1
|
Hu L, Sun C, Yuan K, Yang P. Expression, regulation, and function of PD-L1 on non-tumor cells in the tumor microenvironment. Drug Discov Today 2024; 29:104181. [PMID: 39278561 DOI: 10.1016/j.drudis.2024.104181] [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: 07/14/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Antiprogrammed death ligand 1 (PD-L1) therapy is a leading immunotherapy, but only some patients with solid cancers benefit. Overwhelming evidence has revealed that PD-L1 is expressed on various immune cells in the tumor microenvironment (TME), including macrophages, dendritic cells, and regulatory T cells, modulating tumor immunity and influencing tumor progression. PD-L1 can also be located on tumor cell membranes as well as in exosomes and cytoplasm. Accordingly, the dynamic expression and various forms of PD-L1 might explain the therapy's limited efficacy and resistance. Herein a systematic summary of the expression of PD-L1 on different immune cells and their regulatory mechanisms is provided to offer a solid foundation for future studies.
Collapse
Affiliation(s)
- Lingrong Hu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China.
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
2
|
Loginova N, Aniskin D, Timashev P, Ulasov I, Kharwar RK. GBM Immunotherapy: Macrophage Impacts. Immunol Invest 2024; 53:730-751. [PMID: 38634572 DOI: 10.1080/08820139.2024.2337022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
BACKGROUND Glioblastoma (GBM) is an extremely aggressive form of brain tumor with low survival rates. Current treatments such as chemotherapy, radiation, and surgery are problematic due to tumor growth, invasion, and tumor microenvironment. GBM cells are resistant to these standard treatments, and the heterogeneity of the tumor makes it difficult to find a universal approach. Progression of GBM and acquisition of resistance to therapy are due to the complex interplay between tumor cells and the TME. A significant portion of the TME consists of an inflammatory infiltrate, with microglia and macrophages being the predominant cells. METHODS Analysis of the literature data over a course of 5 years suggest that the tumor-associated macrophages (TAMs) are capable of releasing cytokines and growth factors that promote tumor proliferation, survival, and metastasis while inhibiting immune cell function at the same time. RESULTS Thus, immunosuppressive state, provided with this intensively studied kind of TME cells, is supposed to promote GBM development through TAMs modulation of tumor treatment-resistance and aggressiveness. Therefore, TAMs are an attractive therapeutic target in the treatment of glioblastoma. CONCLUSION This review provides a comprehensive overview of the latest research on the nature of TAMs and the development of therapeutic strategies targeting TAMs, focusing on the variety of macrophage properties, being modulated, as well as molecular targets.
Collapse
Affiliation(s)
- Nina Loginova
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Denis Aniskin
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Peter Timashev
- World-Class Research Centre "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Rajesh Kumar Kharwar
- Endocrine Research Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
| |
Collapse
|
3
|
Almeida PP, Moraes JA, Barja-Fidalgo TC, Renovato-Martins M. Extracellular vesicles as modulators of monocyte and macrophage function in tumors. AN ACAD BRAS CIENC 2024; 96:e20231212. [PMID: 38922279 DOI: 10.1590/0001-3765202420231212] [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: 11/07/2023] [Accepted: 02/17/2024] [Indexed: 06/27/2024] Open
Abstract
The tumor microenvironment (TME) harbors several cell types, such as tumor cells, immune cells, and non-immune cells. These cells communicate through several mechanisms, such as cell-cell contact, cytokines, chemokines, and extracellular vesicles (EVs). Tumor-derived vesicles are known to have the ability to modulate the immune response. Monocytes are a subset of circulating innate immune cells and play a crucial role in immune surveillance, being recruited to tissues where they differentiate into macrophages. In the context of tumors, it has been observed that tumor cells can attract monocytes to the TME and induce their differentiation into tumor-associated macrophages with a pro-tumor phenotype. Tumor-derived EVs have emerged as essential structures mediating this process. Through the transfer of specific molecules and signaling factors, tumor-derived EVs can shape the phenotype and function of monocytes, inducing the expression of cytokines and molecules by these cells, thus modulating the TME towards an immunosuppressive environment.
Collapse
Affiliation(s)
- Palloma P Almeida
- Universidade Federal Fluminense, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Laboratório de Inflamação e Metabolismo, Rua Professor Marcos Waldemar de Freitas Reis, s/n, 24020-140 Niterói, RJ, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Laboratório de Biologia Redox, Av. Carlos Chagas Filho, 373, Prédio do ICB - Anexo B1F3, Ilha do Fundão, 21941-902 Rio de Janeiro, RJ, Brazil
- Universidade do Estado do Rio de Janeiro, Departamento de Biologia Celular, Instituto de Biologia Roberto Alcantara Gomes - IBRAG, Laboratório de Farmacologia Celular e Molecular, Av. 28 de setembro, 87, 20551-030 Rio de Janeiro, RJ, Brazil
| | - João Alfredo Moraes
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Laboratório de Biologia Redox, Av. Carlos Chagas Filho, 373, Prédio do ICB - Anexo B1F3, Ilha do Fundão, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Thereza Christina Barja-Fidalgo
- Universidade do Estado do Rio de Janeiro, Departamento de Biologia Celular, Instituto de Biologia Roberto Alcantara Gomes - IBRAG, Laboratório de Farmacologia Celular e Molecular, Av. 28 de setembro, 87, 20551-030 Rio de Janeiro, RJ, Brazil
| | - Mariana Renovato-Martins
- Universidade Federal Fluminense, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Laboratório de Inflamação e Metabolismo, Rua Professor Marcos Waldemar de Freitas Reis, s/n, 24020-140 Niterói, RJ, Brazil
| |
Collapse
|
4
|
Chern B, Pinto D, Lum JH, Parameswaran R. Nearly Half of Patients with Anaplastic Thyroid Cancer May Be Amenable to Immunotherapy. Biomedicines 2024; 12:1304. [PMID: 38927511 PMCID: PMC11201491 DOI: 10.3390/biomedicines12061304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
IMPORTANCE Poorly differentiated cancer (PDC) and anaplastic thyroid cancer (ATC) have an aggressive course of disease with limited treatment options. The expression of programmed cell death ligand-1 (PD-L1) has been used to determine the responses of many cancers to immunotherapy. The aim of the study was to investigate the expression of PD-L1 in a cohort of patients with PDC and ATC to assess their suitability for immunotherapy. Data, settings, and participants: This study is a retrospective cohort review of patients treated for PDC and ATC treated at a tertiary referral institution during the period 2000-2020. PD-L1 22C3 pharmDx qualitative immunohistochemistry was performed on formalin-fixed, paraffin-embedded (FFPE) specimens of tumours to detect the presence of the PD-L1 protein. MAIN OUTCOME MEASURES The percentage of tumours that were positive for PD-L1 immunohistochemistry and the PD-L1 protein expression as measured by using the Tumour Proportion Score (TPS). Secondary outcomes studied were the associations between demographic, clinicopathological, treatment and disease outcomes and PD-L1 expression. RESULTS Nineteen patients (12F:7M) with a mean age of 65.4 (±14.3 SD) years were diagnosed with PDC in 4 (21%) and fifteen were diagnosed with ATC (79%) during the study period. Fifteen (79%) patients underwent some form of surgery, with R0 resection achieved in only three of the fifteen (20%) patients. Overall, PD-L1 expression was seen in seven of the fifteen (47%) of the patients with ATC, with no positivity seen in the patients with PDC. PD-L1 expression had no impact on treatment modality and positive expression was not significantly associated with stage of disease, metastasis, or survival. CONCLUSION Nearly half of patients with ATC express PD-L1 and may be amenable to immunotherapy with pembrolizumab.
Collapse
Affiliation(s)
- Beverley Chern
- Division of Endocrine Surgery, National University Hospital Health System, Lower Kent Ridge Road, Singapore 119074, Singapore; (B.C.); (D.P.)
| | - Diluka Pinto
- Division of Endocrine Surgery, National University Hospital Health System, Lower Kent Ridge Road, Singapore 119074, Singapore; (B.C.); (D.P.)
- Division of Surgery, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Colombo P.O. Box 6, Sri Lanka
| | - Jeffrey Hy Lum
- Department of Pathology, National University Hospital, Singapore 119074, Singapore;
| | - Rajeev Parameswaran
- Division of Endocrine Surgery, National University Hospital Health System, Lower Kent Ridge Road, Singapore 119074, Singapore; (B.C.); (D.P.)
- Department of Surgery, Yong Loo Lin School of Medicine, Level 8, IE Kent Ridge Road, Singapore 119228, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| |
Collapse
|
5
|
AlHariry NS, El Saftawy EA, Aboulhoda BE, Abozamel AH, Alghamdi MA, Hamoud AE, Khalil Ghanam WAE. Comparison of tissue biomarkers between non-schistosoma and schistosoma-associated urothelial carcinoma. Tissue Cell 2024; 88:102416. [PMID: 38796863 DOI: 10.1016/j.tice.2024.102416] [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: 01/03/2024] [Revised: 04/15/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND High-grade urothelial carcinoma either non-Schistosoma (NS-UBC) or Schistosoma (S-UBC)-associated is the tenth cause of death worldwide and represents a serious therapeutic problem. AIM Evaluation of the immmunohistochemical expression of tumor necrosis factor-alpha (TNFα), epidermal growth factor receptor (EGFR), programmed cell death protein-1 (PDL1), estrogen receptor-alpha (ERα) and UroplakinIII, in the high-grade in NS-UBC and S-UBC as potential prognostic and therapeutic targets analyzed through estimation of area percentage, optical density and international pathological scoring system for each marker. MATERIAL AND METHODS Sixty high grade urothelial carcinoma cases were enrolled in the study (30 cases of NS-UBC and 30 cases of S-UBC). The cases were immunohistochemically-assessed for TNFα, EGFR, PDL1, ERα and Uroplakin III expression. In S-UBC, parasite load was also evaluated for correlation with the immunohistochemical markers' expression in S-UBC. RESULTS The area percentage of immune-expression of TNFα and EGFR was higher in S-UBC compared to NS-UBC. On the other hand, the NS-UBC displayed statistically-higher expression of PDL1 and uroplakinIII (p-value <0.001). ERα revealed higher, yet, non-significant expressions in S-UBC compared to NS-UBC (p-value =0.459). PDL1 expression showed the most superior record regarding area percentage (64.6± 34.5). Regarding optical density, TNF-α showed the highest transmittance expression (2.4 ± 0.9). EGFR positively correlated with PDL1 in S-UBC (r= 0.578, p-value =0.001) whereas in NS-UBC, TNFα and PDL1 (r=0.382, p-value=0.037) had positive correlation. Schistosoma eggs in tissues oppose uroplakin III expression and trigger immunomodulation via PDL1. CONCLUSION Due to lower UroplakinIII expression, S-UBC is supposed to have a poorer prognosis. Hormonal therapy is not hypothesized due to a very minimal ERα expression in both NS-UBC and S-UBC. Regarding immunotherapy, anti-TNF-α is suggested for S-UBC whilst in NS-UBC, blockading PDL1 might be useful. Targeted EGFR therapy seems to carry emphasized outcomes in S-UBC. Correlations encourage combined immune therapy in NS-UBC; nevertheless, in S-UBC, combined anti-EGFR and PDL1 seem to be of benefit.
Collapse
Affiliation(s)
| | - Enas A El Saftawy
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Egypt; Department of Medical Parasitology, Armed Forces College of Medicine, Cairo, Egypt
| | - Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Ahmed H Abozamel
- Department of Urology, Kasr Alainy Hospital, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mansour A Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia; Genomics and Personalized Medicine Unit, The Center for Medical and Health Research, King Khalid University, Abha 62529, Saudi Arabia
| | - Amany E Hamoud
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
6
|
Wang L, Guo W, Guo Z, Yu J, Tan J, Simons DL, Hu K, Liu X, Zhou Q, Zheng Y, Colt EA, Yim J, Waisman J, Lee PP. PD-L1-expressing tumor-associated macrophages are immunostimulatory and associate with good clinical outcome in human breast cancer. Cell Rep Med 2024; 5:101420. [PMID: 38382468 PMCID: PMC10897617 DOI: 10.1016/j.xcrm.2024.101420] [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: 08/02/2023] [Revised: 12/09/2023] [Accepted: 01/18/2024] [Indexed: 02/23/2024]
Abstract
Tumor-associated macrophages (TAMs) are the predominant cells that express programmed cell death ligand 1 (PD-L1) within human tumors in addition to cancer cells, and PD-L1+ TAMs are generally thought to be immunosuppressive within the tumor immune microenvironment (TIME). Using single-cell transcriptomic and spatial multiplex immunofluorescence analyses, we show that PD-L1+ TAMs are mature and immunostimulatory with spatial preference to T cells. In contrast, PD-L1- TAMs are immunosuppressive and spatially co-localize with cancer cells. Either higher density of PD-L1+ TAMs alone or ratio of PD-L1+/PD-L1- TAMs correlate with favorable clinical outcome in two independent cohorts of patients with breast cancer. Mechanistically, we show that PD-L1 is upregulated during the monocyte-to-macrophage maturation and differentiation process and does not require external IFN-γ stimulus. Functionally, PD-L1+ TAMs are more mature/activated and promote CD8+ T cells proliferation and cytotoxic capacity. Together, our findings reveal insights into the immunological significance of PD-L1 within the TIME.
