1
|
Park SY, Pylaeva E, Bhuria V, Gambardella AR, Schiavoni G, Mougiakakos D, Kim SH, Jablonska J. Harnessing myeloid cells in cancer. Mol Cancer 2025; 24:69. [PMID: 40050933 PMCID: PMC11887392 DOI: 10.1186/s12943-025-02249-2] [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: 12/19/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
Cancer-associated myeloid cells due to their plasticity play dual roles in both promoting and inhibiting tumor progression. Myeloid cells with immunosuppressive properties play a critical role in anti-cancer immune regulation. Cells of different origin, such as tumor associated macrophages (TAMs), tumor associated neutrophils (TANs), myeloid derived suppressor cells (also called MDSCs) and eosinophils are often expanded in cancer patients and significantly influence their survival, but also the outcome of anti-cancer therapies. For this reason, the variety of preclinical and clinical studies to modulate the activity of these cells have been conducted, however without successful outcome to date. In this review, pro-tumor activity of myeloid cells, myeloid cell-specific therapeutic targets, in vivo studies on myeloid cell re-polarization and the impact of myeloid cells on immunotherapies/genetic engineering are addressed. This paper also summarizes ongoing clinical trials and the concept of chimeric antigen receptor macrophage (CAR-M) therapies, and suggests future research perspectives, offering new opportunities in the development of novel clinical treatment strategies.
Collapse
Affiliation(s)
- Su-Yeon Park
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ekaterina Pylaeva
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, Essen, 45147, Germany
- German Cancer Consortium (DKTK) Partner Site Düsseldorf/Essen, Essen, Germany
| | - Vikas Bhuria
- Department of Hematology, Oncology, and Cell Therapy, Otto-Von-Guericke University, Magdeburg, Germany
| | | | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto-Von-Guericke University, Magdeburg, Germany
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jadwiga Jablonska
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, Essen, 45147, Germany.
- German Cancer Consortium (DKTK) Partner Site Düsseldorf/Essen, Essen, Germany.
| |
Collapse
|
2
|
Matsuzaki H, Pan C, Komohara Y, Yamada R, Yano H, Fujiwara Y, Kai K, Mukasa A. The roles of glioma-associated macrophages/microglia and potential targets for anti-glioma therapy. Immunol Med 2025; 48:24-32. [PMID: 39391957 DOI: 10.1080/25785826.2024.2411035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
Glioblastoma (GBM) is the central nervous system tumor with the most aggressive behavior, and no definitive therapy has yet been found. The tumor microenvironment of GBM is immunosuppressive and is considered a 'cold tumor' with low lymphocytic infiltration, but is characterized by a high proportion of glioma-associated macrophages/microglia (GAMs). GAMs promote tumor growth and also affect treatment resistance in GBM. In this review, we describe the origin and classification of GAMs in humans and describe the mechanisms of their activation and the cell-cell interactions between tumor cells and GAMs. We also describe the history of GAM detection methods, especially immunohistochemistry, and discusses the merits and limitations of these techniques. In addition, we summarized chemotactic factors for GAMs and the therapies targeting these factors. Recent single-cell RNA analysis and spatial analysis add new insights to our previous knowledge of GAMs. Based on these studies, GBM therapies targeting GAMs are expected to be further developed.
