergized with aPD-1 in several tumor types. Single-cell RNA-seq analysis revealed predominant C5aR1 expression in TAMs, with an immunosuppressive-related expression signature in C5aR1+TAMs. Furthermore, the combination of C5aR1 and PD-L1 was associated with specific molecular characteristics and matched clinical response annotations. Therefore, the abundance of C5aR1 could predict an inferior prognosis in HGSCs, and incorporating PD-L1 may serve as a novel predictive biomarker to guide therapeutic options.
Collapse
Affiliation(s)
- Chen Zhang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Kankan Cao
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Moran Yang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yiying Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Mengdi He
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jiaqi Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yan Huang
- Department of Gynecologic Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Guodong Zhang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Haiou Liu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Salkeni MA, Naing A. Interleukin-10 in cancer immunotherapy: from bench to bedside. Trends Cancer 2023; 9:716-725. [PMID: 37321942 PMCID: PMC10524969 DOI: 10.1016/j.trecan.2023.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023]
Abstract
Interleukin (IL)-10 was one of the first cytokines to be recognized. However, its functionality in promoting antitumor immunity was described more recently. Context- and concentration-dependent biological effects are the hallmarks of the pleiotropic role of IL-10. Despite reducing tumor-promoting inflammation, IL-10 may have a role in rejuvenating exhausted tumor-resident T cells. Contrary to the assumption that IL-10 produces an immunosuppressive tumor microenvironment (TME), it promotes activation of tumor-resident CD8+ T cells, which aids tumor rejection. Emerging data from published early-Phase trials have shown mixed results in different tumor types. In this review, we summarize the biological effects of IL-10 and highlight the clinical experience using pegilodecakin.
Collapse
Affiliation(s)
- Mohamad Adham Salkeni
- Developmental Therapeutics Clinic, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
21
|
Murillo N, Lasso P, Urueña C, Pardo-Rodriguez D, Ballesteros-Ramírez R, Betancourt G, Rojas L, Cala MP, Fiorentino S. Petiveria alliacea Reduces Tumor Burden and Metastasis and Regulates the Peripheral Immune Response in a Murine Myeloid Leukemia Model. Int J Mol Sci 2023; 24:12972. [PMID: 37629156 PMCID: PMC10454792 DOI: 10.3390/ijms241612972] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
The poor response, adverse effects and drug resistance to treatment of acute myeloid leukemia (AML) have led to searching for safer and more effective therapeutic alternatives. We previously demonstrated that the alcoholic extract of Petiveria alliacea (Esperanza) has a significant in vitro antitumor effect on other tumor cells and also the ability to regulate energy metabolism. We evaluated the effect of the Esperanza extract in vitro and in vivo in a murine model of AML with DA-3/ER-GM cells. First, a chemical characterization of the extract was conducted through liquid and gas chromatography coupled with mass spectrometry. In vitro findings showed that the extract modulates tumor metabolism by decreasing glucose uptake and increasing reactive oxygen species, which leads to a reduction in cell proliferation. Then, to evaluate the effect of the extract in vivo, we standardized the mouse model by injecting DA-3/ER-GM cells intravenously. The animals treated with the extract showed a lower percentage of circulating blasts, higher values of hemoglobin, hematocrit, and platelets, less infiltration of blasts in the spleen, and greater production of cytokines compared to the control group. These results suggest that the antitumor activity of this extract on DA-3/ER-GM cells can be attributed to the decrease in glycolytic metabolism, its activity as a mitocan, and the possible immunomodulatory effect by reducing tumor proliferation and metastasis.
Collapse
Affiliation(s)
- Natalia Murillo
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá 110211, Colombia; (N.M.); (P.L.); (C.U.); (R.B.-R.); (G.B.); (L.R.)
| | - Paola Lasso
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá 110211, Colombia; (N.M.); (P.L.); (C.U.); (R.B.-R.); (G.B.); (L.R.)
| | - Claudia Urueña
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá 110211, Colombia; (N.M.); (P.L.); (C.U.); (R.B.-R.); (G.B.); (L.R.)
| | - Daniel Pardo-Rodriguez
- Metabolomics Core Facility—MetCore, Vicepresidency for Research, Universidad de Los Andes, Bogotá 111711, Colombia; (D.P.-R.); (M.P.C.)
| | - Ricardo Ballesteros-Ramírez
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá 110211, Colombia; (N.M.); (P.L.); (C.U.); (R.B.-R.); (G.B.); (L.R.)
| | - Giselle Betancourt
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá 110211, Colombia; (N.M.); (P.L.); (C.U.); (R.B.-R.); (G.B.); (L.R.)
| | - Laura Rojas
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá 110211, Colombia; (N.M.); (P.L.); (C.U.); (R.B.-R.); (G.B.); (L.R.)
| | - Mónica P. Cala
- Metabolomics Core Facility—MetCore, Vicepresidency for Research, Universidad de Los Andes, Bogotá 111711, Colombia; (D.P.-R.); (M.P.C.)
| | - Susana Fiorentino
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá 110211, Colombia; (N.M.); (P.L.); (C.U.); (R.B.-R.); (G.B.); (L.R.)
| |
Collapse
|
22
|
Chang YW, Hsiao HW, Chen JP, Tzeng SF, Tsai CH, Wu CY, Hsieh HH, Carmona SJ, Andreatta M, Di Conza G, Su MT, Koni PA, Ho PC, Chen HK, Yang MH. A CSF-1R-blocking antibody/IL-10 fusion protein increases anti-tumor immunity by effectuating tumor-resident CD8 + T cells. Cell Rep Med 2023; 4:101154. [PMID: 37586318 PMCID: PMC10439276 DOI: 10.1016/j.xcrm.2023.101154] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/04/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
Strategies to increase intratumoral concentrations of an anticancer agent are desirable to optimize its therapeutic potential when said agent is efficacious primarily within a tumor but also have significant systemic side effects. Here, we generate a bifunctional protein by fusing interleukin-10 (IL-10) to a colony-stimulating factor-1 receptor (CSF-1R)-blocking antibody. The fusion protein demonstrates significant antitumor activity in multiple cancer models, especially head and neck cancer. Moreover, this bifunctional protein not only leads to the anticipated reduction in tumor-associated macrophages but also triggers proliferation, activation, and metabolic reprogramming of CD8+ T cells. Furthermore, it extends the clonotype diversity of tumor-infiltrated T cells and shifts the tumor microenvironment (TME) to an immune-active state. This study suggests an efficient strategy for designing immunotherapeutic agents by fusing a potent immunostimulatory molecule to an antibody targeting TME-enriched factors.
Collapse
Affiliation(s)
- Yao-Wen Chang
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | | | - Ju-Pei Chen
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Sheue-Fen Tzeng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11221, Taiwan
| | - Chin-Hsien Tsai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11221, Taiwan
| | - Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Hsin-Hua Hsieh
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Santiago J Carmona
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Massimo Andreatta
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Giusy Di Conza
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Mei-Tzu Su
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | | | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Hung-Kai Chen
- Elixiron Immunotherapeutics (Hong Kong) Ltd., Hong Kong.
| | - Muh-Hwa Yang
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan; Department of Teaching and Research, Taipei City Hospital, Taipei, Taiwan.
| |
Collapse
|
23
|
Dickerson LK, Carter JA, Kohli K, Pillarisetty VG. Emerging interleukin targets in the tumour microenvironment: implications for the treatment of gastrointestinal tumours. Gut 2023; 72:1592-1606. [PMID: 37258094 DOI: 10.1136/gutjnl-2023-329650] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
The effectiveness of antitumour immunity is dependent on intricate cytokine networks. Interleukins (ILs) are important mediators of complex interactions within the tumour microenvironment, including regulation of tumour-infiltrating lymphocyte proliferation, differentiation, migration and activation. Our evolving and increasingly nuanced understanding of the cell type-specific and heterogeneous effects of IL signalling has presented unique opportunities to fine-tune elaborate IL networks and engineer new targeted immunotherapeutics. In this review, we provide a primer for clinicians on the challenges and potential of IL-based treatment. We specifically detail the roles of IL-2, IL-10, IL-12 and IL-15 in shaping the tumour-immune landscape of gastrointestinal malignancies, paying particular attention to promising preclinical findings, early-stage clinical research and innovative therapeutic approaches that may properly place ILs to the forefront of immunotherapy regimens.