Collapse
Affiliation(s)
- Lei Wang
- International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong 518055, China.
| | - Weihua Guo
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Zhikun Guo
- International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong 518055, China
| | - Jiangnan Yu
- International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong 518055, China
| | - Jiayi Tan
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Diana L Simons
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Ke Hu
- Department of Hematology-Oncology, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, Guangdong 518055, China
| | - Xinyu Liu
- International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong 518055, China
| | - Qian Zhou
- International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong 518055, China
| | - Yizi Zheng
- Department of Thyroid and Breast Surgery, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong 518035, China
| | - Egelston A Colt
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - John Yim
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - James Waisman
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA.
| |
Collapse
|
7
|
Zhao X, Li L, Ma X, Li Y, Gao B, Luo W. The role of immune and inflammatory-related indicators in cognitive dysfunction and disease severity in patients with parkinson's disease. J Neural Transm (Vienna) 2024; 131:13-24. [PMID: 37864052 DOI: 10.1007/s00702-023-02704-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/27/2023] [Indexed: 10/22/2023]
Abstract
We aimed to explore the role of immune and inflammatory indicators in cognitive dysfunction and disease severity in patients with Parkinson's disease (PD). A total of 123 patients with Parkinson's disease were enrolled in the PD group and 49 healthy volunteers in the control group. The patients with PD were further divided into 2 subgroups by evaluating cognitive function using the Montreal Cognitive Assessment (MoCA) and Mini-Mental State Examination (MMSE): the normal cognitive function (PD-NCI) group and the mild cognitive impairment (PD-MCI) group. Moreover, the PD patients were also divided into 2 subgroups using the defined scale of the Hoehn and Yahr (H-Y) stage: the early-stage group and the middle- and late-stage group. Immune and inflammatory indicators, including serum Aβ1-42, Tau, CD4+, CD8+, CD3+, B lymphocytes cell, NK cell, Th17 cell, Treg cell, IL-6, IL-17, and TNF-α levels, were evaluated and analyzed to explore the potential correlation with the cognitive dysfunction and disease severity of PD. Among the 123 PD patients, 60 (48.8%) were diagnosed with mild cognitive impairment. Aβ1-42, CD4+, CD8+, CD3+, and Treg levels observed in the PD-NCI group were lower than the control group (P < 0.001), while higher than the PD-MCI group (P < 0.001). The levels of Tau, Th17, IL-6, IL-17, and TNF-α observed in the PD-NCI group were higher than the control group (P < 0.001), while lower than in the PD-MCI group (P < 0.01). Using the same method, the results of the early-stage group and the middle- and the late-stage group were the same as above. Logistic regression analysis and ROC curve estimation were performed and indicated that the variation of Tau, CD8+, Treg, TNF-α levels was associated with cognitive decline in PD patients, and may serve as markers of PD onset. Furthermore, the variation of Aβ1-42, IL-6, and TNF-α levels was found to correlate with the disease severity of PD. The immune and inflammatory-related indicators may represent an important factor in the pathogenesis of PD, cognitive dysfunction, and disease severity. The variation of Tau protein, CD8+, Treg, and TNF-α levels are associated with the cognitive dysfunction of PD, which may be considered as onset markers. Moreover, the variation of Aβ1-42, IL-6, and TNF-α levels can predict the progression of PD.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu Province, China
- Department of General Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, China
| | - Lei Li
- Department of General Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, China
| | - Xiuping Ma
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, China
| | - Yang Li
- Department of Neurology, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, 313000, Zhejiang Province, China
| | - Beibei Gao
- Department of General Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, China
| | - Weifeng Luo
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu Province, China.
| |
Collapse
|
8
|
Knopf P, Stowbur D, Hoffmann SHL, Hermann N, Maurer A, Bucher V, Poxleitner M, Tako B, Sonanini D, Krishnamachary B, Sinharay S, Fehrenbacher B, Gonzalez-Menendez I, Reckmann F, Bomze D, Flatz L, Kramer D, Schaller M, Forchhammer S, Bhujwalla ZM, Quintanilla-Martinez L, Schulze-Osthoff K, Pagel MD, Fransen MF, Röcken M, Martins AF, Pichler BJ, Ghoreschi K, Kneilling M. Acidosis-mediated increase in IFN-γ-induced PD-L1 expression on cancer cells as an immune escape mechanism in solid tumors. Mol Cancer 2023; 22:207. [PMID: 38102680 PMCID: PMC10722725 DOI: 10.1186/s12943-023-01900-0] [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: 04/14/2023] [Accepted: 11/12/2023] [Indexed: 12/17/2023] Open
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy, yet the efficacy of these treatments is often limited by the heterogeneous and hypoxic tumor microenvironment (TME) of solid tumors. In the TME, programmed death-ligand 1 (PD-L1) expression on cancer cells is mainly regulated by Interferon-gamma (IFN-γ), which induces T cell exhaustion and enables tumor immune evasion. In this study, we demonstrate that acidosis, a common characteristic of solid tumors, significantly increases IFN-γ-induced PD-L1 expression on aggressive cancer cells, thus promoting immune escape. Using preclinical models, we found that acidosis enhances the genomic expression and phosphorylation of signal transducer and activator of transcription 1 (STAT1), and the translation of STAT1 mRNA by eukaryotic initiation factor 4F (elF4F), resulting in an increased PD-L1 expression. We observed this effect in murine and human anti-PD-L1-responsive tumor cell lines, but not in anti-PD-L1-nonresponsive tumor cell lines. In vivo studies fully validated our in vitro findings and revealed that neutralizing the acidic extracellular tumor pH by sodium bicarbonate treatment suppresses IFN-γ-induced PD-L1 expression and promotes immune cell infiltration in responsive tumors and thus reduces tumor growth. However, this effect was not observed in anti-PD-L1-nonresponsive tumors. In vivo experiments in tumor-bearing IFN-γ-/- mice validated the dependency on immune cell-derived IFN-γ for acidosis-mediated cancer cell PD-L1 induction and tumor immune escape. Thus, acidosis and IFN-γ-induced elevation of PD-L1 expression on cancer cells represent a previously unknown immune escape mechanism that may serve as a novel biomarker for anti-PD-L1/PD-1 treatment response. These findings have important implications for the development of new strategies to enhance the efficacy of immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Philipp Knopf
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Dimitri Stowbur
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
| | - Sabrina H L Hoffmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Natalie Hermann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
| | - Valentina Bucher
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Marilena Poxleitner
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Bredi Tako
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanhita Sinharay
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | | | - Irene Gonzalez-Menendez
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Institute of Pathology and Neuropathology, Department of Pathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, Tübingen University Hospital, Tübingen, Germany
| | - Felix Reckmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - David Bomze
- Department of Dermatology, Tel-Aviv Medical Center, Tel-Aviv, Israel
| | - Lukas Flatz
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Daniela Kramer
- Interfaculty Institute of Biochemistry, Eberhard Karls University, Tübingen, Germany
| | - Martin Schaller
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | | | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Leticia Quintanilla-Martinez
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Institute of Pathology and Neuropathology, Department of Pathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, Tübingen University Hospital, Tübingen, Germany
| | - Klaus Schulze-Osthoff
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Interfaculty Institute of Biochemistry, Eberhard Karls University, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Mark D Pagel
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Martin Röcken
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - André F Martins
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 10117, Berlin, Germany
| | - Manfred Kneilling
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany.
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
9
|
Minz AP, Mohapatra D, Dutta M, Sethi M, Parida D, Mohapatra AP, Mishra S, Kar S, Sasmal PK, Senapati S. Statins abrogate gemcitabine-induced PD-L1 expression in pancreatic cancer-associated fibroblasts and cancer cells with improved therapeutic outcome. Cancer Immunol Immunother 2023; 72:4261-4278. [PMID: 37926727 PMCID: PMC10992415 DOI: 10.1007/s00262-023-03562-9] [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: 06/18/2023] [Accepted: 10/17/2023] [Indexed: 11/07/2023]
Abstract
A combination of chemotherapy with immunotherapy has been proposed to have better clinical outcomes in Pancreatic Ductal Adenocarcinoma (PDAC). On the other hand, chemotherapeutics is known to have certain unwanted effects on the tumor microenvironment that may mask the expected beneficial effects of immunotherapy. Here, we have investigated the effect of gemcitabine (GEM), on two immune checkpoint proteins (PD-L1 and PD-L2) expression in cancer associated fibroblasts (CAFs) and pancreatic cancer cells (PCCs). Findings of in vitro studies conducted by using in-culture activated mouse pancreatic stellate cells (mPSCs) and human PDAC patients derived CAFs demonstrated that GEM significantly induces PD-L1 and PD-L2 expression in these cells. Moreover, GEM induced phosphorylation of STAT1 and production of multiple known PD-L1-inducing secretory proteins including IFN-γ in CAFs. Upregulation of PD-L1 in PSCs/CAFs upon GEM treatment caused T cell inactivation and apoptosis in vitro. Importantly, Statins suppressed GEM-induced PD-L1 expression both in CAFs and PCCs while abrogating the inactivation of T-cells caused by GEM-treated PSCs/CAFs. Finally, in an immunocompetent syngeneic orthotopic mouse pancreatic tumor model, simvastatin and GEM combination therapy significantly reduced intra-tumor PD-L1 expression and noticeably reduced the overall tumor burden and metastasis incidence. Together, the findings of this study have provided experimental evidence that illustrates potential unwanted side effects of GEM that could hamper the effectiveness of this drug as mono and/or combination therapy. At the same time the findings also suggest use of statins along with GEM will help in overcoming these shortcomings and warrant further clinical investigation.
Collapse
Affiliation(s)
- Aliva Prity Minz
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Debasish Mohapatra
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- CV Raman Global University, Bhubaneswar, Odisha, India
| | - Madhuri Dutta
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Manisha Sethi
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Deepti Parida
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Amlan Priyadarshee Mohapatra
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Swayambara Mishra
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Salona Kar
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Prakash K Sasmal
- Department of General Surgery, All India Institute of Medical Sciences, Bhubaneswar, India
| | | |
Collapse
|
10
|
Siegmund D, Zaitseva O, Wajant H. Fn14 and TNFR2 as regulators of cytotoxic TNFR1 signaling. Front Cell Dev Biol 2023; 11:1267837. [PMID: 38020877 PMCID: PMC10657838 DOI: 10.3389/fcell.2023.1267837] [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: 07/27/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Tumor necrosis factor (TNF) receptor 1 (TNFR1), TNFR2 and fibroblast growth factor-inducible 14 (Fn14) belong to the TNF receptor superfamily (TNFRSF). From a structural point of view, TNFR1 is a prototypic death domain (DD)-containing receptor. In contrast to other prominent death receptors, such as CD95/Fas and the two TRAIL death receptors DR4 and DR5, however, liganded TNFR1 does not instruct the formation of a plasma membrane-associated death inducing signaling complex converting procaspase-8 into highly active mature heterotetrameric caspase-8 molecules. Instead, liganded TNFR1 recruits the DD-containing cytoplasmic signaling proteins TRADD and RIPK1 and empowers these proteins to trigger cell death signaling by cytosolic complexes after their release from the TNFR1 signaling complex. The activity and quality (apoptosis versus necroptosis) of TNF-induced cell death signaling is controlled by caspase-8, the caspase-8 regulatory FLIP proteins, TRAF2, RIPK1 and the RIPK1-ubiquitinating E3 ligases cIAP1 and cIAP2. TNFR2 and Fn14 efficiently recruit TRAF2 along with the TRAF2 binding partners cIAP1 and cIAP2 and can thereby limit the availability of these molecules for other TRAF2/cIAP1/2-utilizing proteins including TNFR1. Accordingly, at the cellular level engagement of TNFR2 or Fn14 inhibits TNFR1-induced RIPK1-mediated effects reaching from activation of the classical NFκB pathway to induction of apoptosis and necroptosis. In this review, we summarize the effects of TNFR2- and Fn14-mediated depletion of TRAF2 and the cIAP1/2 on TNFR1 signaling at the molecular level and discuss the consequences this has in vivo.