Collapse
Affiliation(s)
- Hiroaki Matsuzaki
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Cheng Pan
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Rin Yamada
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Diagnostic Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiromu Yano
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keitaro Kai
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Akitake Mukasa
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
3
|
Zhang C, Yuan Y, Xia Q, Wang J, Xu K, Gong Z, Lou J, Li G, Wang L, Zhou L, Liu Z, Luo K, Zhou X. Machine Learning-Driven Prediction, Preparation, and Evaluation of Functional Nanomedicines Via Drug-Drug Self-Assembly. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415902. [PMID: 39792782 PMCID: PMC11884566 DOI: 10.1002/advs.202415902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Indexed: 01/12/2025]
Abstract
Small molecules as nanomedicine carriers offer advantages in drug loading and preparation. Selecting effective small molecules for stable nanomedicines is challenging. This study used artificial intelligence (AI) to screen drug combinations for self-assembling nanomedicines, employing physiochemical parameters to predict formation via machine learning. Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) are identified as effective carriers for antineoplastic drugs, with high drug loading. Nanomedicines, PEG-coated indomethacin/paclitaxel nanomedicine (PiPTX), and laminarin-modified indomethacin/paclitaxel nanomedicine (LiDOX), are developed with extended circulation and active targeting functions. Indomethacin/paclitaxel nanomedicine iDOX exhibits pH-responsive drug release in the tumor microenvironment. These nanomedicines enhance anti-tumor effects and reduce side effects, offering a rapid approach to clinical nanomedicine development.
Collapse
Affiliation(s)
- Chengyuan Zhang
- Yunnan Key Laboratory of Stem Cell and Regenerative MedicineKunming Medical UniversityKunming650500China
| | - Yuchuan Yuan
- School of MedicineNorthwest UniversityXi'an710068China
| | - Qiong Xia
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Junjie Wang
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Kangkang Xu
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Zhiwei Gong
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Jie Lou
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Gen Li
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Lu Wang
- Department of PharmacySchool of Pharmacy and BioengineeringChongqing University of TechnologyChongqing400054China
| | - Li Zhou
- Department of Biomedical EngineeringSchool of EngineeringChina Pharmaceutical UniversityNanjing210009China
| | - Zhirui Liu
- Department of PharmacyXinan HospitalArmy Medical UniversityChongqing400038China
| | - Kui Luo
- Department of Radiologyand Department of GeriatricsHuaxi MR Research Center (HMRRC)National Clinical Research Center for Geriatrics Frontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityNo. 37 Guoxue AlleyChengdu610041China
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative MedicineKunming Medical UniversityKunming650500China
| |
Collapse
|
4
|
Toghraie FS, Bayat M, Hosseini MS, Ramezani A. Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01051-y. [PMID: 39998754 DOI: 10.1007/s13402-025-01051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.
Collapse
Affiliation(s)
- Fatemeh Sadat Toghraie
- Institute of Biotechnology, Faculty of the Environment and Natural Sciences, Brandenburg University of Technology, Cottbus, Germany
| | - Maryam Bayat
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sadat Hosseini
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Abdalsalam NMF, Ibrahim A, Saliu MA, Liu TM, Wan X, Yan D. MDSC: a new potential breakthrough in CAR-T therapy for solid tumors. Cell Commun Signal 2024; 22:612. [PMID: 39702149 DOI: 10.1186/s12964-024-01995-y] [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: 09/23/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has shown remarkable success in hematologic malignancies but has encountered challenges in effectively treating solid tumors. One major obstacle is the presence of the immunosuppressive tumor microenvironment (TME), which is mainly built by myeloid-derived suppressor cells (MDSCs). Recent studies have shown that MDSCs have a detrimental effect on CAR-T cells due to their potent immunosuppressive capabilities. Targeting MDSCs has shown promising results to enhance CAR-T immunotherapy in preclinical solid tumor models. In this review, we first highlight that MDSCs increase tumor proliferation, transition, angiogenesis and encourage circulating tumor cells (CTCs) extravasation leading to tumor progression and metastasis. Moreover, we describe the main characteristics of the immunosuppressive activities of MDSCs on T cells in TME. Most importantly, we summarize targeting therapeutic strategies of MDSCs in CAR-T therapies against solid tumors. These strategies include (1) therapeutic targeting of MDSCs through small molecule inhibitors and large molecule antibodies; (2) CAR-T targeting cancer cell antigen combination with MDSC modulatory agents; (3) cytokine receptor antigen-targeted CAR-T indirectly or directly targeting MDSCs reshapes TME; (4) modified natural killer (NK) cells expressing activating receptor directly targeting MDSCs; and (5) CAR-T directly targeting MDSC selective antigens. In the near future, we are expected to witness the improvement of CAR-T cell therapies for solid tumors by targeting MDSCs in clinical practice.