Collapse
Affiliation(s)
| | - Jason A Carter
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
| | - Karan Kohli
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
- Flatiron Bio, Palo Alto, California, USA
| | - Venu G Pillarisetty
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
| |
Collapse
|
24
|
Fu Y, Tang R, Zhao X. Engineering cytokines for cancer immunotherapy: a systematic review. Front Immunol 2023; 14:1218082. [PMID: 37483629 PMCID: PMC10357296 DOI: 10.3389/fimmu.2023.1218082] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Cytokines are pivotal mediators of cell communication in the tumor microenvironment. Multiple cytokines are involved in the host antitumor response, but the production and function of these cytokines are usually dysregulated during malignant tumor progression. Considering their clinical potential and the early successful use of cytokines in cancer immunotherapy, such as interferon alpha-2b (IFNα-2b; IntronA®) and IL-2 (Proleukin®), cytokine-based therapeutics have been extensively evaluated in many follow-up clinical trials. Following these initial breakthroughs, however, clinical translation of these natural messenger molecules has been greatly limited owing to their high-degree pleiotropic features and complex biological properties in many cell types. These characteristics, coupled with poor pharmacokinetics (a short half-life), have hampered the delivery of cytokines via systemic administration, particularly because of severe dose-limiting toxicities. New engineering approaches have been developed to widen the therapeutic window, prolong pharmacokinetic effects, enhance tumor targeting and reduce adverse effects, thereby improving therapeutic efficacy. In this review, we focus on the recent progress and competitive landscape in cytokine engineering strategies and preclinical/clinical therapeutics for cancer. In addition, aiming to promote engineered cytokine-based cancer immunotherapy, we present a profound discussion about the feasibility of recently developed methods in clinical medicine translation.
Collapse
Affiliation(s)
- Yong Fu
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
- Jiangsu Simcere Pharmaceutical Co, Ltd., Nanjing, China
| | - Renhong Tang
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
- Simcere Zaiming Pharmaceutical Co, Ltd., Nanjing, China
| | - Xiaofeng Zhao
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
- Jiangsu Simcere Pharmaceutical Co, Ltd., Nanjing, China
| |
Collapse
|
25
|
Shakiba Y, Vorobyev PO, Mahmoud M, Hamad A, Kochetkov DV, Yusubalieva GM, Baklaushev VP, Chumakov PM, Lipatova AV. Recombinant Strains of Oncolytic Vaccinia Virus for Cancer Immunotherapy. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:823-841. [PMID: 37748878 DOI: 10.1134/s000629792306010x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 09/27/2023]
Abstract
Cancer virotherapy is an alternative therapeutic approach based on the viruses that selectively infect and kill tumor cells. Vaccinia virus (VV) is a member of the Poxviridae, a family of enveloped viruses with a large linear double-stranded DNA genome. The proven safety of the VV strains as well as considerable transgene capacity of the viral genome, make VV an excellent platform for creating recombinant oncolytic viruses for cancer therapy. Furthermore, various genetic modifications can increase tumor selectivity and therapeutic efficacy of VV by arming it with the immune-modulatory genes or proapoptotic molecules, boosting the host immune system, and increasing cross-priming recognition of the tumor cells by T-cells or NK cells. In this review, we summarized the data on bioengineering approaches to develop recombinant VV strains for enhanced cancer immunotherapy.
Collapse
Affiliation(s)
- Yasmin Shakiba
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Pavel O Vorobyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Marah Mahmoud
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Azzam Hamad
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Dmitriy V Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Gaukhar M Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Federal Research Clinical Center for Specialized Medical Care and Medical Technologies, Federal Medical-Biological Agency (FMBA), Moscow, 115682, Russia
- Federal Center of Brain Research and Neurotechnologies of the FMBA of Russia, Moscow, 117513, Russia
| | - Vladimir P Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Federal Research Clinical Center for Specialized Medical Care and Medical Technologies, Federal Medical-Biological Agency (FMBA), Moscow, 115682, Russia
- Federal Center of Brain Research and Neurotechnologies of the FMBA of Russia, Moscow, 117513, Russia
| | - Peter M Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | - Anastasia V Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
26
|
Fortunato M, Amodio G, Gregori S. IL-10-Engineered Dendritic Cells Modulate Allogeneic CD8 + T Cell Responses. Int J Mol Sci 2023; 24:9128. [PMID: 37298076 PMCID: PMC10252493 DOI: 10.3390/ijms24119128] [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: 04/17/2023] [Revised: 05/12/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Tolerogenic dendritic cells (tolDC) play a central role in regulating immune homeostasis and in promoting peripheral tolerance. These features render tolDC a promising tool for cell-based approaches aimed at inducing tolerance in T-cell mediated diseases and in allogeneic transplantation. We developed a protocol to generate genetically engineered human tolDC overexpressing IL-10 (DCIL-10) by means of a bidirectional lentiviral vector (LV) encoding for IL-10. DCIL-10 promote allo-specific T regulatory type 1 (Tr1) cells, modulate allogeneic CD4+ T cell responses in vitro and in vivo, and are stable in a pro-inflammatory milieu. In the present study, we investigated the ability of DCIL-10 to modulate cytotoxic CD8+ T cell responses. We demonstrate that DCIL-10 reduces allogeneic CD8+ T cell proliferation and activation in primary mixed lymphocyte reactions (MLR). Moreover, long-term stimulation with DCIL-10 induces allo-specific anergic CD8+ T cells without signs of exhaustion. DCIL-10-primed CD8+ T cells display limited cytotoxic activity. These findings indicate that stable over-expression of IL-10 in human DC leads to a population of cells able to modulate cytotoxic allogeneic CD8+ T cell responses, overall indicating that DCIL-10 represent a promising cellular product for clinical applications aimed at inducing tolerance after transplantation.
Collapse
Affiliation(s)
- Marta Fortunato
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.F.); (G.A.)
- PhD Course in Molecular Medicine, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Giada Amodio
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.F.); (G.A.)
| | - Silvia Gregori
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.F.); (G.A.)
| |
Collapse
|
27
|
Liang Z, Chen W, Guo Y, Ren Y, Tian Y, Cai W, Bao Y, Liu Q, Ding P, Li Y. Soluble monomeric human programmed cell death-ligand 1 inhibits the functions of activated T cells. Front Immunol 2023; 14:1133883. [PMID: 37266424 PMCID: PMC10229872 DOI: 10.3389/fimmu.2023.1133883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/05/2023] [Indexed: 06/03/2023] Open
Abstract
Introduction The presence of soluble human programmed cell death-ligand 1 (shPD-L1) in the blood of patients with cancer has been reported to be negatively correlated with disease prognosis. However, little information exists about the mechanisms underlying high levels of shPD-L1 for promoting disease progression. Methods In this study, we first analyzed the correlations between shPD-L1 and apoptosis of T cells in patients with cancer, then tested the effect of shPD-L1 on T-cell functions and the production of regulatory T cells. Results We found that the apoptosis of human peripheral PD-1+CD4+ T cells was significantly elevated in patients with cancer compared with healthy donors and was positively correlated with circulating PD-L1 levels in patients with cancer. In vitro, monomeric shPD-L1 significantly inhibited the proliferation, cytokine secretion, and cancer cell-killing activity of peripheral blood mononuclear cells (PBMCs) activated by either agonist antibodies or HATac (high-affinity T cell activation core)-NYE (NY-ESO-1 antigen). It also promoted CD4+ T cells to express forkhead family transcription factor 3 (FoxP3) for the conversion of induced T regulatory cells, which was more significant than that mediated by soluble human PD-L1 fusion protein (shPD-L1-Fc). Discussion These results confirm that soluble PD-L1 could be a candidate for inhibiting the functions of activated T cells, promoting peripheral tolerance to tumor cells, and implicating in system tumor immune escape in addition to the tumor microenvironment. This is an important mechanism explaining the negative correlation between peripheral blood PD-L1 levels and cancer prognosis. Therefore, understanding the roles of hPD-L1 in peripheral blood will be helpful for the development of precision immunotherapy programs in treating various tumors.