Collapse
Affiliation(s)
| | | | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
11
|
Yuan H, You Y, He Y, Wei Y, Zhang Y, Min H, Li C, Chen J. Crystalline Silica-Induced Proinflammatory Interstitial Macrophage Recruitment through Notch3 Signaling Promotes the Pathogenesis of Silicosis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:14502-14514. [PMID: 37721423 DOI: 10.1021/acs.est.3c03980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Crystalline silica (CS) particles are ubiquitous in the environment, especially in occupational conditions, and exposure to respirable CS causes silicosis. The initial response to CS is mediated by innate immunity, where pulmonary macrophages act as central orchestrators. However, the repercussions of CS on functionally distinct macrophage subsets remain to be inconclusive. Herein, to study the effects of inhaled CS, we divided macrophages into three subsets: circulating monocytes, interstitial macrophages (IMs), and alveolar macrophages (AMs). CS-induced massive IMs increase in the lung, the phenotype and function of which differed from those of tissue-resident AMs and circulating monocytes. The augmented IMs were driven by recruitment of circulating macrophages rather than cell proliferation in situ. Moreover, the IMs predominantly exerted a classic activated (M1) phenotype and expressed proinflammatory cytokines, contributing to CS-induced lung injury. Notably, we demonstrated that IMs augmented Notch3 expression. Mechanistically, using myeloid-specific Notch3-knockout mice, we demonstrated that Notch3 signaling not only promoted IMs recruitment by regulating CCR2 expression but also manipulated the proinflammatory phenotype. Mice with conditional Notch3-knockout exhibited alleviation of CS-induced inflammation and fibrosis in lung. Overall, our study identifies IMs as critical mediators in response to CS and highlights the role of Notch3 in IMs recruitment and activation, providing new insights into CS toxicological effects in the lung.
Collapse
Affiliation(s)
- Haoyang Yuan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yichuan You
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yangyang He
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yungeng Wei
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yuting Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Chao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Jie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| |
Collapse
|
12
|
Chang CY, Armstrong D, Corry DB, Kheradmand F. Alveolar macrophages in lung cancer: opportunities challenges. Front Immunol 2023; 14:1268939. [PMID: 37822933 PMCID: PMC10562548 DOI: 10.3389/fimmu.2023.1268939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
Alveolar macrophages (AMs) are critical components of the innate defense mechanism in the lung. Nestled tightly within the alveoli, AMs, derived from the yolk-sac or bone marrow, can phagocytose foreign particles, defend the host against pathogens, recycle surfactant, and promptly respond to inhaled noxious stimuli. The behavior of AMs is tightly dependent on the environmental cues whereby infection, chronic inflammation, and associated metabolic changes can repolarize their effector functions in the lungs. Several factors within the tumor microenvironment can re-educate AMs, resulting in tumor growth, and reducing immune checkpoint inhibitors (ICIs) efficacy in patients treated for non-small cell lung cancer (NSCLC). The plasticity of AMs and their critical function in altering tumor responses to ICIs make them a desirable target in lung cancer treatment. New strategies have been developed to target AMs in solid tumors reprograming their suppressive function and boosting the efficacy of ICIs. Here, we review the phenotypic and functional changes in AMs in response to sterile inflammation and in NSCLC that could be critical in tumor growth and metastasis. Opportunities in altering AMs' function include harnessing their potential function in trained immunity, a concept borrowed from memory response to infections, which could be explored therapeutically in managing lung cancer treatment.
Collapse
Affiliation(s)
- Cheng-Yen Chang
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Dominique Armstrong
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - David B. Corry
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, United States
| | - Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, United States
| |
Collapse
|
13
|
Singh J, Minz RW, Saikia B, Nada R, Sharma A, Jha S, Anand S, Rathi M, D'Cruz S. Diminished PD-L1 regulation along with dysregulated T lymphocyte subsets and chemokine in ANCA-associated vasculitis. Clin Exp Med 2023; 23:1801-1813. [PMID: 36219364 DOI: 10.1007/s10238-022-00908-y] [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: 06/22/2022] [Accepted: 09/29/2022] [Indexed: 11/03/2022]
Abstract
ANCA-associated vasculitis (AAV) is a life-threatening disease characterized by small vessel inflammation and pathogenic self-directed antibodies. Programmed death-ligand 1 receptor (PD-1) and programmed cell death ligand-1 (PD-L1) are immune checkpoint molecules crucial for maintaining tolerance and immune homeostasis. After checkpoint inhibition therapy, development of various autoimmune diseases and immune-related adverse events (irAEs) have been observed. Here, we investigated the immunomodulatory roles of neutrophils through the expression of immune checkpoint molecule (PD-L1), migratory molecules (CXCR2), chemotactic chemokines (CXCL5) and other important molecules (BAFF and HMGB1) in development of AAV. We also scrutinized the immune mechanism responsible for development of pauci-immune crescentic GN (PICGN). We demonstrate for the first time that the frequency of PD-L1 expressing neutrophils was significantly reduced in AAV patients compared to healthy controls and correlated negatively with disease severity (BVASv3). Further, in renal biopsy, reduced PD-L1 immune checkpoint expression provides a microenvironment that unleashes uncontrolled activated CD4 + T cells, B cells, neutrophils and macrophages and ultimately causes engulfment of immune complexes leading to PICGN. Furthermore, during remission, reduced neutrophils PD-L1 and CXCR2 expression, increased neutrophils CXCL5 expression and increased peripheral effector memory T cells and increased HMGB1 and BAFF levels in serum, demonstrate the propensity for the persistence of sub-clinical inflammation, which could explain relapse, in this group of diseases.
Collapse
Affiliation(s)
- Jagdeep Singh
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Ranjana Walker Minz
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| | - Biman Saikia
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Ritambhra Nada
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Aman Sharma
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Saket Jha
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Shashi Anand
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Manish Rathi
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Sanjay D'Cruz
- Department of General Medicine, Government Medical College and Hospital, Chandigarh, 160030, India
| |
Collapse
|
14
|
Fruntealată RF, Marius M, Boboc IKS, Mitran SI, Ciurea ME, Stoica GA. Mechanisms of Altered Immune Response in Skin Melanoma. CURRENT HEALTH SCIENCES JOURNAL 2023; 49:297-311. [PMID: 38314217 PMCID: PMC10832881 DOI: 10.12865/chsj.49.03.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/18/2023] [Indexed: 02/06/2024]
Abstract
Melanoma, a deadly form of skin cancer, poses significant challenges to the host immune system, allowing tumor cells to evade immune surveillance and persist. This complex interplay between melanoma and the immune system involves a multitude of mechanisms that impair immune recognition and promote tumor progression. This review summarizes the intricate strategies employed by melanoma cells to evade the immune response, including defective immune recognition, immune checkpoint activation, and the role of regulatory T-cells, myeloid-derived suppressor cells, and exosomes in suppressing anti-tumor immunity. Additionally, we discuss potential therapeutic targets aimed at reversing immune evasion in melanoma, highlighting the importance of understanding these mechanisms for developing more effective immunotherapies. Improved insights into the interactions between melanoma and the immune system will aid in the development of novel treatment strategies to enhance anti-tumor immune responses and improve patient outcomes.
Collapse
Affiliation(s)
| | - Matei Marius
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Ianis Kevyn Stefan Boboc
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Romania
| | | | - Marius Eugen Ciurea
- Department of Physiology, University of Medicine and Pharmacy of Craiova, Romania
| | - George-Alin Stoica
- Department of Pediatric Surgery, University of Medicine and Pharmacy of Craiova, Romania
| |
Collapse
|
15
|
Sutherland SIM, Ju X, Silveira PA, Kupresanin F, Horvath LG, Clark GJ. CD300f signalling induces inhibitory human monocytes/macrophages. Cell Immunol 2023; 390:104731. [PMID: 37302321 DOI: 10.1016/j.cellimm.2023.104731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 05/11/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
The CD300 glycoproteins are a family of related leucocyte surface molecules that regulate the immune response via their paired triggering and inhibitory receptors. Here we studied CD300f, an apoptotic cell receptor, and how it modulates the function of human monocytes and macrophages. We showed that CD300f signalling by crosslinking with anti-CD300f mAb (DCR-2) suppressed monocytes causing upregulation of the inhibitory molecule, CD274 (PD-L1) and their inhibition of T cell proliferation. Furthermore, CD300f signalling drove macrophages preferentially towards M2-type with upregulation of CD274, which was further enhanced by IL-4. CD300f signalling activates the PI3K/Akt pathway in monocytes. Inhibition of PI3K/Akt signalling resulting from CD300f crosslinking leads to downregulation of CD274 expression on monocytes. These findings highlight the potential use of CD300f blockade in cancer immune therapy to target immune suppressive macrophages in the tumour microenvironment, a known resistance mechanism to PD-1/PD-L1 checkpoint inhibitors.
Collapse
Affiliation(s)
- Sarah I M Sutherland
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | - Xinsheng Ju
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Pablo A Silveira
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Fiona Kupresanin
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
| | - Lisa G Horvath
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Chris O'Brien Lifehouse, Sydney, NSW, Australia; Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Georgina J Clark
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Xu S, Wang C, Yang L, Wu J, Li M, Xiao P, Xu Z, Xu Y, Wang K. Targeting immune checkpoints on tumor-associated macrophages in tumor immunotherapy. Front Immunol 2023; 14:1199631. [PMID: 37313405 PMCID: PMC10258331 DOI: 10.3389/fimmu.2023.1199631] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/16/2023] [Indexed: 06/15/2023] Open
Abstract
Unprecedented breakthroughs have been made in cancer immunotherapy in recent years. Particularly immune checkpoint inhibitors have fostered hope for patients with cancer. However, immunotherapy still exhibits certain limitations, such as a low response rate, limited efficacy in certain populations, and adverse events in certain tumors. Therefore, exploring strategies that can improve clinical response rates in patients is crucial. Tumor-associated macrophages (TAMs) are the predominant immune cells that infiltrate the tumor microenvironment and express a variety of immune checkpoints that impact immune functions. Mounting evidence indicates that immune checkpoints in TAMs are closely associated with the prognosis of patients with tumors receiving immunotherapy. This review centers on the regulatory mechanisms governing immune checkpoint expression in macrophages and strategies aimed at improving immune checkpoint therapies. Our review provides insights into potential therapeutic targets to improve the efficacy of immune checkpoint blockade and key clues to developing novel tumor immunotherapies.
Collapse
Affiliation(s)
- Shumin Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Chenyang Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lingge Yang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Jiaji Wu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Mengshu Li
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Peng Xiao
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Yun Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|
17
|
Han J, Dong L, Wu M, Ma F. Dynamic polarization of tumor-associated macrophages and their interaction with intratumoral T cells in an inflamed tumor microenvironment: from mechanistic insights to therapeutic opportunities. Front Immunol 2023; 14:1160340. [PMID: 37251409 PMCID: PMC10219223 DOI: 10.3389/fimmu.2023.1160340] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/20/2023] [Indexed: 05/31/2023] Open
Abstract
Immunotherapy has brought a paradigm shift in the treatment of tumors in recent decades. However, a significant proportion of patients remain unresponsive, largely due to the immunosuppressive tumor microenvironment (TME). Tumor-associated macrophages (TAMs) play crucial roles in shaping the TME by exhibiting dual identities as both mediators and responders of inflammation. TAMs closely interact with intratumoral T cells, regulating their infiltration, activation, expansion, effector function, and exhaustion through multiple secretory and surface factors. Nevertheless, the heterogeneous and plastic nature of TAMs renders the targeting of any of these factors alone inadequate and poses significant challenges for mechanistic studies and clinical translation of corresponding therapies. In this review, we present a comprehensive summary of the mechanisms by which TAMs dynamically polarize to influence intratumoral T cells, with a focus on their interaction with other TME cells and metabolic competition. For each mechanism, we also discuss relevant therapeutic opportunities, including non-specific and targeted approaches in combination with checkpoint inhibitors and cellular therapies. Our ultimate goal is to develop macrophage-centered therapies that can fine-tune tumor inflammation and empower immunotherapy.
Collapse
Affiliation(s)
- Jiashu Han
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, China
| | - Luochu Dong
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Fei Ma
- Center for National Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Massicano AVF, Song PN, Mansur A, White SL, Sorace AG, Lapi SE. [ 89Zr]-Atezolizumab-PET Imaging Reveals Longitudinal Alterations in PDL1 during Therapy in TNBC Preclinical Models. Cancers (Basel) 2023; 15:2708. [PMID: 37345044 PMCID: PMC10216761 DOI: 10.3390/cancers15102708] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Triple-negative breast cancers (TNBCs) currently have limited treatment options; however, PD-L1 is an indicator of susceptibility to immunotherapy. Currently, assessment of PD-L1 is limited to biopsy samples. These limitations may be overcome with molecular imaging. In this work, we describe chemistry development and optimization, in vitro, in vivo, and dosimetry of [89Zr]-Atezolizumab for PD-L1 imaging. Atezolizumab was conjugated to DFO and radiolabeled with 89Zr. Tumor uptake and heterogeneity in TNBC xenograft and patient-derived xenograft (PDX) mouse models were quantified following [89Zr]-Atezolizumab-PET imaging. PD-L1 expression in TNBC PDX models undergoing therapy and immunohistochemistry (IHC) was used to validate imaging. SUV from PET imaging was quantified and used to identify heterogeneity. PET/CT imaging using [89Zr]-Atezolizumab identified a significant increase in tumor:muscle SUVmean 1 and 4 days after niraparib therapy and revealed an increased trend in PD-L1 expression following other cytotoxic therapies. A preliminary dosimetry study indicated the organs that will receive a higher dose are the spleen, adrenals, kidneys, and liver. [89Zr]-Atezolizumab PET/CT imaging reveals potential for the noninvasive detection of PD-L1-positive TNBC tumors and allows for quantitative and longitudinal assessment. This has potential significance for understanding tumor heterogeneity and monitoring early expression changes in PD-L1 induced by therapy.