Collapse
Affiliation(s)
- Nada Mohamady Farouk Abdalsalam
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Abdulrahman Ibrahim
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Muhammad Auwal Saliu
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, Taipa, China.
| | - Xiaochun Wan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100864, China.
| | - Dehong Yan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100864, China.
| |
Collapse
|
6
|
Montoya M, Collins SA, Chuntova P, Patel TS, Nejo T, Yamamichi A, Kasahara N, Okada H. Interferon regulatory factor 8-driven reprogramming of the immune microenvironment enhances antitumor adaptive immunity and reduces immunosuppression in murine glioblastoma. Neuro Oncol 2024; 26:2272-2287. [PMID: 39115195 PMCID: PMC11630541 DOI: 10.1093/neuonc/noae149] [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] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) has a highly immunosuppressive tumor immune microenvironment (TIME), largely mediated by myeloid-derived suppressor cells (MDSCs). Here, we utilized a retroviral replicating vector (RRV) to deliver Interferon Regulatory Factor 8 (IRF8), a master regulator of type 1 conventional dendritic cell (cDC1) development, in a syngeneic murine GBM model. We hypothesized that RRV-mediated delivery of IRF8 could "reprogram" intratumoral MDSCs into antigen-presenting cells and thereby restore T-cell responses. METHODS Effects of RRV-IRF8 on survival and tumor growth kinetics were examined in the SB28 murine GBM model. The immunophenotype was analyzed by flow cytometry and gene expression assays. We assayed functional immunosuppression and antigen presentation by ex vivo T-cell-myeloid co-culture. RESULTS Intratumoral injection of RRV-IRF8 in mice bearing intracerebral SB28 glioma significantly suppressed tumor growth and prolonged survival. RRV-IRF8 treated tumors exhibited significant enrichment of cDC1s and CD8+ T-cells. Additionally, myeloid cells derived from RRV-IRF8 tumors showed decreased expression of the immunosuppressive markers Arg1 and IDO1 and demonstrated reduced suppression of naïve T-cell proliferation in ex vivo co-culture, compared to controls. Furthermore, DCs from RRV-IRF8 tumors showed increased antigen presentation compared to those from control tumors. In vivo treatment with azidothymidine (AZT), a viral replication inhibitor, showed that IRF8 transduction in both tumor and non-tumor cells is necessary for survival benefit, associated with a reprogrammed, cDC1- and CD8 T-cell-enriched TIME. CONCLUSIONS Our results indicate that reprogramming of glioma-infiltrating myeloid cells by in vivo expression of IRF8 may reduce immunosuppression and enhance antigen presentation, achieving improved tumor control.
Collapse
Affiliation(s)
- Megan Montoya
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Sara A Collins
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Pavlina Chuntova
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Trishna S Patel
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Takahide Nejo
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Akane Yamamichi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Noriyuki Kasahara
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
- The Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| |
Collapse
|
7
|
Taheri M, Tehrani HA, Farzad SA, Korourian A, Arefian E, Ramezani M. The potential of mesenchymal stem cell coexpressing cytosine deaminase and secretory IL18-FC chimeric cytokine in suppressing glioblastoma recurrence. Int Immunopharmacol 2024; 142:113048. [PMID: 39236459 DOI: 10.1016/j.intimp.2024.113048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Glioblastoma multiforme (GBM) patients have a high recurrence rate of 90%, and the 5-year survival rate is only about 5%. Cytosine deaminase (CDA)/5-fluorocytosine (5-FC) gene therapy is a promising glioma treatment as 5-FC can cross the blood-brain barrier (BBB), while 5-fluorouracil (5-FU) cannot. Furthermore, 5-FU can assist reversing the immunological status of cold solid tumors. This study developed mesenchymal stem cells (MSCs) co-expressing yeast CDA and the secretory