Collapse
Affiliation(s)
- Zhaoduan Liang
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health GuangDong Laboratory, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Wenfang Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Yunzhuo Guo
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuefei Ren
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ye Tian
- T-cell Immunity Optimized Cure (TIOC) Therapeutics Limited, Hangzhou, Zhejiang, China
| | - Wenxuan Cai
- T-cell Immunity Optimized Cure (TIOC) Therapeutics Limited, Hangzhou, Zhejiang, China
| | - Yifeng Bao
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Qi Liu
- T-cell Immunity Optimized Cure (TIOC) Therapeutics Limited, Hangzhou, Zhejiang, China
| | - Peng Ding
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Yi Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- T-cell Immunity Optimized Cure (TIOC) Therapeutics Limited, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Sun Q, Zhao X, Li R, Liu D, Pan B, Xie B, Chi X, Cai D, Wei P, Xu W, Wei K, Zhao Z, Fu Y, Ni L, Dong C. STAT3 regulates CD8+ T cell differentiation and functions in cancer and acute infection. J Exp Med 2023; 220:e20220686. [PMID: 36688918 PMCID: PMC9884582 DOI: 10.1084/jem.20220686] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 11/05/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
In cancer, persistent antigens drive CD8+ T cell differentiation into exhausted progenitor (Texprog) and terminally exhausted (Texterm) cells. However, how the extrinsic and intrinsic regulatory mechanisms cooperate during this process still remains not well understood. Here, we found that STAT3 signaling plays essential roles in promoting intratumor Texterm cell development by enhancing their effector functions and survival, which results in better tumor control. In tumor microenvironments, STAT3 is predominantly activated by IL-10 and IL-21, but not IL-6. Besides, STAT3 also plays critical roles in the development and function of terminally differentiated effector CD8+ T cells in acute infection. Mechanistically, STAT3 transcriptionally promotes the expression of effector function-related genes, while it suppresses those expressed by the progenitor Tex subset. Moreover, STAT3 functions in collaboration with BATF and IRF4 to mediate chromatin activation at the effector gene loci. Thus, we have elucidated the roles of STAT3 signaling in terminally differentiated CD8+ T cell development, especially in cancer, which benefits the development of more effective immunotherapies against tumors.
Collapse
Affiliation(s)
- Qinli Sun
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Xiaohong Zhao
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Ruifeng Li
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Dingfeng Liu
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Birui Pan
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Bowen Xie
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xinxin Chi
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Dongli Cai
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Peng Wei
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Wei Xu
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Kun Wei
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Zixuan Zhao
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Yujie Fu
- Institute for Immunology, Tsinghua University, Beijing, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Ling Ni
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Chen Dong
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
- Research Unit of Immune Regulation and Immune Diseases of Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
29
|
Cavallazzi Sebold B, Ni G, Li J, Li H, Liu X, Wang T. PEGylated IL-10: Clinical Development in Cancer Immunotherapy, Where to Go? Curr Oncol Rep 2023; 25:115-122. [PMID: 36585961 DOI: 10.1007/s11912-022-01355-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 01/01/2023]
Abstract
PURPOSEOF REVIEW The purpose of this review is to discuss the current understanding of the pegilodecakin (PEGylated interleukin 10) and its role in the inhibition of tumour growth and metastasis. This review also focuses on clinical data published to date that have evaluated the efficacy and safety of pegilodecakin. RECENT FINDINGS Pegilodecakin has shown significant promise in preclinical models, notable for decreased tumour burden and fewer sites of metastatic disease across various malignancies. It has been most widely assessed in a phase I/Ib clinical trial against several solid tumours, leading to the phase II and III clinical trials containing pegilodecakin and its combination with other current treatments. However, the updated data have not shown higher efficacy in renal cell carcinoma, metastatic non-small cell lung cancer or pancreatic cancer, with respect to the controls, yet the adverse events presented more mixed results. Further investigation into combination therapies including pegilodecakin is ongoing. Pegilodecakin showed promise in preclinical and phase I clinical trials on its efficacy in several solid tumours, with expected regulation of IL-10 signalling pathway observed. However, the phase II and III trials did not justify its application as potential immunotherapy in selected cancers. Further evaluation of pegilodecakin's efficacy in other cancers, either as monotherapy or in combination with the current treatments, is worth investigating clinically, which warrants to better understand its potential clinical utility.
Collapse
Affiliation(s)
- Bernardo Cavallazzi Sebold
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.,School of Science, Engineering and Technology, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia
| | - Guoying Ni
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.,The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China.,Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Junjie Li
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Hejie Li
- School of Science, Engineering and Technology, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia
| | - Xiaosong Liu
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China. .,Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China.
| | - Tianfang Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia. .,School of Science, Engineering and Technology, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
| |
Collapse
|
30
|
Sullivan KM, Jiang X, Guha P, Lausted C, Carter JA, Hsu C, Labadie KP, Kohli K, Kenerson HL, Daniel SK, Yan X, Meng C, Abbasi A, Chan M, Seo YD, Park JO, Crispe IN, Yeung RS, Kim TS, Gujral TS, Tian Q, Katz SC, Pillarisetty VG. Blockade of interleukin 10 potentiates antitumour immune function in human colorectal cancer liver metastases. Gut 2023; 72:325-337. [PMID: 35705369 PMCID: PMC9872249 DOI: 10.1136/gutjnl-2021-325808] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 05/25/2022] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Programmed cell death protein 1 (PD-1) checkpoint inhibition and adoptive cellular therapy have had limited success in patients with microsatellite stable colorectal cancer liver metastases (CRLM). We sought to evaluate the effect of interleukin 10 (IL-10) blockade on endogenous T cell and chimeric antigen receptor T (CAR-T) cell antitumour function in CRLM slice cultures. DESIGN We created organotypic slice cultures from human CRLM (n=38 patients' tumours) and tested the antitumour effects of a neutralising antibody against IL-10 (αIL-10) both alone as treatment and in combination with exogenously administered carcinoembryonic antigen (CEA)-specific CAR-T cells. We evaluated slice cultures with single and multiplex immunohistochemistry, in situ hybridisation, single-cell RNA sequencing, reverse-phase protein arrays and time-lapse fluorescent microscopy. RESULTS αIL-10 generated a 1.8-fold increase in T cell-mediated carcinoma cell death in human CRLM slice cultures. αIL-10 significantly increased proportions of CD8+ T cells without exhaustion transcription changes, and increased human leukocyte antigen - DR isotype (HLA-DR) expression of macrophages. The antitumour effects of αIL-10 were reversed by major histocompatibility complex class I or II (MHC-I or MHC-II) blockade, confirming the essential role of antigen presenting cells. Interrupting IL-10 signalling also rescued murine CAR-T cell proliferation and cytotoxicity from myeloid cell-mediated immunosuppression. In human CRLM slices, αIL-10 increased CEA-specific CAR-T cell activation and CAR-T cell-mediated cytotoxicity, with nearly 70% carcinoma cell apoptosis across multiple human tumours. Pretreatment with an IL-10 receptor blocking antibody also potentiated CAR-T function. CONCLUSION Neutralising the effects of IL-10 in human CRLM has therapeutic potential as a stand-alone treatment and to augment the function of adoptively transferred CAR-T cells.
Collapse
Affiliation(s)
- Kevin M Sullivan
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Xiuyun Jiang
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Prajna Guha
- Immuno-Oncology Institute and Department of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA,Department of Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Jason A Carter
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Cynthia Hsu
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Kevin P Labadie
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Karan Kohli
- Department of Surgery, University of Washington, Seattle, Washington, USA,Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Heidi L Kenerson
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Sara K Daniel
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Xiaowei Yan
- Institute for Systems Biology, Seattle, Washington, USA
| | | | - Arezou Abbasi
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Marina Chan
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Y David Seo
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - James O Park
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | | | - Raymond S Yeung
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Teresa S Kim
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Taranjit S Gujral
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Qiang Tian
- Institute for Systems Biology, Seattle, Washington, USA .,National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Steven C Katz
- Immuno-Oncology Institute and Department of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA,Department of Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Venu G Pillarisetty
- Department of Surgery, University of Washington, Seattle, Washington, USA .,Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| |
Collapse
|
31
|
Deckers J, Anbergen T, Hokke AM, de Dreu A, Schrijver DP, de Bruin K, Toner YC, Beldman TJ, Spangler JB, de Greef TFA, Grisoni F, van der Meel R, Joosten LAB, Merkx M, Netea MG, Mulder WJM. Engineering cytokine therapeutics. NATURE REVIEWS BIOENGINEERING 2023; 1:286-303. [PMID: 37064653 PMCID: PMC9933837 DOI: 10.1038/s44222-023-00030-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Cytokines have pivotal roles in immunity, making them attractive as therapeutics for a variety of immune-related disorders. However, the widespread clinical use of cytokines has been limited by their short blood half-lives and severe side effects caused by low specificity and unfavourable biodistribution. Innovations in bioengineering have aided in advancing our knowledge of cytokine biology and yielded new technologies for cytokine engineering. In this Review, we discuss how the development of bioanalytical methods, such as sequencing and high-resolution imaging combined with genetic techniques, have facilitated a better understanding of cytokine biology. We then present an overview of therapeutics arising from cytokine re-engineering, targeting and delivery, mRNA therapeutics and cell therapy. We also highlight the application of these strategies to adjust the immunological imbalance in different immune-mediated disorders, including cancer, infection and autoimmune diseases. Finally, we look ahead to the hurdles that must be overcome before cytokine therapeutics can live up to their full potential.