Collapse
Affiliation(s)
| | - Patrick N. Song
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Ameer Mansur
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Sharon L. White
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Anna G. Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Suzanne E. Lapi
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
19
|
Bruni S, Mauro FL, Proietti CJ, Cordo-Russo RI, Rivas MA, Inurrigarro G, Dupont A, Rocha D, Fernández EA, Deza EG, Lopez Della Vecchia D, Barchuk S, Figurelli S, Lasso D, Friedrich AD, Santilli MC, Regge MV, Lebersztein G, Levit C, Anfuso F, Castiglione T, Elizalde PV, Mercogliano MF, Schillaci R. Blocking soluble TNFα sensitizes HER2-positive breast cancer to trastuzumab through MUC4 downregulation and subverts immunosuppression. J Immunother Cancer 2023; 11:jitc-2022-005325. [PMID: 36889811 PMCID: PMC10016294 DOI: 10.1136/jitc-2022-005325] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND The success of HER2-positive (HER2+) breast cancer treatment with trastuzumab, an antibody that targets HER2, relies on immune response. We demonstrated that TNFα induces mucin 4 (MUC4) expression, which shields the trastuzumab epitope on the HER2 molecule decreasing its therapeutic effect. Here, we used mouse models and samples from HER2+ breast cancer patients to unravel MUC4 participation in hindering trastuzumab effect by fostering immune evasion. METHODS We used a dominant negative TNFα inhibitor (DN) selective for soluble TNFα (sTNFα) together with trastuzumab. Preclinical experiments were performed using two models of conditionally MUC4-silenced tumors to characterize the immune cell infiltration. A cohort of 91 patients treated with trastuzumab was used to correlate tumor MUC4 with tumor-infiltrating lymphocytes. RESULTS In mice bearing de novo trastuzumab-resistant HER2+ breast tumors, neutralizing sTNFα with DN induced MUC4 downregulation. Using the conditionally MUC4-silenced tumor models, the antitumor effect of trastuzumab was reinstated and the addition of TNFα-blocking agents did not further decrease tumor burden. DN administration with trastuzumab modifies the immunosuppressive tumor milieu through M1-like phenotype macrophage polarization and NK cells degranulation. Depletion experiments revealed a cross-talk between macrophages and NK cells necessary for trastuzumab antitumor effect. In addition, tumor cells treated with DN are more susceptible to trastuzumab-dependent cellular phagocytosis. Finally, MUC4 expression in HER2+ breast cancer is associated with immune desert tumors. CONCLUSIONS These findings provide rationale to pursue sTNFα blockade combined with trastuzumab or trastuzumab drug conjugates for MUC4+ and HER2+ breast cancer patients to overcome trastuzumab resistance.
Collapse
Affiliation(s)
- Sofia Bruni
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Florencia L Mauro
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Cecilia J Proietti
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Rosalia I Cordo-Russo
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Martin A Rivas
- Division of Hematology & Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | | | - Agustina Dupont
- Servicio de Patología, Sanatorio Mater Dei, Buenos Aires, Argentina
| | - Dario Rocha
- Bioscience Data Mining Group at CIDIE-CONICET-UCC, Córdoba, Argentina
| | - Elmer A Fernández
- Bioscience Data Mining Group at CIDIE-CONICET-UCC, Córdoba, Argentina
| | | | | | - Sabrina Barchuk
- Sección Patología Mamaria Hospital General de Agudos "Juan A Fernández, Buenos Aires, Argentina
| | - Silvina Figurelli
- Servicio de Patología, Hospital General de Agudos "Juan A. Fernández,", Buenos Aires, Argentina
| | - David Lasso
- Hospital Oncológico Provincial de Córdoba, Córdoba, Argentina
| | - Adrián D Friedrich
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - María C Santilli
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - María V Regge
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | | | - Claudio Levit
- Servicio de Cirugía, Sanatorio Sagrado Corazón, Buenos Aires, Argentina
| | - Fabiana Anfuso
- Servicio de Cirugía, Sanatorio Sagrado Corazón, Buenos Aires, Argentina
| | | | - Patricia V Elizalde
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Maria F Mercogliano
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Roxana Schillaci
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| |
Collapse
|
20
|
Chen L, Deng J. Role of non-coding RNA in immune microenvironment and anticancer therapy of gastric cancer. J Mol Med (Berl) 2022; 100:1703-1719. [PMID: 36329206 DOI: 10.1007/s00109-022-02264-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
Gastric cancer remains one of the cancers with the highest mortality in the world; therefore, it is very important to investigate its pathogenesis to improve the prognosis of gastric cancer patients. Recently, noncoding RNAs have become a research hotspot in the field of oncology. These RNA molecules play complex roles in the regulation of tumor cells, immune cells, and the tumor microenvironment. Therefore, studying their ability to regulate the gastric cancer immune microenvironment will provide us with a better perspective to understand their potential role in anticancer therapy. In this review, we discuss the regulatory effects of several common noncoding RNAs on the immune microenvironment of gastric cancer and their prospects in anticancer therapy to provide some novel insight into the identification of valuable diagnostic markers and improving the prognosis of gastric cancer patients.
Collapse
Affiliation(s)
- Liqiao Chen
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Jingyu Deng
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China.
| |
Collapse
|
21
|
Wen Y, Wang X, Meng W, Guo W, Duan C, Cao J, Kang L, Guo N, Lin Q, Lv P, Zhang R, Xing L, Zhang X, Shen H. TNF-α-dependent lung inflammation upregulates PD-L1 in monocyte-derived macrophages to contribute to lung tumorigenesis. FASEB J 2022; 36:e22595. [PMID: 36205325 DOI: 10.1096/fj.202200434rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/20/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022]
Abstract
Chronic inflammation, which is dominated by macrophage-involved inflammatory responses, is an instigator of cancer initiation. Macrophages are the most abundant immune cells in healthy lungs, and associated with lung tumor development and promotion. PD-L1 is a negative molecule in macrophages and correlated with an immunosuppressive function in tumor environment. Macrophages expressing PD-L1, rather than tumor cells, exhibits a critical role in tumor growth and progression. However, whether and how PD-L1 in macrophages contributes to inflammation-induced lung tumorigenesis requires further elucidation. Here, we found that higher expression of PD-L1 in CD11b+ CD206+ macrophages was positively correlated with tumor progression and PD-1+ CD8+ T cells population in human adenocarcinoma patients. In the urethane-induced inflammation-driven lung adenocarcinoma (IDLA) mouse model, the infiltration of circulating CD11bhigh F4/80+ monocyte-derived macrophages (MoMs) was increased in pro-tumor inflamed lung tissues and lung adenocarcinoma. PD-L1 was mainly upregulated in MoMs associated with enhanced T cells exhaustion in lung tissues. Anti-PD-L1 treatment can reduce T cells exhaustion at pro-tumor inflammatory stage, and then inhibit tumorigenesis in IDLA. The pro-tumor lung inflammation depended on TNF-α to upregulate PD-L1 and CSN6 expression in MoMs, and induced cytokines production by alveolar type-II cells (AT-II). Furthermore, inflammatory AT-II cells could secret TNF-α to upregulate PD-L1 expression in bone-marrow driven macrophages (BM-M0). Inhibition of CSN6 decreased PD-L1 expression in TNF-α-activated macrophage in vitro, suggesting a critical role of CSN6 in PD-L1 upregulation. Thus, pro-tumor inflammation can depend on TNF-α to upregulate PD-L1 in recruited MoMs, which may be essential for lung tumorigenesis.
Collapse
Affiliation(s)
- Yue Wen
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Department of Ultrasound, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiuqing Wang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Wei Meng
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Wenli Guo
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Chenyang Duan
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Jingjing Cao
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Lifei Kang
- Department of Pathology, Hebei Chest Hospital, Shijiazhuang, China
| | - Ningfei Guo
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Qiang Lin
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, Renqiu, China
| | - Ping Lv
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Lingxiao Xing
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Xianghong Zhang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China.,Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
22
|
Tojo M, Horie H, Koinuma K, Miyato H, Tsukui H, Kaneko Y, Futoh Y, Kimura Y, Takahashi K, Saito A, Ohzawa H, Yamaguchi H, Lefor AK, Sata N, Kitayama J. Programmed cell death ligand 1 expression on monocytes is inversely correlated with tumour response to preoperative chemoradiotherapy for locally advanced rectal cancer. Colorectal Dis 2022; 24:1140-1149. [PMID: 35502766 PMCID: PMC9790410 DOI: 10.1111/codi.16167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/17/2022] [Accepted: 04/23/2022] [Indexed: 12/30/2022]
Abstract
AIM The clinical efficacy of chemoradiotherapy (CRT) is largely dependent on host immune status. The aim of this study was to identify possible markers expressed on circulating mononuclear cells to predict tumour response in patients with locally advanced rectal cancer (LARC). METHODS Peripheral blood samples were obtained from 47 patients diagnosed with LARC before and after CRT. The numbers of lymphocytes and monocyte subsets were analysed using flow cytometry. Based on clinical and pathological findings, patients were classified as high or low responders. RESULTS Lymphocyte counts were markedly decreased after CRT. Total numbers of lymphocytes (p = 0.030) and CD4(+) T cells (p = 0.041) in post-CRT samples were significantly lower in low responders than in high responders. In contrast, monocyte counts were not reduced and the number of CD14dim (+) CD16(+) nonclassical (patrolling) monocytes were somewhat increased after CRT (p = 0.050). Moreover, the ratios of programmed cell death ligand 1 (PD-L1) (+) cells on patrolling monocytes before and after CRT were significantly higher in low responders than in high responders (p = 0.0046, p = 0.0006). The same trend was observed for classical and intermediate monocytes. The expression of PD-L1 on patrolling monocytes before CRT correlated inversely with the number of T cells and natural killer (NK) cells after CRT. PD-L1(+) ratio in patrolling monocytes was an independent predictor for response to CRT. CONCLUSION Programmed cell death ligand 1 (PD-L1) expression on patrolling monocytes suppresses cell-mediated immunity in patients receiving CRT which could be related to tumour response, and may be a useful biomarker for decision-making in the management of patients with LARC.
Collapse
Affiliation(s)
- Mineyuki Tojo
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Hisanaga Horie
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Koji Koinuma
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Hideyo Miyato
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan,Jichi Medical University HospitalDivision of Translational Research, Center for Clinical ResearchShimotsukeTochigiJapan
| | - Hidenori Tsukui
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Yuki Kaneko
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Yurie Futoh
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Yuki Kimura
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Kazuya Takahashi
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Akira Saito
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Hideyuki Ohzawa
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan,Department of Clinical OncologyJichi Medical UniversityShimotsukeTochigiJapan
| | - Hironori Yamaguchi
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan,Department of Clinical OncologyJichi Medical UniversityShimotsukeTochigiJapan
| | | | - Naohiro Sata
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Joji Kitayama
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan,Jichi Medical University HospitalDivision of Translational Research, Center for Clinical ResearchShimotsukeTochigiJapan
| |
Collapse
|
23
|
The integration of systemic and tumor PD-L1 as a predictive biomarker of clinical outcomes in patients with advanced NSCLC treated with PD-(L)1blockade agents. Cancer Immunol Immunother 2022; 71:1823-1835. [PMID: 34984538 DOI: 10.1007/s00262-021-03107-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 11/05/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Tumor PD-L1 expression is a predictive biomarker for patients with NSCLC receiving PD-(L)1 blockade agents. However, although increased tumor PD-L1 expression predicts responsiveness, clinical benefit has been observed regardless of tumor PD-L1 expression, suggesting the existence of other PD-L1 sources. The aim of our study was to analyze whether integrating systemic and tumor PD-L1 is more predictive of efficacy in patients with advanced NSCLC receiving PD-(L)1 blockade agents. MATERIAL AND METHODS Twenty-nine healthy donors and 119 consecutive patients with advanced NSCLC treated with PD-(L)1 drug were prospectively included. Pretreatment blood samples were collected to evaluate PD-L1 levels on circulating immune cells, platelets (PLTs), platelet microparticles (PMPs), and the plasma soluble PD-L1 concentration (sPD-L1). Tumor PD-L1 status was assessed by immunohistochemistry. The percentages of circulating PD-L1 + leukocytes, sPD-L1 levels, and tumor PD-L1 were correlated with efficacy. RESULTS No differences in the percentages of circulating PD-L1 + leukocytes were observed according to tumor PD-L1 expression. Significantly longer progression-free survival was observed in patients with higher percentages of PD-L1 + CD14 + , PD-L1 + neutrophils, PD-L1 + PLTs, and PD-L1 + PMPs and significantly longer overall survival was observed in patients with higher percentages of PD-L1 + CD14 + and high tumor PD-L1 expression. Integrating the PD-L1 data of circulating and tumor PD-L1 results significantly stratified patients according to the efficacy of PD-(L1) blockade agents. CONCLUSIONS Our results suggest that integrating circulating PD-L1 + leukocytes, PLT, PMPs, and sPD-L1 and tumor PD-L1 expression may be helpful to decide on the best treatment strategy in patients with advanced NSCLC who are candidates for PD-(L)1 blockade agents.