IL18-FC superkine to prevent recurrent tumor progression by simultaneously exerting cytotoxic effects and enhancing immune responses. IL18 was fused with Igk and IgG2a FC domains to enhance its secretion and serum half-life. The study confirmed the expression and activity of the CDA enzyme, as well as the expression, secretion, and activity of secretory IL18 and IL18-FC superkine, which were expressed by lentiviruses transduced-MSCs. In the transwell tumor-tropism assay, it was observed that the genetically modified MSCs retained their selective tumor-tropism ability following transduction. CDA-expressing MSCs, in the presence of 5-FC (200 µg/ml), induced cell cycle arrest and apoptosis in glioma cells through bystander effects in an indirect transwell co-culture system. They reduced the viability of the direct co-culture system when they constituted only 12.5 % of the cell population. The effectiveness of engineered MSCs in suppressing tumor progression was assessed by intracerebral administration of a lethal dose of GL261 cells combined in a ratio of 1:1 with MSCs expressing CDA, or CDA and sIL18, or CDA and sIL18-FC, into C57BL/6 mice. PET scan showed no conspicuous tumor mass in the MSC-CDA-sIL18-FC group that received 5-FC treatment. The pathological analysis showed that tumor progression suppressed in this group until 20th day after cell inoculation. Cytokine assessment showed that both interferon-gamma (IFN-γ) and interleukin-4 (IL-4) increased in the serum of MSC-CDA-sIL18 and MSC-CDA-sIL18-FC, treated with normal saline (NS) compared to those of the control group. The MSC-CDA-sIL18-FC group that received 5-FC treatment showed reduced serum levels of IL-6 and a considerably improved survival rate compared to the control group. Therefore, MSCs co-expressing yeast CDA and secretory IL18-FC, with tumor tropism capability, may serve as a supplementary approach to standard GBM treatment to effectively inhibit tumor progression and prevent recurrence.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sara Amel Farzad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Korourian
- Quality Control Department Pathobiology Laboratory Center, Tehran, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Stem Cells Technology and Tissue Regeneration Department, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Rajkumari S, Singh J, Agrawal U, Agrawal S. Myeloid-derived suppressor cells in cancer: Current knowledge and future perspectives. Int Immunopharmacol 2024; 142:112949. [PMID: 39236460 DOI: 10.1016/j.intimp.2024.112949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
MDSCs (myeloid-derived suppressor cells) are crucial for immune system evasion in cancer. They accumulate in peripheral blood and tumor microenvironment, suppressing immune cells like T-cells, natural killer cells and dendritic cells. They promote tumor angiogenesis and metastasis by secreting cytokines and growth factors and contribute to a tumor-promoting environment. The accumulation of MDSCs in cancer patients has been linked to poor prognosis and resistance to various cancer therapies. Targeting MDSCs and their immunosuppressive mechanisms may improve treatment outcomes and enhance immune surveillance by developing drugs that inhibit MDSC function, by preventing their accumulation and by disrupting the tumor-promoting environment. This review presents a detailed overview of the MDSC research in cancer with regulation of their development and function. The relevance of MDSC as a prognostic and predictive biomarker in different types of cancers, along with recent advancements on the therapeutic approaches to target MDSCs are discussed in detail.
Collapse
Affiliation(s)
- Sunanda Rajkumari
- ICMR National Institute of Medical Statistics, Ansari Nagar, New Delhi 110029, India
| | - Jaspreet Singh
- ICMR National Institute of Pathology, Safdarjung Hospital Campus, Ansari Nagar, New Delhi 110029, India
| | - Usha Agrawal
- Asian Institute of Public Health University (AIPH) University, 1001 Haridamada, Jatani, Near IIT Bhubaneswar, Bhubaneswar 751002, India
| | - Sandeep Agrawal
- Discovery Research Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India.