Collapse
Affiliation(s)
- Jeroen Deckers
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Tom Anbergen
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Ayla M. Hokke
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Anne de Dreu
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - David P. Schrijver
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Koen de Bruin
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Yohana C. Toner
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Thijs J. Beldman
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jamie B. Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Tom F. A. de Greef
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
- Centre for Living Technologies, Alliance Eindhoven University of Technology, Wageningen University & Research, Utrecht University and University Medical Center Utrecht (EWUU), Utrecht, Netherlands
| | - Francesca Grisoni
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Centre for Living Technologies, Alliance Eindhoven University of Technology, Wageningen University & Research, Utrecht University and University Medical Center Utrecht (EWUU), Utrecht, Netherlands
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Present Address: Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Leo A. B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
- Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, Netherlands
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Maarten Merkx
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Present Address: Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
- Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, Netherlands
- Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Willem J. M. Mulder
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Present Address: Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
32
|
Lu M, Lee Y, Lillehoj HS. Evolution of developmental and comparative immunology in poultry: The regulators and the regulated. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 138:104525. [PMID: 36058383 DOI: 10.1016/j.dci.2022.104525] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
Avian has a unique immune system that evolved in response to environmental pressures in all aspects of innate and adaptive immune responses, including localized and circulating lymphocytes, diversity of immunoglobulin repertoire, and various cytokines and chemokines. All of these attributes make birds an indispensable vertebrate model for studying the fundamental immunological concepts and comparative immunology. However, research on the immune system in birds lags far behind that of humans, mice, and other agricultural animal species, and limited immune tools have hindered the adequate application of birds as disease models for mammalian systems. An in-depth understanding of the avian immune system relies on the detailed studies of various regulated and regulatory mediators, such as cell surface antigens, cytokines, and chemokines. Here, we review current knowledge centered on the roles of avian cell surface antigens, cytokines, chemokines, and beyond. Moreover, we provide an update on recent progress in this rapidly developing field of study with respect to the availability of immune reagents that will facilitate the study of regulatory and regulated components of poultry immunity. The new information on avian immunity and available immune tools will benefit avian researchers and evolutionary biologists in conducting fundamental and applied research.
Collapse
Affiliation(s)
- Mingmin Lu
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Youngsub Lee
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| |
Collapse
|
33
|
Huang Y, Jia A, Wang Y, Liu G. CD8 + T cell exhaustion in anti-tumour immunity: The new insights for cancer immunotherapy. Immunology 2023; 168:30-48. [PMID: 36190809 DOI: 10.1111/imm.13588] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/29/2022] [Indexed: 12/27/2022] Open
Abstract
CD8+ T cells play a crucial role in anti-tumour immunity, but they often undergo exhaustion, which affects the anti-tumour activity of CD8+ T cells. The effect and mechanism of exhausted CD8+ T cells have become the focus of anti-tumour immunity research. Recently, a large number of studies have confirmed that long-term antigen exposure can induce exhaustion. Cytokines previously have identified their effects (such as IL-2 and IL-10) may play a dual role in the exhaustion process of CD8+ T cells, suggesting a new mechanism of inducing exhaustion. This review just focuses our current understanding of the biology of exhausted CD8+ T cells, including differentiation pathways, cellular characteristics and signalling pathways involved in inducing exhaustion, and summarizes how these can be applied to tumour immunotherapy.
Collapse
Affiliation(s)
- Yijin Huang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Anna Jia
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yufei Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
34
|
Regulatory T Cells in Ovarian Carcinogenesis and Future Therapeutic Opportunities. Cancers (Basel) 2022; 14:cancers14225488. [PMID: 36428581 PMCID: PMC9688690 DOI: 10.3390/cancers14225488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022] Open
Abstract
Regulatory T cells (Tregs) have been shown to play a role in the development of solid tumors. A better understanding of the biology of Tregs, immune suppression by Tregs, and how cancer developed with the activity of Tregs has facilitated the development of strategies used to improve immune-based therapy. In ovarian cancer, Tregs have been shown to promote cancer development and resistance at different cancer stages. Understanding the various Treg-mediated immune escape mechanisms provides opportunities to establish specific, efficient, long-lasting anti-tumor immunity. Here, we review the evidence of Treg involvement in various stages of ovarian cancer. We further provide an overview of the current and prospective therapeutic approaches that arise from the modulation of Treg-related tumor immunity at those specific stages. Finally, we propose combination strategies of Treg-related therapies with other anti-tumor therapies to improve clinical efficacy and overcome tumor resistance in ovarian cancer.
Collapse
|
35
|
Immunoregulatory signal networks and tumor immune evasion mechanisms: insights into therapeutic targets and agents in clinical development. Biochem J 2022; 479:2219-2260. [DOI: 10.1042/bcj20210233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022]
Abstract
Through activation of immune cells, the immune system is responsible for identifying and destroying infected or otherwise damaged cells including tumorigenic cells that can be recognized as foreign, thus maintaining homeostasis. However, tumor cells have evolved several mechanisms to avoid immune cell detection and killing, resulting in tumor growth and progression. In the tumor microenvironment, tumor infiltrating immune cells are inactivated by soluble factors or tumor promoting conditions and lose their effects on tumor cells. Analysis of signaling and crosstalk between immune cells and tumor cells have helped us to understand in more detail the mechanisms of tumor immune evasion and this forms basis for drug development strategies in the area of cancer immunotherapy. In this review, we will summarize the dominant signaling networks involved in immune escape and describe the status of development of therapeutic strategies to target tumor immune evasion mechanisms with focus on how the tumor microenvironment interacts with T cells.
Collapse
|
36
|
Zhong Y, Ren X, Cao X, Xu Y, Song Y, Zhou Y, Mao F, Shen S, Wang Z, Sun Q. Insulin-like growth factor 2 receptor is a key immune-related gene that is correlated with a poor prognosis in patients with triple-negative breast cancer: A bioinformatics analysis. Front Oncol 2022; 12:871786. [PMID: 36330486 PMCID: PMC9624382 DOI: 10.3389/fonc.2022.871786] [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: 02/08/2022] [Accepted: 09/27/2022] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Immunotherapy plays an important role in the treatment of triple-negative breast cancer (TNBC). This study aimed to identify immune-related genes that are associated with the prognosis of patients with TNBC as possible targets of immunotherapy, alongside their related tumor-infiltrating lymphocytes (TILs). METHODS The clinical data and gene expression profiles of patients with breast cancer were extracted from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases and divided into training (n = 1,053) and verification (n = 508) groups. CIBERSORT was used to predict the differences in immune cell infiltration in patient subsets that were stratified according to risk. Gene Ontology (GO) enrichment analysis was used to identify pathways associated with immune-related genes in patient subsets that were stratified according to risk. The clinical data and insulin-like growth factor 2 receptor (IGF2R) expression profiles of patients with breast cancer were extracted from METABRIC. The expression of IGF2R and TILs were evaluated in a cohort containing 282 untreated patients with TNBC. The correlations of IGF2R expression, TILs, and clinicopathological parameters with patient prognosis were analyzed in the whole cohort. RESULTS The prognostic model, which was composed of 26 immune-related gene pairs, significantly distinguished between high- and low-risk patients. Univariate and multivariate analyses indicated that the model was an independent prognostic factor for breast cancer. Among the identified genes, the expression of IGF2R significantly distinguished between high- and low-risk patients in TCGA (P = 0.008) and in METABRIC patients (P < 0.001). The expression of IGF2R was significantly associated with clinical risk factors such as TNBC, estrogen receptor (ER)-negative expression, human epidermal growth factor receptor 2 (HER2)-positive expression, and age ≤60 years old in METABRIC patients. In addition, the patients with IGF2R-positive expression had lower disease-free survival (DFS) rates than those with IGF2R-negative expression in the TNBC cohort (67.8% vs. 78.5%, P = 0.023). IGF2R expression also was significantly negatively correlated with TILs, particularly with CD8+ TILs and CD19+ TILs in the cohort of patients with TNBC. CONCLUSION IGF2R can be used as an indicator of a poor prognosis in patients with TNBC and as a potential target and research direction for TNBC immunotherapy in the future.