Collapse
|
24
|
Li W, Wu F, Zhao S, Shi P, Wang S, Cui D. Correlation between PD-1/PD-L1 expression and polarization in tumor-associated macrophages: A key player in tumor immunotherapy. Cytokine Growth Factor Rev 2022; 67:49-57. [PMID: 35871139 DOI: 10.1016/j.cytogfr.2022.07.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/13/2022]
Abstract
Tumor immunotherapy, such as PD-1/PD-L1 blockade, has shown promising clinical efficacy in patients with various types of tumors. However, the response to PD-1/PD-L1 blockade in a majority of malignancies is limited, indicating an urgent need for a deeper understanding of the mechanisms of PD-1/PD-L1 axis-mediated tumor tolerance. As the most abundant immune cells in the tumor stroma, macrophages display multiple phenotypes and functions in response to the stimuli of the tumor microenvironment. PD-1/PD-L1 has been demonstrated to be highly expressed in tumor-associated macrophages (TAMs), and TAM polarization has been shown to be important during tumor progression. In this review, we outline the relationship between TAM PD-1/PD-L1 expression and polarizations, summarize the involvement of M2 TAMs in PD-1/PD-L1-mediated T-cell exhaustion, and discuss improved approaches for overcoming PD-1/PD-L1 blockade resistance by inducing M2/M1 switching of TAMs.
Collapse
Affiliation(s)
- Wei Li
- Center of Research Laboratory, Department of Laboratory Medicine, The First People's Hospital of Lianyungang, Lianyungang, China.
| | - Fenglei Wu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Shaolin Zhao
- Center of Research Laboratory, Department of Laboratory Medicine, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Peiqin Shi
- Center of Research Laboratory, Department of Laboratory Medicine, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
25
|
Numata Y, Akutsu N, Ishigami K, Koide H, Wagatsuma K, Motoya M, Sasaki S, Nakase H. Synergistic effect of IFN-γ and IL-1β on PD-L1 expression in hepatocellular carcinoma. Biochem Biophys Rep 2022; 30:101270. [PMID: 35573813 PMCID: PMC9095738 DOI: 10.1016/j.bbrep.2022.101270] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/15/2022] [Accepted: 04/26/2022] [Indexed: 12/30/2022] Open
Abstract
Immunotherapy using anti-programmed death 1 ligand 1 (PD-L1) antibodies has shown clinical efficacy against hepatocellular carcinoma (HCC) and is recognized as the first-line treatment for unresectable HCC. PD-L1 expression is affected by various cytokines produced by immune cells in the tumor microenvironment; however, there is limited information about the effects of cytokine interactions on PD-L1 expression. In this study, we examined how cytokines induce PD-L1 expression in HCC cells. Both interferon gamma (IFN-γ) and interleukin 1 beta (IL-1β) induced PD-L1 expression, and the two cytokines enhanced PD-L1 expression in combination compared to that when administered alone. The Janus kinase/signal transducer and activator of transcription signaling pathway activated by IFN-γ is the major pathway of PD-L1 expression. The increase in interferon regulatory factor 1 expression and IFN-γ receptor expression induced by IL-1β was associated with the synergistic effect of IFN-γ and IL-1β on PD-L1 expression. These findings strongly indicate that IFN-γ and IL-1β affect the mechanism underlying immune resistance in HCC cells. IFN-γ and IL-1β synergistically increase the expression of PD-L1 in HCC cells. IFN-γ enhances PD-L1 expression via STAT1 signaling. IL-1β enhances PD-L1 expression via the NF-κB and the p38 MAPK pathways. IRF-1 and IFNGR also contribute to the synergistic effect of IFN-γ and IL-1β in HCC.
Collapse
Affiliation(s)
| | - Noriyuki Akutsu
- Corresponding author. Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan.
| | | | | | | | | | | | | |
Collapse
|
26
|
Multi-Omics Approaches for the Prediction of Clinical Endpoints after Immunotherapy in Non-Small Cell Lung Cancer: A Comprehensive Review. Biomedicines 2022; 10:biomedicines10061237. [PMID: 35740259 PMCID: PMC9219996 DOI: 10.3390/biomedicines10061237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized the management of locally advanced and advanced non-small lung cancer (NSCLC). With an improvement in the overall survival (OS) as both first- and second-line treatments, ICIs, and especially programmed-death 1 (PD-1) and programmed-death ligands 1 (PD-L1), changed the landscape of thoracic oncology. The PD-L1 level of expression is commonly accepted as the most used biomarker, with both prognostic and predictive values. However, even in a low expression level of PD-L1, response rates remain significant while a significant number of patients will experience hyperprogression or adverse events. The dentification of such subtypes is thus of paramount importance. While several studies focused mainly on the prediction of the PD-L1 expression status, others aimed directly at the development of prediction/prognostic models. The response to ICIs depends on a complex physiopathological cascade, intricating multiple mechanisms from the molecular to the macroscopic level. With the high-throughput extraction of features, omics approaches aim for the most comprehensive assessment of each patient. In this article, we will review the place of the different biomarkers (clinical, biological, genomics, transcriptomics, proteomics and radiomics), their clinical implementation and discuss the most recent trends projecting on the future steps in prediction modeling in NSCLC patients treated with ICI.
Collapse
|
27
|
De Ridder K, Locy H, Piccioni E, Zuazo MI, Awad RM, Verhulst S, Van Bulck M, De Vlaeminck Y, Lecocq Q, Reijmen E, De Mey W, De Beck L, Ertveldt T, Pintelon I, Timmermans JP, Escors D, Keyaerts M, Breckpot K, Goyvaerts C. TNF-α-Secreting Lung Tumor-Infiltrated Monocytes Play a Pivotal Role During Anti-PD-L1 Immunotherapy. Front Immunol 2022; 13:811867. [PMID: 35493461 PMCID: PMC9046849 DOI: 10.3389/fimmu.2022.811867] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/03/2022] [Indexed: 11/18/2022] Open
Abstract
Immune checkpoint blockade (ICB) of the PD-1 pathway revolutionized the survival forecast for advanced non-small cell lung cancer (NSCLC). Yet, the majority of PD-L1+ NSCLC patients are refractory to anti-PD-L1 therapy. Recent observations indicate a pivotal role for the PD-L1+ tumor-infiltrating myeloid cells in therapy failure. As the latter comprise a heterogenous population in the lung tumor microenvironment, we applied an orthotopic Lewis Lung Carcinoma (LLC) model to evaluate 11 different tumor-residing myeloid subsets in response to anti-PD-L1 therapy. While we observed significantly reduced fractions of tumor-infiltrating MHC-IIlow macrophages and monocytes, serological levels of TNF-α restored in lung tumor-bearing mice. Notably, we demonstrated in vivo and in vitro that anti-PD-L1 therapy mediated a monocyte-specific production of, and response to TNF-α, further accompanied by their significant upregulation of CD80, VISTA, LAG-3, SIRP-α and TIM-3. Nevertheless, co-blockade of PD-L1 and TNF-α did not reduce LLC tumor growth. A phenomenon that was partly explained by the observation that monocytes and TNF-α play a Janus-faced role in anti-PD-L1 therapy-mediated CTL stimulation. This was endorsed by the observation that monocytes appeared crucial to effectively boost T cell-mediated LLC killing in vitro upon combined PD-L1 with LAG-3 or SIRP-α blockade. Hence, this study enlightens the biomarker potential of lung tumor-infiltrated monocytes to define more effective ICB combination strategies.
Collapse
Affiliation(s)
- Kirsten De Ridder
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hanne Locy
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Elisa Piccioni
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Miren Ibarra Zuazo
- Immunomodulation Group, Navarrabiomed, Navarrabiomed-UPNA-IdISNA, Pamplona, Spain
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Stefaan Verhulst
- Liver Cell Biology Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Mathias Van Bulck
- Laboratory of Molecular and Medical Oncology, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Eva Reijmen
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Wout De Mey
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Lien De Beck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology & Histology, Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology & Histology, Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Antwerp, Belgium
| | - David Escors
- Immunomodulation Group, Navarrabiomed, Navarrabiomed-UPNA-IdISNA, Pamplona, Spain
- Rayne Institute, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
28
|
He Z, Khatib AM, Creemers JWM. The proprotein convertase furin in cancer: more than an oncogene. Oncogene 2022; 41:1252-1262. [PMID: 34997216 DOI: 10.1038/s41388-021-02175-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/13/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023]
Abstract
Furin is the first discovered proprotein convertase member and is present in almost all mammalian cells. Therefore, by regulating the maturation of a wide range of proproteins, Furin expression and/or activity is involved in various physiological and pathophysiological processes ranging from embryonic development to carcinogenesis. Since many of these protein precursors are involved in initiating and maintaining the hallmarks of cancer, Furin has been proposed as a potential target for treating several human cancers. In contrast, other studies have revealed that some types of cancer do not benefit from Furin inhibition. Therefore, understanding the heterogeneous functions of Furin in cancer will provide important insights into the design of effective strategies targeting Furin in cancer treatment. Here, we present recent advances in understanding how Furin expression and activity are regulated in cancer cells and their influences on the activity of Furin substrates in carcinogenesis. Furthermore, we discuss how Furin represses tumorigenic properties of several cancer cells and why Furin inhibition leads to aggressive phenotypes in other tumors. Finally, we summarize the clinical applications of Furin inhibition in treating human cancers.
Collapse
Affiliation(s)
- Zongsheng He
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Abdel-Majid Khatib
- INSERM, LAMC, UMR 1029, Allée Geoffroy St Hilaire, Pessac, France.
- Institut Bergoinié, Bordeaux, France.
| | - John W M Creemers
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Liu W, Stachura P, Xu HC, Váraljai R, Shinde P, Ganesh NU, Mack M, Van Lierop A, Huang A, Sundaram B, Lang KS, Picard D, Fischer U, Remke M, Homey B, Roesch A, Häussinger D, Lang PA, Borkhardt A, Pandyra AA. BAFF Attenuates Immunosuppressive Monocytes in the Melanoma Tumor Microenvironment. Cancer Res 2022; 82:264-277. [PMID: 34810198 PMCID: PMC9397630 DOI: 10.1158/0008-5472.can-21-1171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/17/2021] [Accepted: 11/15/2021] [Indexed: 01/07/2023]
Abstract
Emerging evidence indicates B-cell activating factor (BAFF, Tnfsf13b) to be an important cytokine for antitumor immunity. In this study, we generated a BAFF-overexpressing B16.F10 melanoma cell model and found that BAFF-expressing tumors grow more slowly in vivo than control tumors. The tumor microenvironment (TME) of BAFF-overexpressing tumors had decreased myeloid infiltrates with lower PD-L1 expression. Monocyte depletion and anti-PD-L1 antibody treatment confirmed the functional importance of monocytes for the phenotype of BAFF-mediated tumor growth delay. RNA sequencing analysis confirmed that monocytes isolated from BAFF-overexpressing tumors were characterized by a less exhaustive phenotype and were enriched for in genes involved in activating adaptive immune responses and NF-κB signaling. Evaluation of patients with late-stage metastatic melanoma treated with inhibitors of the PD-1/PD-L1 axis demonstrated a stratification of patients with high and low BAFF plasma levels. Patients with high BAFF levels experienced lower responses to anti-PD-1 immunotherapies. In summary, these results show that BAFF, through its effect on tumor-infiltrating monocytes, not only impacts primary tumor growth but can serve as a biomarker to predict response to anti-PD-1 immunotherapy in advanced disease. SIGNIFICANCE: The BAFF cytokine regulates monocytes in the melanoma microenvironment to suppress tumor growth, highlighting the importance of BAFF in antitumor immunity.