Collapse
Affiliation(s)
- Ying Zhong
- Department of Breast Disease, Peking Union Medical College Hospital, Beijing, China
| | - Xinyu Ren
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Xi Cao
- Department of Breast Disease, Peking Union Medical College Hospital, Beijing, China
| | - Yali Xu
- Department of Breast Disease, Peking Union Medical College Hospital, Beijing, China
| | - Yu Song
- Department of Breast Disease, Peking Union Medical College Hospital, Beijing, China
| | - Yidong Zhou
- Department of Breast Disease, Peking Union Medical College Hospital, Beijing, China
| | - Feng Mao
- Department of Breast Disease, Peking Union Medical College Hospital, Beijing, China
| | - Songjie Shen
- Department of Breast Disease, Peking Union Medical College Hospital, Beijing, China
| | - Zhe Wang
- Department of Breast Disease, Peking Union Medical College Hospital, Beijing, China
| | - Qiang Sun
- Department of Breast Disease, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
37
|
McRitchie BR, Akkaya B. Exhaust the exhausters: Targeting regulatory T cells in the tumor microenvironment. Front Immunol 2022; 13:940052. [PMID: 36248808 PMCID: PMC9562032 DOI: 10.3389/fimmu.2022.940052] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022] Open
Abstract
The concept of cancer immunotherapy has gained immense momentum over the recent years. The advancements in checkpoint blockade have led to a notable progress in treating a plethora of cancer types. However, these approaches also appear to have stalled due to factors such as individuals' genetic make-up, resistant tumor sub-types and immune related adverse events (irAE). While the major focus of immunotherapies has largely been alleviating the cell-intrinsic defects of CD8+ T cells in the tumor microenvironment (TME), amending the relationship between tumor specific CD4+ T cells and CD8+ T cells has started driving attention as well. A major roadblock to improve the cross-talk between CD4+ T cells and CD8+ T cells is the immune suppressive action of tumor infiltrating T regulatory (Treg) cells. Despite their indispensable in protecting tissues against autoimmune threats, Tregs have also been under scrutiny for helping tumors thrive. This review addresses how Tregs establish themselves at the TME and suppress anti-tumor immunity. Particularly, we delve into factors that promote Treg migration into tumor tissue and discuss the unique cellular and humoral composition of TME that aids survival, differentiation and function of intratumoral Tregs. Furthermore, we summarize the potential suppression mechanisms used by intratumoral Tregs and discuss ways to target those to ultimately guide new immunotherapies.
Collapse
Affiliation(s)
- Bayley R. McRitchie
- Department of Neurology, The College of Medicine, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Billur Akkaya
- Department of Neurology, The College of Medicine, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
38
|
Rallis KS, Corrigan AE, Dadah H, Stanislovas J, Zamani P, Makker S, Szabados B, Sideris M. IL-10 in cancer: an essential thermostatic regulator between homeostatic immunity and inflammation - a comprehensive review. Future Oncol 2022; 18:3349-3365. [PMID: 36172856 DOI: 10.2217/fon-2022-0063] [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] [Indexed: 12/30/2022] Open
Abstract
Cytokines are soluble proteins that mediate intercellular signaling regulating immune and inflammatory responses. Cytokine modulation represents a promising cancer immunotherapy approach for immune-mediated tumor regression. However, redundancy in cytokine signaling and cytokines' pleiotropy, narrow therapeutic window, systemic toxicity, short half-life and limited efficacy represent outstanding challenges for cytokine-based cancer immunotherapies. Recently, there has been interest in the paradoxical role of IL-10 in cancer, its controversial prognostic utility and novel strategies to enhance its therapeutic profile. Here, the authors review the literature surrounding the role of IL-10 within the tumor microenvironment, its prognostic correlates to cancer patient outcomes and its pro- and antitumor effects, and they assess the legitimacy of potential therapeutic strategies harnessing IL-10 by outlining the notable preclinical and clinical evidence to date.
Collapse
Affiliation(s)
- Kathrine S Rallis
- Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AD, UK.,Barts Cancer Institute, Queen Mary University of London, London, EC1M 5PZ, UK
| | - Amber E Corrigan
- GKT School of Medicine, King's College London, London, SE1 9RT, UK
| | - Hashim Dadah
- GKT School of Medicine, King's College London, London, SE1 9RT, UK
| | - Justas Stanislovas
- Barts Cancer Institute, Queen Mary University of London, London, EC1M 5PZ, UK
| | - Parisa Zamani
- GKT School of Medicine, King's College London, London, SE1 9RT, UK
| | - Shania Makker
- Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AD, UK
| | - Bernadett Szabados
- Barts Cancer Institute, Queen Mary University of London, London, EC1M 5PZ, UK
| | - Michail Sideris
- Women's Health Research Unit, Queen Mary University of London, London, E1 2AB, UK
| |
Collapse
|
39
|
Wang Y, Johnson KCC, Gatti-Mays ME, Li Z. Emerging strategies in targeting tumor-resident myeloid cells for cancer immunotherapy. J Hematol Oncol 2022; 15:118. [PMID: 36031601 PMCID: PMC9420297 DOI: 10.1186/s13045-022-01335-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/09/2022] [Indexed: 12/11/2022] Open
Abstract
Immune checkpoint inhibitors targeting programmed cell death protein 1, programmed death-ligand 1, and cytotoxic T-lymphocyte-associated protein 4 provide deep and durable treatment responses which have revolutionized oncology. However, despite over 40% of cancer patients being eligible to receive immunotherapy, only 12% of patients gain benefit. A key to understanding what differentiates treatment response from non-response is better defining the role of the innate immune system in anti-tumor immunity and immune tolerance. Teleologically, myeloid cells, including macrophages, dendritic cells, monocytes, and neutrophils, initiate a response to invading pathogens and tissue repair after pathogen clearance is successfully accomplished. However, in the tumor microenvironment (TME), these innate cells are hijacked by the tumor cells and are imprinted to furthering tumor propagation and dissemination. Major advancements have been made in the field, especially related to the heterogeneity of myeloid cells and their function in the TME at the single cell level, a topic that has been highlighted by several recent international meetings including the 2021 China Cancer Immunotherapy workshop in Beijing. Here, we provide an up-to-date summary of the mechanisms by which major myeloid cells in the TME facilitate immunosuppression, enable tumor growth, foster tumor plasticity, and confer therapeutic resistance. We discuss ongoing strategies targeting the myeloid compartment in the preclinical and clinical settings which include: (1) altering myeloid cell composition within the TME; (2) functional blockade of immune-suppressive myeloid cells; (3) reprogramming myeloid cells to acquire pro-inflammatory properties; (4) modulating myeloid cells via cytokines; (5) myeloid cell therapies; and (6) emerging targets such as Siglec-15, TREM2, MARCO, LILRB2, and CLEVER-1. There is a significant promise that myeloid cell-based immunotherapy will help advance immuno-oncology in years to come.
Collapse
Affiliation(s)
- Yi Wang
- Division of Medical Oncology, Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Margaret E Gatti-Mays
- Division of Medical Oncology, Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
- Stefanie Spielman Comprehensive Breast Center, Columbus, OH, USA.
| | - Zihai Li
- Division of Medical Oncology, Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
40
|
Shi Y, Xie T, Wang B, Wang R, Cai Y, Yuan B, Gleber-Netto FO, Tian X, Rodriguez-Rosario AE, Osman AA, Wang J, Pickering CR, Ren X, Sikora AG, Myers JN, Rangel R. Mutant p53 drives an immune cold tumor immune microenvironment in oral squamous cell carcinoma. Commun Biol 2022; 5:757. [PMID: 35902768 PMCID: PMC9334280 DOI: 10.1038/s42003-022-03675-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 07/06/2022] [Indexed: 02/03/2023] Open
Abstract
The critical role of the tumor immune microenvironment (TIME) in determining response to immune checkpoint inhibitor (ICI) therapy underscores the importance of understanding cancer cell-intrinsic mechanisms driving immune-excluded ("cold") TIMEs. One such cold tumor is oral cavity squamous cell carcinoma (OSCC), a tobacco-associated cancer with mutations in the TP53 gene which responds poorly to ICI therapy. Because altered TP53 function promotes tumor progression and plays a potential role in TIME modulation, here we developed a syngeneic OSCC models with defined Trp53 (p53) mutations and characterized their TIMEs and degree of ICI responsiveness. We observed that a carcinogen-induced p53 mutation promoted a cold TIME enriched with immunosuppressive M2 macrophages highly resistant to ICI therapy. p53-mutated cold tumors failed to respond to combination ICI treatment; however, the combination of a programmed cell death protein 1 (PD-1) inhibitor and stimulator of interferon genes (STING) agonist restored responsiveness. These syngeneic OSCC models can be used to gain insights into tumor cell-intrinsic drivers of immune resistance and to develop effective immunotherapeutic approaches for OSCC and other ICI-resistant solid tumors.