Collapse
Affiliation(s)
- Wei Liu
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Paweł Stachura
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Haifeng C. Xu
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Renáta Váraljai
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Prashant Shinde
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Nikkitha Umesh Ganesh
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Matthias Mack
- Department of Nephrology, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Anke Van Lierop
- Department of Dermatology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Anfei Huang
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Balamurugan Sundaram
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Karl S. Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany.,Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany
| | - Marc Remke
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany.,Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Bernhard Homey
- Department of Dermatology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University of Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Dieter Häussinger
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany
| | - Aleksandra A. Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany.,Corresponding Author: Aleksandra A. Pandyra, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Universitätsstraβe 1, Düsseldorf, 40225, Germany. E-mail:
| |
Collapse
|
30
|
Tamene W, Abebe M, Wassie L, Mollalign H, Bauer K, Kebede A, Marconi VC, Howe R, Sack U. PDL1 expression on monocytes is associated with plasma cytokines in Tuberculosis and HIV. PLoS One 2021; 16:e0258122. [PMID: 34597347 PMCID: PMC8486133 DOI: 10.1371/journal.pone.0258122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/20/2021] [Indexed: 11/19/2022] Open
Abstract
Introduction PDL1 and its interaction with PD1 is implicated in immune dysfunction in TB and HIV. The expression of PDL1 on multiple subsets of monocytes as well as their associations with cytokines and microbial products have not been well studied. Method HIV (TB-HIV+), TB (TB+HIV-) and TB/HIV co-infected (TB+HIV+) patients as well as apparently healthy controls (TB-HIV-) were recruited. TB and HIV patients were treatment naïve while TB/HIV patients were both ART naïve and experienced but not yet started TB therapy. Monocyte subsets were evaluated for PDL1 expression by flow cytometry; plasma TNFα, IL6, IP10, IFNγ and IL10 were measured by Luminex; and cytokine mRNA from purified monocytes quantitated by qPCR. The association of PDL1 with cytokines, clinical and microbial indices, including HIV viral load, TB smear microscopy and TB urinary lipoarabinomannan (LAM) were assessed. Results Monocyte expression of PDL1 was significantly higher in TB, HIV and TB/HIV co-infected patients compared with healthy controls (p = 0.0001), with the highest levels in TB/HIV co-infected patients. The highest expression of PDL1 was on intermediate (CD14+CD16+) monocytes in all participant groups. PDL1 strongly correlated with HIV viral load in TB/HIV while weakly correlated in HIV. PDL1 levels moderately correlated with plasma TNFα, IL6, IP10, IFNγ and IL10 level in TB subjects whereas weakly correlated with TNFα and IP10 in HIV patients. However, cytokine mRNA from purified monocytes showed no association with either plasma cytokines or monocyte PDL1 expression, implying that if cytokines modulate PDL1, they are likely not originating from circulating monocytes themselves. These results underscore the importance of further characterization of multiple monocyte subsets and their phenotypic and functional differences in different disease states.
Collapse
Affiliation(s)
- Wegene Tamene
- HIV and TB Research Directorate, Ethiopian Public Health Institute (EPHI), Addis Ababa, Ethiopia
- * E-mail: ,
| | - Meseret Abebe
- Mycobacterial Disease Research Directorate, Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Liya Wassie
- Mycobacterial Disease Research Directorate, Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Helina Mollalign
- HIV and TB Research Directorate, Ethiopian Public Health Institute (EPHI), Addis Ababa, Ethiopia
| | - Katrin Bauer
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Amha Kebede
- HIV and TB Research Directorate, Ethiopian Public Health Institute (EPHI), Addis Ababa, Ethiopia
| | - Vincent C. Marconi
- School of Medicine and Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Rawleigh Howe
- Mycobacterial Disease Research Directorate, Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Ulrich Sack
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
31
|
Lucas ED, Schafer JB, Matsuda J, Kraus M, Burchill MA, Tamburini BAJ. PD-L1 Reverse Signaling in Dermal Dendritic Cells Promotes Dendritic Cell Migration Required for Skin Immunity. Cell Rep 2021; 33:108258. [PMID: 33053342 PMCID: PMC7688291 DOI: 10.1016/j.celrep.2020.108258] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/06/2020] [Accepted: 09/21/2020] [Indexed: 12/26/2022] Open
Abstract
Although the function of the extracellular region of programmed death ligand 1 (PD-L1) through its interactions with PD-1 on T cells is well studied, little is understood regarding the intracellular domain of PD-L1. Here, we outline a major role for PD-L1 intracellular signaling in the control of dendritic cell (DC) migration from the skin to the draining lymph node (dLN). Using a mutant mouse model, we identify a TSS signaling motif within the intracellular domain of PD-L1. The TSS motif proves critical for chemokine-mediated DC migration to the dLN during inflammation. This loss of DC migration, in the PD-L1 TSS mutant, leads to a significant decline in T cell priming when DC trafficking is required for antigen delivery to the dLN. Finally, the TSS motif is required for chemokine receptor signaling downstream of the Gα subunit of the heterotrimeric G protein complex, ERK phosphorylation, and actin polymerization in DCs. Lucas et al. define three residues within the cytoplasmic tail of PD-L1 that are required for proper dendritic cell migration from the skin to the lymph node. These three-amino-acid residues promote chemokine signaling in dendritic cells and productive T cell responses to skin infections.
Collapse
Affiliation(s)
- Erin D Lucas
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Johnathon B Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Molecular Biology Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | | | - Madison Kraus
- Gates Summer Research Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Matthew A Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Molecular Biology Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA.
| |
Collapse
|
32
|
Reis-Sobreiro M, Teixeira da Mota A, Jardim C, Serre K. Bringing Macrophages to the Frontline against Cancer: Current Immunotherapies Targeting Macrophages. Cells 2021; 10:2364. [PMID: 34572013 PMCID: PMC8464913 DOI: 10.3390/cells10092364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/07/2021] [Accepted: 08/29/2021] [Indexed: 12/21/2022] Open
Abstract
Macrophages are found in all tissues and display outstanding functional diversity. From embryo to birth and throughout adult life, they play critical roles in development, homeostasis, tissue repair, immunity, and, importantly, in the control of cancer growth. In this review, we will briefly detail the multi-functional, protumoral, and antitumoral roles of macrophages in the tumor microenvironment. Our objective is to focus on the ever-growing therapeutic opportunities, with promising preclinical and clinical results developed in recent years, to modulate the contribution of macrophages in oncologic diseases. While the majority of cancer immunotherapies target T cells, we believe that macrophages have a promising therapeutic potential as tumoricidal effectors and in mobilizing their surroundings towards antitumor immunity to efficiently limit cancer progression.
Collapse
Affiliation(s)
| | | | | | - Karine Serre
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal; (M.R.-S.); (A.T.d.M.); (C.J.)
| |
Collapse
|
33
|
Moon J, Oh YM, Ha SJ. Perspectives on immune checkpoint ligands: expression, regulation, and clinical implications. BMB Rep 2021. [PMID: 34078531 PMCID: PMC8411045 DOI: 10.5483/bmbrep.2021.54.8.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the tumor microenvironment, immune checkpoint ligands (ICLs) must be expressed in order to trigger the inhibitory signal via immune checkpoint receptors (ICRs). Although ICL expression frequently occurs in a manner intrinsic to tumor cells, extrinsic factors derived from the tumor microenvironment can fine-tune ICL expression by tumor cells or prompt non-tumor cells, including immune cells. Considering the extensive interaction between T cells and other immune cells within the tumor microenvironment, ICL expression on immune cells can be as significant as that of ICLs on tumor cells in promoting anti-tumor immune responses. Here, we introduce various regulators known to induce or suppress ICL expression in either tumor cells or immune cells, and concise mechanisms relevant to their induction. Finally, we focus on the clinical significance of understanding the mechanisms of ICLs for an optimized immunotherapy for individual cancer patients.
Collapse
Affiliation(s)
- Jihyun Moon
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Yoo Min Oh
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
34
|
Shwe TH, Pothacharoen P, Phitak T, Wudtiwai B, Kongtawelert P. Atorvastatin Attenuates Programmed Death Ligand-1 (PD-L1) Induction in Human Hepatocellular Carcinoma Cells. Int J Mol Sci 2021; 22:ijms22168755. [PMID: 34445462 PMCID: PMC8395726 DOI: 10.3390/ijms22168755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 01/05/2023] Open
Abstract
Liver cancer is the sixth most common cancer worldwide with high morbidity and mortality. Programmed death ligand 1 (PD-L1) is a major ligand of programmed death 1 receptor (PD1), and PD1/PD-L1 checkpoint acts as a negative regulator of the immune system. Cancers evade the host’s immune defense via PD-L1 expression. This study aimed to investigate the effects of tumor-related cytokines, interferon gamma (IFNγ), and tumor necrosis factor alpha (TNFα) on PD-L1 expression in human hepatocellular carcinoma cells, HepG2. Furthermore, as atorvastatin, a cholesterol-lowering agent, is documented for its immunomodulatory properties, its effect on PD-L1 expression was investigated. In this study, through real-time RT-PCR, Western blot, and immunocytochemistry methods, PD-L1 expression in both mRNA and protein levels was found to be synergistically upregulated in HepG2 by a combination of IFNγ and TNFα, and STAT1 activation was mainly responsible for that synergistic effect. Next, atorvastatin can inhibit the induction of PD-L1 by either IFNγ alone or IFNγ/TNFα combination treatment in HepG2 cells. In conclusion, in HepG2 cells, expression of PD-L1 was augmented by cytokines in the tumor microenvironment, and the effect of atorvastatin on tumor immune response through inhibition of PD-L1 induction should be taken into consideration in cancer patients who have been prescribed atorvastatin.
Collapse
Affiliation(s)
- Thuzar Hla Shwe
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (T.H.S.); (P.P.); (T.P.); (B.W.)
- Department of Medical Research, Ministry of Health, Yangon P.O. Box 11191, Myanmar
| | - Peraphan Pothacharoen
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (T.H.S.); (P.P.); (T.P.); (B.W.)
| | - Thanyaluck Phitak
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (T.H.S.); (P.P.); (T.P.); (B.W.)
| | - Benjawan Wudtiwai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (T.H.S.); (P.P.); (T.P.); (B.W.)
| | - Prachya Kongtawelert
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (T.H.S.); (P.P.); (T.P.); (B.W.)
- Correspondence: ; Tel.: +66-53-94-5325 (ext. 217)
| |
Collapse
|
35
|
Benoot T, Piccioni E, De Ridder K, Goyvaerts C. TNFα and Immune Checkpoint Inhibition: Friend or Foe for Lung Cancer? Int J Mol Sci 2021; 22:ijms22168691. [PMID: 34445397 PMCID: PMC8395431 DOI: 10.3390/ijms22168691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor-alpha (TNFα) can bind two distinct receptors (TNFR1/2). The transmembrane form (tmTNFα) preferentially binds to TNFR2. Upon tmTNFα cleavage by the TNF-alpha-converting enzyme (TACE), its soluble (sTNFα) form is released with higher affinity for TNFR1. This assortment empowers TNFα with a plethora of opposing roles in the processes of tumor cell survival (and apoptosis) and anti-tumor immune stimulation (and suppression), in addition to angiogenesis and metastases. Its functions and biomarker potential to predict cancer progression and response to immunotherapy are reviewed here, with a focus on lung cancer. By mining existing sequencing data, we further demonstrate that the expression levels of TNF and TACE are significantly decreased in lung adenocarcinoma patients, while the TNFR1/TNFR2 balance are increased. We conclude that the biomarker potential of TNFα alone will most likely not provide conclusive findings, but that TACE could have a key role along with the delicate balance of sTNFα/tmTNFα as well as TNFR1/TNFR2, hence stressing the importance of more research into the potential of rationalized treatments that combine TNFα pathway modulators with immunotherapy for lung cancer patients.
Collapse
|
36
|
Xian D, Niu L, Zeng J, Wang L. LncRNA KCNQ1OT1 Secreted by Tumor Cell-Derived Exosomes Mediates Immune Escape in Colorectal Cancer by Regulating PD-L1 Ubiquitination via MiR-30a-5p/USP22. Front Cell Dev Biol 2021; 9:653808. [PMID: 34350172 PMCID: PMC8326752 DOI: 10.3389/fcell.2021.653808] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Background: This study tried to explore the mechanism of long non-coding RNA (lncRNA) KCNQ1OT1 in tumor immune escape. Methods: Gene Expression Omnibus (GEO) and microarray analysis were used to screen the differentially expressed lncRNA and microRNA (miRNA) in normal tissues and tumor tissues. Quantitative reverse transcription PCR (RT-qPCR) was used to quantify KCNQ1OT1, miR-30a-5p, ubiquitin-specific peptidase 22 (USP22), and programmed death-ligand 1 (PD-L1). The interactive relationship between KCNQ1OT1 and miR-30a-5p was verified using dual-luciferase reporter gene assay and ribonucleoprotein immunoprecipitation (RIP) assay. Cell Counting Kit (CCK)-8, clone formation, wound healing, and apoptosis are used to detect the occurrence of tumor cells after different treatments. Protein half-life and ubiquitination detection are used to study the influence of USP22 on PD-L1 ubiquitination. BALB/c mice and BALB/c nude mice are used to detect the effects of different treatments on tumor growth and immune escape in vivo. Results: The expression of lncRNA KCNQ1OT1 in tumor tissues and tumor cell-derived exosomes was significantly increased. The tumor-promoting effect of lncRNA KCNQ1OT1 was through the autocrine effect of tumor cell-derived exosomes, which mediates the miR-30a-5p/USP22 pathway to regulate the ubiquitination of PD-L1 and inhibits CD8+ T-cell response, thereby promoting colorectal cancer development. Conclusion: Tumor cell-derived exosomes' KCNQ1OT1 could regulate PD-L1 ubiquitination through miR-30a-5p/USP22 to promote colorectal cancer immune escape.