Collapse
Affiliation(s)
- Yewen Shi
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA ,grid.452672.00000 0004 1757 5804Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| | - Tongxin Xie
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Bingbing Wang
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Rong Wang
- grid.49470.3e0000 0001 2331 6153Department of Endodontics, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu Cai
- grid.49470.3e0000 0001 2331 6153Department of Oral & Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bo Yuan
- grid.240145.60000 0001 2291 4776Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Frederico O. Gleber-Netto
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Xiangjun Tian
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Alanis E. Rodriguez-Rosario
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA ,grid.449853.70000 0001 2051 0540Department of Biology, University of Puerto Rico, Bayamon, Puerto Rico USA
| | - Abdullah A. Osman
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Jing Wang
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Curtis R. Pickering
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Xiaoyong Ren
- grid.452672.00000 0004 1757 5804Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004 China
| | - Andrew G. Sikora
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Jeffrey N. Myers
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| | - Roberto Rangel
- grid.240145.60000 0001 2291 4776Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 7030 USA
| |
Collapse
|
41
|
Zhang Y, Vu T, Palmer DC, Kishton RJ, Gong L, Huang J, Nguyen T, Chen Z, Smith C, Livák F, Paul R, Day CP, Wu C, Merlino G, Aldape K, Guan XY, Jiang P. A T cell resilience model associated with response to immunotherapy in multiple tumor types. Nat Med 2022; 28:1421-1431. [PMID: 35501486 PMCID: PMC9406236 DOI: 10.1038/s41591-022-01799-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 03/24/2022] [Indexed: 01/10/2023]
Abstract
Despite breakthroughs in cancer immunotherapy, most tumor-reactive T cells cannot persist in solid tumors due to an immunosuppressive environment. We developed Tres (tumor-resilient T cell), a computational model utilizing single-cell transcriptomic data to identify signatures of T cells that are resilient to immunosuppressive signals, such as transforming growth factor-β1, tumor necrosis factor-related apoptosis-inducing ligand and prostaglandin E2. Tres reliably predicts clinical responses to immunotherapy in melanoma, lung cancer, triple-negative breast cancer and B cell malignancies using bulk T cell transcriptomic data from pre-treatment tumors from patients who received immune-checkpoint inhibitors (n = 38), infusion products for chimeric antigen receptor T cell therapies (n = 34) and pre-manufacture samples for chimeric antigen receptor T cell or tumor-infiltrating lymphocyte therapies (n = 84). Further, Tres identified FIBP, whose functions are largely unknown, as the top negative marker of tumor-resilient T cells across many solid tumor types. FIBP knockouts in murine and human donor CD8+ T cells significantly enhanced T cell-mediated cancer killing in in vitro co-cultures. Further, Fibp knockout in murine T cells potentiated the in vivo efficacy of adoptive cell transfer in the B16 tumor model. Fibp knockout T cells exhibit reduced cholesterol metabolism, which inhibits effector T cell function. These results demonstrate the utility of Tres in identifying biomarkers of T cell effectiveness and potential therapeutic targets for immunotherapies in solid tumors.
Collapse
Affiliation(s)
- Yu Zhang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Trang Vu
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Douglas C Palmer
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- AstraZeneca, Gaithersburg, MD, USA
| | - Rigel J Kishton
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Lyell Immunopharma, South San Francisco, CA, USA
| | - Lanqi Gong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Jiao Huang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Thanh Nguyen
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Gaia Foods, Singapore, Singapore
| | - Zuojia Chen
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cari Smith
- Laboratory Animal Science Program, Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Ferenc Livák
- Flow Cytometry Core, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rohit Paul
- Office of the Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chuan Wu
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
42
|
Yang Z, Loy J, Poirson B, Dai Y, Rajendran S, Xu S, Spires V, Gururajan M, Lin Z, Arbanas J, Carl S, Pace S, Wang Y, Mehl J, Vasudevan K, Spires T, Novosiadly R, Coker S, Perez R, Covello K, Morin P, Graziano R, Broz M, Lehman-McKeeman L. Application of Pharmacokinetic/Pharmacodynamic Modeling to Bridge Mouse Antitumor Efficacy and Monkey Toxicology Data for Determining the Therapeutic Index of an Interleukin-10 Fc Fusion Protein. Front Pharmacol 2022; 13:829063. [PMID: 35795558 PMCID: PMC9251408 DOI: 10.3389/fphar.2022.829063] [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: 12/04/2021] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Pharmacokinetic/pharmacodynamic (PK/PD) modeling was performed to quantitatively integrate preclinical pharmacology and toxicology data for determining the therapeutic index (TI) of an interleukin-10 (IL-10) fragment crystallizable (Fc) fusion protein. Mouse Fc fused with mouse IL-10 (mFc-mIL-10) was studied in mice for antitumor efficacy, and the elevation of interleukin-18 (IL-18) was examined as a PD biomarker. The in vivo mFc-mIL-10 EC50 for the IL-18 induction was estimated to be 2.4 nM, similar to the in vitro receptor binding affinity (Kd) of 3.2 nM. The IL-18 induction was further evaluated in cynomolgus monkeys, where the in vivo induction EC50 by a human IL-10 human Fc-fusion protein (hFc-hIL-10) was 0.08 nM vs. 0.3 nM measured as the in vitro Kd. The extent of the IL-18 induction correlated with mouse antitumor efficacy and was used to connect mouse efficacy to that in monkeys. The PD-based efficacious dose projected in monkeys was comparable to the results obtained using a PK-based method in which mouse efficacious exposure was targeted and corrected for affinity differences between the species. Furthermore, PK/PD relationships were developed for anemia and thrombocytopenia in monkeys treated with hFc-hIL-10, with thrombocytopenia predicted to be dose-limiting toxicity. Using quantitative pharmacology and toxicology information obtained through modeling work in the same species, the TI of hFc-hIL-10 in monkeys was determined to be 2.4 (vs. PD-based efficacy) and 1.2–3 (vs. PK-based efficacy), indicating a narrow safety margin. The model-based approaches were proven valuable to the developability assessment of the IL-10 Fc-fusion protein.
Collapse
Affiliation(s)
- Zheng Yang
- Metabolism and Pharmacokinetics, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
- *Correspondence: Zheng Yang,
| | - James Loy
- Discovery Toxicology, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Cambridge, MA, United States
| | - Brian Poirson
- Discovery Oncology, Bristol Myers Squibb, Princeton, NJ, United States
| | - Yanshan Dai
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, United States
| | - Surendran Rajendran
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, United States
| | - Shihua Xu
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, United States
| | - Vanessa Spires
- Discovery Oncology, Bristol Myers Squibb, Princeton, NJ, United States
| | - Murali Gururajan
- Discovery Oncology, Bristol Myers Squibb, Princeton, NJ, United States
| | - Zheng Lin
- Discovery Biotherapeutics, Bristol Myers Squibb, Princeton, NJ, United States
| | - Jaren Arbanas
- Discovery Biotherapeutics, Bristol Myers Squibb, Princeton, NJ, United States
| | - Stephen Carl
- Discovery Pharmaceutics and Analytical Sciences, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| | - Samantha Pace
- Discovery Pharmaceutics and Analytical Sciences, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| | - Yun Wang
- Discovery Pharmaceutics and Analytical Sciences, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| | - John Mehl
- Discovery Pharmaceutics and Analytical Sciences, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| | - Krishna Vasudevan
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Thomas Spires
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Ruslan Novosiadly
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Shodeinde Coker
- Oncology Early Clinical Development, Bristol Myers Squibb, Princeton, NJ, United States
| | - Raymond Perez
- Oncology Early Clinical Development, Bristol Myers Squibb, Princeton, NJ, United States
| | - Kelly Covello
- Oncology Early Clinical Development, Bristol Myers Squibb, Princeton, NJ, United States
| | - Paul Morin
- Discovery Biotherapeutics, Bristol Myers Squibb, Princeton, NJ, United States
| | - Robert Graziano
- Discovery Oncology, Bristol Myers Squibb, Princeton, NJ, United States
| | - Miranda Broz
- Discovery Oncology, Bristol Myers Squibb, Redwood City, CA, United States
| | - Lois Lehman-McKeeman
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Princeton, NJ, United States
| |
Collapse
|
43
|
Pei Y, Xiang Z, Wen K, Tu CR, Wang X, Zhang Y, Mu X, Liu Y, Tu W. CD137 Costimulation Enhances the Antitumor Activity of Vγ9Vδ2-T Cells in IL-10-Mediated Immunosuppressive Tumor Microenvironment. Front Immunol 2022; 13:872122. [PMID: 35784354 PMCID: PMC9247142 DOI: 10.3389/fimmu.2022.872122] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/17/2022] [Indexed: 11/29/2022] Open
Abstract
Although γδ-T cell-based tumor immunotherapy using phosphoantigens to boost γδ-T cell immunity has shown success in some cancer patients, the clinical application is limited due to the rapid exhaustion of Vγ9Vδ2-T cells caused by repetitive stimulation from phosphoantigens and the profoundly immunosuppressive tumor microenvironment (TME). In this study, using a cell culture medium containing human and viral interleukin-10 (hIL-10 and vIL-10) secreted from EBV-transformed lymphoblastoid B cell lines (EBV-LCL) to mimic the immunosuppressive TEM, we found that the antitumor activity of Vγ9Vδ2-T cells was highly suppressed by endogenous hIL-10 and vIL-10 within the TME. CD137 costimulation could provide an anti-exhaustion signal to mitigate the suppressive effects of IL-10 in TME by suppressing IL-10R1 expression on Vγ9Vδ2-T cells. CD137 costimulation also improved the compromised antitumor activity of Vγ9Vδ2-T cells in TME with high levels of IL-10 in Rag2-/- γc-/- mice. In humanized mice, CD137 costimulation boosted the therapeutic effects of aminobisphosphonate pamidronate against EBV-induced lymphoma. Our study offers a novel approach to overcoming the obstacle of the hIL-10 and vIL-10-mediated immunosuppressive microenvironment by costimulating CD137 and enhancing the efficacy of γδ-T cell-based tumor therapy.
Collapse
Affiliation(s)
- Yujun Pei
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Zheng Xiang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Kun Wen
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Chloe Ran Tu
- Computational and Systems Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Xiwei Wang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yanmei Zhang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiaofeng Mu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yinping Liu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wenwei Tu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Wenwei Tu,
| |
Collapse
|
44
|
Yang Z, Rajendran S, Spires V, Poirson B, Gururajan M, Lin Z, Arbanas J, Krystek S, Loy J, Cheng Y, Carl S, Pace S, Wang Y, Mehl J, Xu S, Vasudevan K, Broz M, Lehman-McKeeman LD, Morin P, Graziano RF. Target-Mediated Drug Disposition Affects the Pharmacokinetics of Interleukin-10 Fc-fusion Proteins at Pharmacologically Active Doses. Drug Metab Dispos 2022; 50:898-908. [PMID: 35545256 DOI: 10.1124/dmd.121.000799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Fragment crystallizable (Fc) fusion is commonly used for extending the half-life of biotherapeutics such as cytokines. In this work, we studied the pharmacokinetics of Fc-fused interleukin-10 (IL-10) proteins that exhibited potent anti-tumor activity in mouse syngeneic tumor models. At pharmacologically active doses of {greater than or equal to}0.1 mg/kg, both mouse Fc-mouse IL-10 and human Fc-human IL-10, constructed as the C-terminus of the Fc domain fused with IL-10 via a glycine-serine polypeptide linker, exhibited nonlinear pharmacokinetics after intravenous administration to mice at the doses of 0.05, 0.5, and 5 mg/kg. With a nominal dose ratio of 1:10:100; the ratio of the area under the curve for mouse Fc-mouse IL-10 and human Fc-human IL-10 was 1:181:1830 and 1:75:633, respectively. In contrast, recombinant mouse or human IL-10 proteins exhibited linear pharmacokinetics in mice. Compartmental analysis, using the Michaelis-Menten equation with the in vitro IL-10 receptor alpha binding affinity inputted as the Km, unified the pharmacokinetic data across the dose range. Additionally, non-target-mediated clearance estimated for fusion proteins was ~200-fold slower than that for cytokines, causing the manifestation of target-mediated drug disposition (TMDD) in the fusion protein pharmacokinetics. The experimental data generated with a mouse IL-10 receptor alpha-blocking antibody and a human Fc-human IL-10 mutant with a reduced receptor binding affinity showed significant improvements in pharmacokinetics, supporting TMDD as the cause of nonlinearity. Target expression and its effect on pharmacokinetics must be determined when considering using Fc as a half-life extension strategy, and pharmacokinetic evaluations need to be performed at a range of doses covering pharmacological activity. Significance Statement Target-mediated drug disposition can manifest to affect the pharmacokinetics of a fragment crystallizable (Fc)-fused cytokine when the non-target-mediated clearance of the cytokine is decreased due to neonatal Fc receptor-mediated recycling and molecular weight increases that reduce the renal clearance. The phenomenon was demonstrated with interleukin-10 Fc-fusion proteins in mice at pharmacologically active doses. Future drug designs using Fc as a half-life extension approach for cytokines need to consider target expression and its effect on pharmacokinetics at relevant doses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yun Wang
- Bristol Myers Squibb, United States
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Liu S, Meng Y, Liu L, Lv Y, Yu W, Liu T, Wang L, Mu D, Zhou Q, Liu M, Ren Y, Zhang D, Li B, Sun Q, Ren X. CD4 + T cells are required to improve the efficacy of CIK therapy in non-small cell lung cancer. Cell Death Dis 2022; 13:441. [PMID: 35523765 PMCID: PMC9076680 DOI: 10.1038/s41419-022-04882-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
Abstract
As a widely studied adoptive treatment method, CIK (cytokine-induced killer cells) treatment has shown clinical benefits in many clinical trials on non-small cell lung cancer. As a heterogeneous cell population, however, CIK cells have a strong instability and individual differences in their efficacies, which are collaboratively regulated by the tumor microenvironment and CIK subpopulations. Among them, CD4+ T cells belong to a crucial subgroup of the CIK cell population, and their influence on CIK therapy is still unclear. Herein, we show how CD4+ T cells positively regulate the functions of CD3+CD56+ T and CD3+CD8+ T cells. During this process, we found that Th1/Th17 CD4+ subgroups can induce the phosphorylation of the AKT pathway by secreting IL-17A, and upregulate the expression of T-bet/Eomes transcription factors, thereby restoring the function of CD8+/CD3+CD56+ T cells and reversing the exhaustion of PD-1+Tim-3+ T cells. These findings will provide guidance for the clinical screening of suitable populations for CIK treatment and formulation of strategies for CIK therapy plus immune checkpoint treatment. Based on these findings, we are conducting an open-label phase II study (NCT04836728) is to evaluate the effects of autologous CIKs in combination with PD-1 inhibitor in the first-line treatment of IV NSCLC, and hope to observe patients' benefits in this clinical trial.
Collapse
Affiliation(s)
- Shaochuan Liu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuan Meng
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Liang Liu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yingge Lv
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenwen Yu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ting Liu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Limei Wang
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Di Mu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qiuru Zhou
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Min Liu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yulin Ren
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Dong Zhang
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Baihui Li
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qian Sun
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiubao Ren
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
46
|
Li Y, Wang J, Wang H, Zhang S, Wei Y, Liu S. The Interplay Between Inflammation and Stromal Components in Pancreatic Cancer. Front Immunol 2022; 13:850093. [PMID: 35493517 PMCID: PMC9046560 DOI: 10.3389/fimmu.2022.850093] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/24/2022] [Indexed: 01/18/2023] Open
Abstract
Inflammation involves interactions between various immune cells, inflammatory cells, chemokines and cytokines in pancreatic cancer. Cancer cells as well as surrounding stromal and inflammatory cells establish an inflammatory tumor microenvironment (TME). Inflammation is closely associated with immunity. Meanwhile, immune cells are involved in both inflammation and immune response. Tumor-promoting inflammation and tumor-suppressive immunity are two main characteristics of the tumor microenvironment in pancreatic cancer. Yet, the mechanism of inflammation and immune response in pancreatic cancer development is still unclear due to the dual role of some cytokines and the complicated crosstalk between tumor and stromal components in TME. In this review, we outline the principal cytokines and stromal cells in the pancreatic TME that are involved in the tumor-promoting and immunosuppressive effects of inflammation, and discuss the interaction between inflammation and stromal components in pancreatic cancer progression. Moreover, the clinical approaches based on targeting TME in pancreatic cancer are also summarized. Defining the mechanisms of interplay between inflammation and stromal components will be essential for further development of anti-cancer therapies.