Collapse
Affiliation(s)
- Di Xian
- Department of Emergency Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Liangbo Niu
- Department of Emergency Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Jie Zeng
- Department of Emergency Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Lei Wang
- Department of Emergency Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
37
|
Pose E, Coll M, Martínez‐Sánchez C, Zeng Z, Surewaard BGJ, Català C, Velasco‐de Andrés M, Lozano JJ, Ariño S, Fuster D, Niñerola‐Bazán A, Graupera I, Muñoz É, Lozano F, Sancho‐Bru P, Kubes P, Ginès P. Programmed Death Ligand 1 Is Overexpressed in Liver Macrophages in Chronic Liver Diseases, and Its Blockade Improves the Antibacterial Activity Against Infections. Hepatology 2021; 74:296-311. [PMID: 33219516 PMCID: PMC8362175 DOI: 10.1002/hep.31644] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 10/12/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Bacterial infections are common and severe in cirrhosis, but their pathogenesis is poorly understood. Dysfunction of liver macrophages may play a role, but information about their function in cirrhosis is limited. Our aims were to investigate the specific profile and function of liver macrophages in cirrhosis and their contribution to infections. Macrophages from human cirrhotic livers were characterized phenotypically by transcriptome analysis and flow cytometry; function was assessed in vivo by single photon emission computerized tomography in patients with cirrhosis. Serum levels of specific proteins and expression in peripheral monocytes were determined by ELISA and flow cytometry. In vivo phagocytic activity of liver macrophages was measured by spinning disk intravital microscopy in a mouse model of chronic liver injury. APPROACH AND RESULTS Liver macrophages from patients with cirrhosis overexpressed proteins related to immune exhaustion, such as programmed death ligand 1 (PD-L1), macrophage receptor with collagenous structure (MARCO), and CD163. In vivo phagocytic activity of liver macrophages in patients with cirrhosis was markedly impaired. Monocytes from patients with cirrhosis showed overexpression of PD-L1 that paralleled disease severity, correlated with its serum levels, and was associated with increased risk of infections. Blockade of PD-L1 with anti-PD-L1 antibody caused a shift in macrophage phenotype toward a less immunosuppressive profile, restored liver macrophage in vivo phagocytic activity, and reduced bacterial dissemination. CONCLUSION Liver cirrhosis is characterized by a remarkable impairment of phagocytic function of macrophages associated with an immunosuppressive transcriptome profile. The programmed cell death receptor 1/PD-L1 axis plays a major role in the impaired activity of liver macrophages. PD-L1 blockade reverses the immune suppressive profile and increases antimicrobial activity of liver macrophages in cirrhosis.
Collapse
Affiliation(s)
- Elisa Pose
- Liver UnitHospital ClínicBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Mar Coll
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain
| | | | - Zhutian Zeng
- Snyder Institute for Chronic Diseases, University of CalgaryCalgaryABCanada
| | | | - Cristina Català
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | | | - Juan José Lozano
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain
| | - Sílvia Ariño
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - David Fuster
- Nuclear Medicine Department, Hospital ClínicUniversity of BarcelonaBarcelonaSpain,Centro Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER‐bbn)BarcelonaSpain
| | - Aida Niñerola‐Bazán
- Nuclear Medicine Department, Hospital ClínicUniversity of BarcelonaBarcelonaSpain,Centro Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER‐bbn)BarcelonaSpain
| | - Isabel Graupera
- Liver UnitHospital ClínicBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Érica Muñoz
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Francisco Lozano
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain,Immunology UnitHospital ClínicBarcelonaSpain,Biomedicine DepartmentUniversity of BarcelonaBarcelonaSpain
| | - Pau Sancho‐Bru
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Paul Kubes
- Snyder Institute for Chronic Diseases, University of CalgaryCalgaryABCanada
| | - Pere Ginès
- Liver UnitHospital ClínicBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain,Medicine Department, Faculty of MedicineUniversity of BarcelonaBarcelonaSpain
| |
Collapse
|
38
|
Cai H, Zhang Y, Wang J, Gu J. Defects in Macrophage Reprogramming in Cancer Therapy: The Negative Impact of PD-L1/PD-1. Front Immunol 2021; 12:690869. [PMID: 34248982 PMCID: PMC8260839 DOI: 10.3389/fimmu.2021.690869] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/02/2021] [Indexed: 12/23/2022] Open
Abstract
Classically activated M1 macrophages and alternatively activated M2 macrophages are two polarized subsets of macrophages at the extreme ends of a constructed continuum. In the field of cancer research, M2 macrophage reprogramming is defined as the repolarization of pro-tumoral M2 to anti-tumoral M1 macrophages. It is known that colony-stimulating factor 1 (CSF1)/CSF1 receptor (CSF1R) and CSF2/CSF2R signaling play important roles in macrophage polarization. Targeting CSF1/CSF1R for M2 macrophage reprogramming has been widely performed in clinical trials for cancer therapy. Other targets for M2 macrophage reprogramming include Toll-like receptor 7 (TLR7), TLR8, TLR9, CD40, histone deacetylase (HDAC), and PI3Kγ. Although macrophages are involved in innate and adaptive immune responses, M1 macrophages are less effective at phagocytosis and antigen presenting, which are required properties for the activation of T cells and eradication of cancer cells. Similar to T and dendritic cells, the “functionally exhausted” status might be attributed to the high expression of programmed death-ligand 1 (PD-L1) or programmed cell death protein 1 (PD-1). PD-L1 is expressed on both M1 and M2 macrophages. Macrophage reprogramming from M2 to M1 might increase the expression of PD-L1, which can be transcriptionally activated by STAT3. Macrophage reprogramming or PD-L1/PD-1 blockade alone is less effective in the treatment of most cancers. Since PD-L1/PD-1 blockade could make up for the defect in macrophage reprogramming, the combination of macrophage reprogramming and PD-L1/PD-1 blockade might be a novel treatment strategy for cancer therapy.
Collapse
Affiliation(s)
- Hao Cai
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichi Zhang
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinyang Gu
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Narayanapillai SC, Han YH, Song JM, Kebede ME, Upadhyaya P, Kassie F. Modulation of the PD-1/PD-L1 immune checkpoint axis during inflammation-associated lung tumorigenesis. Carcinogenesis 2021; 41:1518-1528. [PMID: 32602900 DOI: 10.1093/carcin/bgaa059] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 05/28/2020] [Indexed: 12/27/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a significant risk factor for lung cancer. One potential mechanism through which COPD contributes to lung cancer development could be through generation of an immunosuppressive microenvironment that allows tumor formation and progression. In this study, we compared the status of immune cells and immune checkpoint proteins in lung tumors induced by the tobacco smoke carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) or NNK + lipopolysaccharide (LPS), a model for COPD-associated lung tumors. Compared with NNK-induced lung tumors, NNK+LPS-induced lung tumors exhibited an immunosuppressive microenvironment characterized by higher relative abundances of PD-1+ tumor-associated macrophages, PD-L1+ tumor cells, PD-1+ CD4 and CD8 T lymphocytes and FOXP3+ CD4 and CD8 T lymphocytes. Also, these markers were more abundant in the tumor tissue than in the surrounding 'normal' lung tissue of NNK+LPS-induced lung tumors. PD-L1 expression in lung tumors was associated with IFNγ/STAT1/STAT3 signaling axis. In cell line models, PD-L1 expression was found to be significantly enhanced in phorbol-12-myristate 13-acetate activated THP-1 human monocytes (macrophages) treated with LPS or incubated in conditioned media (CM) generated by non-small cell lung cancer (NSCLC) cells. Similarly, when NSCLC cells were incubated in CM generated by activated THP-1 cells, PD-L1 expression was upregulated in EGFR- and ERK-dependent manner. Overall, our observations indicate that COPD-like chronic inflammation creates a favorable immunosuppressive microenvironment for tumor development and COPD-associated lung tumors might show a better response to immune checkpoint therapies.
Collapse
Affiliation(s)
| | - Yong Hwan Han
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jung Min Song
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | | | - Pramod Upadhyaya
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Fekadu Kassie
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| |
Collapse
|
40
|
Zeisbrich M, Chevalier N, Sehnert B, Rizzi M, Venhoff N, Thiel J, Voll RE. CMTM6-Deficient Monocytes in ANCA-Associated Vasculitis Fail to Present the Immune Checkpoint PD-L1. Front Immunol 2021; 12:673912. [PMID: 34108971 PMCID: PMC8183471 DOI: 10.3389/fimmu.2021.673912] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022] Open
Abstract
Objectives ANCA-associated vasculitides (AAV) affect small- and medium-sized blood vessels. In active disease, vessel wall infiltrates are mainly composed of monocytes and macrophages. Immune checkpoint molecules are crucial for the maintenance of self-tolerance and the prevention of autoimmune diseases. After checkpoint inhibitor therapy, the development of autoimmune vasculitis has been observed. However, defects of immune checkpoint molecules in AAV patients have not been identified yet. Methods Monocytes and monocyte-derived macrophages from AAV patients and healthy age-matched controls were tested for surface expression of immunoinhibitory checkpoint programmed cell death ligand-1 (PD-L1). Using in vitro co-culture approaches, the effect of monocyte PD-L1 expression on CD4+ T cell activation and proliferation was tested. Results Monocytes from AAV patients displayed lower PD-L1 expression and a defective PD-L1 presentation upon activation, an effect that was correlated with disease activity. Lower PD-L1 expression was due to increased lysosomal degradation of PD-L1 in AAV monocytes. We identified a reduced expression of CMTM6, a protein protecting PD-L1 from lysosomal breakdown, as the underlying molecular defect. PD-L1low AAV monocytes showed increased stimulatory capacity and induced T cell activation and proliferation. Inhibiting lysosomal function corrected this phenotype by increasing PD-L1, thus normalizing the pro-stimulatory behavior of AAV monocytes. Conclusions This study identifies a defect of the immunoinhibitory checkpoint PD-L1 in monocytes from patients with AAV. Low expression of CMTM6 results in enhanced lysosomal degradation of PD-L1, thus providing insufficient negative signaling to T cells. Correcting this defect by targeting lysosomal function may represent a novel strategy to treat AAV.
Collapse
Affiliation(s)
- Markus Zeisbrich
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Nina Chevalier
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Bettina Sehnert
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Marta Rizzi
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Nils Venhoff
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Jens Thiel
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| |
Collapse
|
41
|
DeRogatis JM, Viramontes KM, Neubert EN, Tinoco R. PSGL-1 Immune Checkpoint Inhibition for CD4 + T Cell Cancer Immunotherapy. Front Immunol 2021; 12:636238. [PMID: 33708224 PMCID: PMC7940186 DOI: 10.3389/fimmu.2021.636238] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 01/15/2023] Open
Abstract
Immune checkpoint inhibition targeting T cells has shown tremendous promise in the treatment of many cancer types and are now standard therapies for patients. While standard therapies have focused on PD-1 and CTLA-4 blockade, additional immune checkpoints have shown promise in promoting anti-tumor immunity. PSGL-1, primarily known for its role in cellular migration, has also been shown to function as a negative regulator of CD4+ T cells in numerous disease settings including cancer. PSGL-1 is highly expressed on T cells and can engage numerous ligands that impact signaling pathways, which may modulate CD4+ T cell differentiation and function. PSGL-1 engagement in the tumor microenvironment may promote CD4+ T cell exhaustion pathways that favor tumor growth. Here we highlight that blocking the PSGL-1 pathway on CD4+ T cells may represent a new cancer therapy approach to eradicate tumors.
Collapse
Affiliation(s)
| | | | | | - Roberto Tinoco
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
42
|
Lee KY, Wong HY, Zeng Q, Le Lin J, Cheng MS, Kuick CH, Chang KTE, Loh AHP, Schwarz H. Ectopic CD137 expression by rhabdomyosarcoma provides selection advantages but allows immunotherapeutic targeting. Oncoimmunology 2021; 10:1877459. [PMID: 33643694 PMCID: PMC7872024 DOI: 10.1080/2162402x.2021.1877459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is a heterogeneous soft tissue neoplasm most frequently found in children and adolescents. As the prognosis for recurrent and metastatic RMS remains poor, immunotherapies are hoped to improve quality of life and survival. CD137 is a member of tumor necrosis factor receptor family and a T cell costimulatory molecule which induces potent cellular immune responses that are able to eliminate malignant cells. Therefore, it was puzzling to find expression of CD137 on an RMS tissue microarray by multiplex staining. CD137 is not only expressed by infiltrating T cells but also by malignant RMS cells. Functional in vitro experiments demonstrate that CD137 on RMS cells is being transferred to adjacent antigen-presenting cells by trogocytosis, where it downregulates CD137 ligand, and thereby reduces T cell costimulation which results in reduced killing of RMS cells. The transfer of CD137 and the subsequent downregulation of CD137 ligand is a physiological negative feedback mechanism that is likely usurped by RMS, and may facilitate its escape from immune surveillance. In addition, CD137 signals into RMS cells and induces IL-6 and IL-8 secretion, which are linked to RMS metastasis and poor prognosis. However, the ectopic expression of CD137 on RMS cells is an Achilles’ heel that may be utilized for immunotherapy. Natural killer cells expressing an anti-CD137 chimeric antigen receptor specifically kill CD137-expressing RMS cells. Our study implicates ectopic CD137 expression as a pathogenesis mechanism in RMS, and it demonstrates that CD137 may be a novel target for immunotherapy of RMS.