Collapse
Affiliation(s)
- Ying Li
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Wang
- Department of Operating Room, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haiyan Wang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shaoqiang Zhang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingxin Wei
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
47
|
Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol 2022; 19:237-253. [PMID: 34997230 DOI: 10.1038/s41571-021-00588-9] [Citation(s) in RCA: 379] [Impact Index Per Article: 189.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 12/14/2022]
Abstract
During the past 40 years, cytokines and cytokine receptors have been extensively investigated as either cancer targets or cancer treatments. A strong preclinical rationale supports therapeutic strategies to enhance the growth inhibitory and immunostimulatory effects of interferons and interleukins, including IL-2, IL-7, IL-12 and IL-15, or to inhibit the inflammatory and tumour-promoting actions of cytokines such as TNF, IL-1β and IL-6. This rationale is underscored by the discovery of altered and dysregulated cytokine expression in all human cancers. These findings prompted clinical trials of several cytokines or cytokine antagonists, revealing relevant biological activity but limited therapeutic efficacy. However, most trials involved patients with advanced-stage disease, which might not be the optimal setting for cytokine-based therapy. The advent of more effective immunotherapies and an increased understanding of the tumour microenvironment have presented new approaches to harnessing cytokine networks in the treatment of cancer, which include using cytokine-based therapies to enhance the activity or alleviate the immune-related toxicities of other treatments as well as to target early stage cancers. Many challenges remain, especially concerning delivery methods, context dependencies, and the pleiotropic, redundant and often conflicting actions of many cytokines. Herein, we discuss the lessons learnt from the initial trials of single-agent cytokine-based therapies and subsequent efforts to better exploit such agents for the treatment of solid tumours.
Collapse
Affiliation(s)
- David J Propper
- Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Frances R Balkwill
- Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
48
|
CD4 + T-cell-derived IL-10 promotes CNS inflammation in mice by sustaining effector T cell survival. Cell Rep 2022; 38:110565. [PMID: 35354043 DOI: 10.1016/j.celrep.2022.110565] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 01/05/2022] [Accepted: 03/03/2022] [Indexed: 11/24/2022] Open
Abstract
Interleukin (IL)-10 is considered a prototypical anti-inflammatory cytokine, significantly contributing to the maintenance and reestablishment of immune homeostasis. Accordingly, it has been shown in the intestine that IL-10 produced by Tregs can act on effector T cells, thereby limiting inflammation. Herein, we investigate whether this role also applies to IL-10 produced by T cells during central nervous system (CNS) inflammation. During neuroinflammation, both CNS-resident and -infiltrating cells produce IL-10; yet, as IL-10 has a pleotropic function, the exact contribution of the different cellular sources is not fully understood. We find that T-cell-derived IL-10, but not other relevant IL-10 sources, can promote inflammation in experimental autoimmune encephalomyelitis. Furthermore, in the CNS, T-cell-derived IL-10 acts on effector T cells, promoting their survival and thereby enhancing inflammation and CNS autoimmunity. Our data indicate a pro-inflammatory role of T-cell-derived IL-10 in the CNS.
Collapse
|
49
|
Xu Y, Zeng H, Jin K, Liu Z, Zhu Y, Xu L, Wang Z, Chang Y, Xu J. Immunosuppressive tumor-associated macrophages expressing interlukin-10 conferred poor prognosis and therapeutic vulnerability in patients with muscle-invasive bladder cancer. J Immunother Cancer 2022; 10:jitc-2021-003416. [PMID: 35338085 PMCID: PMC8961180 DOI: 10.1136/jitc-2021-003416] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Background Tumor-associated macrophages (TAMs) secreting IL-10 could be a specific functional cell subset with distinct polarization state and suppressive role in antitumor immune response. Here, we assessed the associations of clinical outcome, therapeutic responses and molecular features with IL-10+TAMs infiltration, and potential impact of IL-10+TAMs on the immune contexture in muscle-invasive bladder cancer (MIBC). Methods In this retrospective study, 128 patients and 391 patients with MIBC from Zhongshan hospital (ZS cohort) and The Cancer Genome Atlas cohort were included respectively. Immunohistochemistry was performed to quantify various immune cell infiltration in the ZS cohort. Single cell RNA sequencing and flow cytometry were performed to examine the functional status of IL-10+TAMs and its correlation with other immune cells. Survival analyses and assessment of the adjuvant chemotherapy (ACT) benefit analyses were also performed. Results High IL-10+TAMs infiltration was associated with inferior prognosis in terms of overall survival and recurrence-free survival, but superior chemotherapeutic response in MIBC. IL-10+TAMs with suppressive features were associated with immunoevasive tumor microenviroment characterized by exhausted CD8+ T cells, immature NK cells and increased immune checkpoint expression. Additionally, high IL-10+TAMs infiltration showed a strong linkage with basal-featured subtype and augmented EGF signaling. Conclusions Immunosuppresive IL-10+TAMs contributed to an evasive contexture with incapacitated immune effector cells and increased immune checkpoint expression, therefore, predicting unfavorable clinical outcomes despite better ACT responsiveness. IL-10+TAMs might provide guidance for customized selection of EGFR-targeted therapy, FGFR3-targeted therapy as well as immunotherapy. The potential of immunosuppressive IL-10+TAMs as a therapeutic target is worth further exploration.
Collapse
Affiliation(s)
- Yijia Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Han Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kaifeng Jin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhaopei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Le Xu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zewei Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Chang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Balakrishnan PB, Ledezma DK, Cano-Mejia J, Andricovich J, Palmer E, Patel VA, Latham PS, Yvon ES, Villagra A, Fernandes R, Sweeney EE. CD137 agonist potentiates the abscopal efficacy of nanoparticle-based photothermal therapy for melanoma. NANO RESEARCH 2022; 15:2300-2314. [PMID: 36089987 PMCID: PMC9455608 DOI: 10.1007/s12274-021-3813-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Despite the promise of immunotherapy such as the immune checkpoint inhibitors (ICIs) anti-PD-1 and anti-CTLA-4 for advanced melanoma, only 26%-52% of patients respond, and many experience grade III/IV immune-related adverse events. Motivated by the need for an effective therapy for patients non-responsive to clinically approved ICIs, we have developed a novel nanoimmunotherapy that combines locally administered Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) with systemically administered agonistic anti-CD137 monoclonal antibody therapy (aCD137). PBNP-PTT was administered at various thermal doses to melanoma cells in vitro, and was combined with aCD137 in vivo to test treatment effects on melanoma tumor progression, animal survival, immunological protection against tumor rechallenge, and hepatotoxicity. When administered at a melanoma-specific thermal dose, PBNP-PTT elicits immunogenic cell death (ICD) in melanoma cells and upregulates markers associated with antigen presentation and immune cell co-stimulation in vitro. Consequently, PBNP-PTT eliminates primary melanoma tumors in vivo, yielding long-term tumor-free survival. However, the antitumor immune effects generated by PBNP-PTT cannot eliminate secondary tumors, despite significantly slowing their growth. The addition of aCD137 enables significant abscopal efficacy and improvement of survival, functioning through activated dendritic cells and tumor-infiltrating CD8+ T cells, and generates CD4+ and CD8+ T cell memory that manifests in the rejection of tumor rechallenge, with no long-term hepatotoxicity. This study describes for the first time a novel and effective nanoimmunotherapy combination of PBNP-PTT with aCD137 mAb therapy for melanoma.
Collapse
Affiliation(s)
- Preethi Bala Balakrishnan
- GW Cancer Center, Department of Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Debbie K. Ledezma
- The Institute for Biomedical Sciences, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Juliana Cano-Mejia
- GW Cancer Center, Department of Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Jaclyn Andricovich
- The Institute for Biomedical Sciences, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Erica Palmer
- GW Cancer Center, Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Vishal A. Patel
- Department of Dermatology & Oncology, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Patricia S. Latham
- Department of Pathology, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Eric S. Yvon
- GW Cancer Center, Department of Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Alejandro Villagra
- GW Cancer Center, Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Rohan Fernandes
- GW Cancer Center, Department of Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
- The Institute for Biomedical Sciences, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
- ImmunoBlue, Bethesda, MD 20817, USA
| | - Elizabeth E. Sweeney
- GW Cancer Center, Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
- ImmunoBlue, Bethesda, MD 20817, USA
| |
Collapse
|