Collapse
Affiliation(s)
- Kang Yi Lee
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Hiu Yi Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Qun Zeng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Jia Le Lin
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Man Si Cheng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | | | | | | | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
43
|
Mercogliano MF, Bruni S, Mauro F, Elizalde PV, Schillaci R. Harnessing Tumor Necrosis Factor Alpha to Achieve Effective Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13030564. [PMID: 33540543 PMCID: PMC7985780 DOI: 10.3390/cancers13030564] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/17/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor alpha (TNFα) is a pleiotropic cytokine known to have contradictory roles in oncoimmunology. Indeed, TNFα has a central role in the onset of the immune response, inducing both activation and the effector function of macrophages, dendritic cells, natural killer (NK) cells, and B and T lymphocytes. Within the tumor microenvironment, however, TNFα is one of the main mediators of cancer-related inflammation. It is involved in the recruitment and differentiation of immune suppressor cells, leading to evasion of tumor immune surveillance. These characteristics turn TNFα into an attractive target to overcome therapy resistance and tackle cancer. This review focuses on the diverse molecular mechanisms that place TNFα as a source of resistance to immunotherapy such as monoclonal antibodies against cancer cells or immune checkpoints and adoptive cell therapy. We also expose the benefits of TNFα blocking strategies in combination with immunotherapy to improve the antitumor effect and prevent or treat adverse immune-related effects.
Collapse
Affiliation(s)
- María Florencia Mercogliano
- Laboratorio de Biofisicoquímica de Proteínas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales-Consejo Nacional de Investigaciones Científicas y Técnicas (IQUIBICEN-CONICET), Buenos Aires 1428, Argentina;
| | - Sofía Bruni
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Florencia Mauro
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Patricia Virginia Elizalde
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Roxana Schillaci
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
- Correspondence: ; Tel.: +54-11-4783-2869; Fax: +54-11-4786-2564
| |
Collapse
|
44
|
Stromal Protein-Mediated Immune Regulation in Digestive Cancers. Cancers (Basel) 2021; 13:cancers13010146. [PMID: 33466303 PMCID: PMC7795083 DOI: 10.3390/cancers13010146] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Solid cancers are surrounded by a network of non-cancerous cells comprising different cell types, including fibroblasts, and acellular protein structures. This entire network is called the tumor microenvironment (TME) and it provides a physical barrier to the tumor shielding it from infiltrating immune cells, such as lymphocytes, or therapeutic agents. In addition, the TME has been shown to dampen efficient immune responses of infiltrated immune cells, which are key in eliminating cancer cells from the organism. In this review, we will discuss how TME proteins in particular are involved in this dampening effect, known as immunosuppression. We will focus on three different types of digestive cancers: pancreatic cancer, colorectal cancer, and gastric cancer. Moreover, we will discuss current therapeutic approaches using TME proteins as targets to reverse their immunosuppressive effects. Abstract The stromal tumor microenvironment (TME) consists of immune cells, vascular and neural structures, cancer-associated fibroblasts (CAFs), as well as extracellular matrix (ECM), and favors immune escape mechanisms promoting the initiation and progression of digestive cancers. Numerous ECM proteins released by stromal and tumor cells are crucial in providing physical rigidity to the TME, though they are also key regulators of the immune response against cancer cells by interacting directly with immune cells or engaging with immune regulatory molecules. Here, we discuss current knowledge of stromal proteins in digestive cancers including pancreatic cancer, colorectal cancer, and gastric cancer, focusing on their functions in inhibiting tumor immunity and enabling drug resistance. Moreover, we will discuss the implication of stromal proteins as therapeutic targets to unleash efficient immunotherapy-based treatments.
Collapse
|
45
|
Eddy K, Chen S. Overcoming Immune Evasion in Melanoma. Int J Mol Sci 2020; 21:E8984. [PMID: 33256089 PMCID: PMC7730443 DOI: 10.3390/ijms21238984] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/17/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
Melanoma is the most aggressive and dangerous form of skin cancer that develops from transformed melanocytes. It is crucial to identify melanoma at its early stages, in situ, as it is "curable" at this stage. However, after metastasis, it is difficult to treat and the five-year survival is only 25%. In recent years, a better understanding of the etiology of melanoma and its progression has made it possible for the development of targeted therapeutics, such as vemurafenib and immunotherapies, to treat advanced melanomas. In this review, we focus on the molecular mechanisms that mediate melanoma development and progression, with a special focus on the immune evasion strategies utilized by melanomas, to evade host immune surveillances. The proposed mechanism of action and the roles of immunotherapeutic agents, ipilimumab, nivolumab, pembrolizumab, and atezolizumab, adoptive T- cell therapy plus T-VEC in the treatment of advanced melanoma are discussed. In this review, we implore that a better understanding of the steps that mediate melanoma onset and progression, immune evasion strategies exploited by these tumor cells, and the identification of biomarkers to predict treatment response are critical in the design of improved strategies to improve clinical outcomes for patients with this deadly disease.
Collapse
Affiliation(s)
- Kevinn Eddy
- Graduate Program in Cellular and Molecular Pharmacology, School of Graduate Studies Rutgers University, Piscataway, NJ 08854, USA;
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA
| | - Suzie Chen
- Graduate Program in Cellular and Molecular Pharmacology, School of Graduate Studies Rutgers University, Piscataway, NJ 08854, USA;
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Environmental & Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
46
|
Abstract
In this issue of JEM, Du et al. (https://doi.org/10.1084/jem.20191115) report that enhancement of the β-catenin signaling by Wnt or EGF treatment increases the expression of PD-L1 in an AKT and β-catenin-dependent manner, and blocking the AKT pathway synergizes with anti-PD-1 in a glioblastoma model.
Collapse
Affiliation(s)
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
47
|
Lin D, Liu Q, Wang W, Li Y, Li Y, Lin B, Ye Z, Huang J, Yu X, Chen Y, Mei Y, Huang M, Yang W, Zhou J, Liu X, Zeng J. Aberrant expression of miR-16, B12 and CD272 in peripheral blood mononuclear cells from patients with active tuberculosis. Am J Transl Res 2020; 12:6076-6091. [PMID: 33194015 PMCID: PMC7653578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
Tuberculosis (TB) immunity is affected by complex immune regulation processes, which involve various immune cells, immune molecules, and cytokines. Here, we evaluated the expression of B12, CD272 and miR-16 in peripheral blood mononuclear cells (PBMC) of patients with active pulmonary tuberculosis. The results showed that monocytes expressing CD272 or B12 were down-regulated in patients with tuberculosis. The expression of B12 and CD272 in T cells and monocytes is related to tuberculosis. In TB patients, the up-regulation of miR-16 was negatively correlated with B12 mRNA expression, miR-16 was mainly expressed in CD14+ monocytes, and CD272 mRNA was mainly expressed in CD19+ B cells. It is worth noting that the overexpression of miR-16 inhibits the expression of CD272 and B12 in monocytes of TB patients. After BCG stimulation, miR-16 expression of CD14+ monocytes was up-regulated and B12 mRNA and CD272 mRNA expressions were down-regulated in TB patients. Finally, we found that miR-16 may participate in the TB immunization process through targeted regulation of B12 expression. These studies indicate that the expression of B12, CD272 and miR-16 in PBMC may be related to tuberculosis.
Collapse
Affiliation(s)
- Dongzi Lin
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
- Department of Laboratory Medicine, Foshan Forth People’s HospitalFoshan 528041, Guangdong, China
| | - Qiankun Liu
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Wei Wang
- Department of Laboratory Medicine, Foshan Forth People’s HospitalFoshan 528041, Guangdong, China
| | - Yanyun Li
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Yumei Li
- Department of Laboratory Medicine, Dongguan Sixth People’s HospitalDongguan 523008, Guangdong, China
| | - Bihua Lin
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Ziyu Ye
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Juan Huang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Xiaolin Yu
- Department of Laboratory Medicine, Dongguan Sixth People’s HospitalDongguan 523008, Guangdong, China
| | - Yinwen Chen
- Department of Laboratory Medicine, Dongguan Sixth People’s HospitalDongguan 523008, Guangdong, China
| | - Yuezhi Mei
- Department of Laboratory Medicine, Dongguan Sixth People’s HospitalDongguan 523008, Guangdong, China
| | - Minyuan Huang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Weiqin Yang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Jie Zhou
- Department of Laboratory Medicine, Foshan Forth People’s HospitalFoshan 528041, Guangdong, China
| | - Xinguang Liu
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Jincheng Zeng
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| |
Collapse
|
48
|
Spatial and Temporal Changes in PD-L1 Expression in Cancer: The Role of Genetic Drivers, Tumor Microenvironment and Resistance to Therapy. Int J Mol Sci 2020; 21:ijms21197139. [PMID: 32992658 PMCID: PMC7583014 DOI: 10.3390/ijms21197139] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapies blocking immune inhibitory receptors programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) on T-cells have dramatically improved patient outcomes in a range of advanced cancers. However, the lack of response, and the development of resistance remain major obstacles to long-term improvements in patient outcomes. There is significant interest in the clinical use of biomarkers to improve patient selection, and the expression of PD-1 ligand 1 (PD-L1) is often reported as a potential biomarker of response. However, accumulating evidence suggests that the predictive value of PD-L1 expression in tumor biopsies is relatively low due, in part, to its complex biology. In this review, we discuss the biological consequences of PD-L1 expression by various cell types within the tumor microenvironment, and the complex mechanisms that regulate PD-L1 expression at the genomic, transcriptomic and proteomic levels.
Collapse
|
49
|
Hossain MA, Liu G, Dai B, Si Y, Yang Q, Wazir J, Birnbaumer L, Yang Y. Reinvigorating exhausted CD8 + cytotoxic T lymphocytes in the tumor microenvironment and current strategies in cancer immunotherapy. Med Res Rev 2020; 41:156-201. [PMID: 32844499 DOI: 10.1002/med.21727] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/26/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy has revolutionized the treatment of cancer in recent years and achieved overall success and long-term clinical benefit in patients with a wide variety of cancer types. However, there is still a large proportion of patients exhibiting limited or no responses to immunotherapeutic strategy, some of which were even observed with hyperprogressive disease. One major obstacle restricting the efficacy is that tumor-reactive CD8+ T cells, which are central for tumor control, undergo exhaustion, and lose their ability to eliminate cancer cells after infiltrating into the strongly immunosuppressive tumor microenvironment. Thus, as a potential therapeutic rationale in the development of cancer immunotherapy, targeting or reinvigorating exhausted CD8+ T cells has been attracting much interest. Hitherto, both intrinsic and extrinsic mechanisms that govern CD8+ T-cell exhaustion have been explored. Specifically, the transcriptional and epigenetic landscapes have been depicted utilizing single-cell RNA sequencing or mass cytometry (CyTOF). In addition, cellular metabolism dictating the tumor-infiltrating CD8+ T-cell fate is currently under investigation. A series of clinical trials are being carried out to further establish the current strategies targeting CD8+ T-cell exhaustion. Taken together, despite the proven benefit of immunotherapy in cancer patients, additional efforts are still needed to fully circumvent limitations of exhausted T cells in the treatment. In this review, we will focus on the current cellular and molecular understanding of metabolic changes, epigenetic remodeling, and transcriptional regulation in CD8+ T-cell exhaustion and describe hypothetical treatment approaches based on immunotherapy aiming at reinvigorating exhausted CD8+ T cells.
Collapse
Affiliation(s)
- Md Amir Hossain
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guilai Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Beiying Dai
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yaxuan Si
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qitao Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Junaid Wazir
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Durham, North Carolina, USA.,Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina
| | - Yong Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.,Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
50
|
Shu C, Li Q. Current advances in PD-1/PD-L1 axis-related tumour-infiltrating immune cells and therapeutic regimens in glioblastoma. Crit Rev Oncol Hematol 2020; 151:102965. [PMID: 32442903 DOI: 10.1016/j.critrevonc.2020.102965] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/21/2020] [Accepted: 04/15/2020] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant tumour in the brain, and current treatments are not curative and cannot control recurrence. This limitation indirectly places immunotherapy at the focus of translational GBM research. Many studies on the PD-1/PD-L1 axis in GBM are ongoing, and the immunosuppressive mechanism of PD-1/PD-L1 in GBM is different from that in other solid tumours. This review focuses on the effect of the PD-1/PD-L1 axis on infiltrating immune cells in the suppressive GBM immune microenvironment and summarizes the recent progress in PD-1/PD-L1 axis-related therapies reported in preclinical and clinical GBM studies, providing a reference for the systematic study of PD-1/PD-L1 axis-related anti-GBM immunity.
Collapse
Affiliation(s)
- Chang Shu
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Tianjin Neurosurgery Institute, Tianjin Huan Hu Hospital, Tianjin, 300350, China; Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Qingguo Li
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| |
Collapse
|