| |
Collapse
|
9
|
Cuenca-Escalona J, Bödder J, Subtil B, Sánchez-Sánchez M, Vidal-Manrique M, Sweep MWD, Fauerbach JA, Cambi A, Flórez-Grau G, de Vries JM. EP2/EP4 targeting prevents tumor-derived PGE2-mediated immunosuppression in cDC2s. J Leukoc Biol 2024; 116:1554-1567. [PMID: 39041661 DOI: 10.1093/jleuko/qiae164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/11/2024] [Accepted: 07/22/2024] [Indexed: 07/24/2024] Open
Abstract
Tumor-derived prostaglandin E2 (PGE2) impairs antitumor immunity by priming suppressive functions on various immune cell types, including dendritic cells (DCs). In this way, tumors mediate DC dysfunction and hamper their antitumoral activity. PGE2 is known to modulate DC function via signaling through the E-type prostanoid receptor 2 (EP2) and EP4. Preclinical studies have demonstrated the therapeutic value of targeting EP2/4 receptor signaling in DCs. Ongoing phase 1 clinical trials with EP antagonists have shown immunomodulation in cancer patients. However, the systemic drug administration leads to off-target events and subsequent side effects. To limit the off-target effects of EP targeting, EP2 and EP4 antagonists were encapsulated in polymeric nanoparticles (NPs). In this study, we evaluated the efficacy of EP2/4-specific antagonists encapsulated in NPs to protect conventional type 2 DCs (cDC2s) from suppressive effects of tumor-derived PGE2 in different tumor models. We show that tumor-derived PGE2 signals via EP2/4 to mediate the acquisition of a suppressive phenotype of cDC2s. EP2/4 antagonists encapsulated in NPs impaired the conversion of cDC2s toward a suppressive state and inhibited the occurrence of suppressive features such as interleukin-10 production or the ability to expand regulatory T cells. Importantly, the NPs abolished the transition toward this suppressive state in different tumor models: melanoma-conditioned media, ascites fluid derived from ovarian cancer patients (2-dimensional), and upon coculture with colorectal cancer patient-derived organoids (3-dimensional). We propose that targeting the PGE2-EP2/4 axis using NPs can achieve immunomodulation in the immune system of cancer patients, alleviate tumor-derived suppression, and thus facilitate the development of potent antitumor immunity in cancer patients.
Collapse
MESH Headings
- Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Dinoprostone/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/drug effects
- Animals
- Mice
- Cell Line, Tumor
- Female
- Humans
- Mice, Inbred C57BL
- Immune Tolerance/drug effects
- Nanoparticles/chemistry
Collapse
Affiliation(s)
- Jorge Cuenca-Escalona
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA, Nijmegen, the Netherlands
| | - Johanna Bödder
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA, Nijmegen, the Netherlands
| | - Beatriz Subtil
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA, Nijmegen, the Netherlands
| | - Marta Sánchez-Sánchez
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA, Nijmegen, the Netherlands
| | - Marcos Vidal-Manrique
- Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Mark W D Sweep
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA, Nijmegen, the Netherlands
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Jonathan A Fauerbach
- R&D Reagents, Chemical Biology Department; Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429, Bergisch Gladbach, Germany
| | - Alessandra Cambi
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA, Nijmegen, the Netherlands
| | - Georgina Flórez-Grau
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA, Nijmegen, the Netherlands
| | - Jolanda M de Vries
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA, Nijmegen, the Netherlands
| |
Collapse
|
10
|
Corsaro A, Tremonti B, Bajetto A, Barbieri F, Thellung S, Florio T. Chemokine signaling in tumors: potential role of CXC chemokines and their receptors as glioblastoma therapeutic targets. Expert Opin Ther Targets 2024; 28:937-952. [PMID: 39582130 DOI: 10.1080/14728222.2024.2433130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
INTRODUCTION Glioblastoma is the most aggressive brain tumor, typically associated with poor prognosis. Its treatment is challenging due to the peculiar glioblastoma cell biology and its microenvironment complexity. Specifically, a small fraction of glioma stem cells within the tumor mass drives tumor growth and invasiveness by hijacking brain resident and immune cells. This process also involves modification of extracellular matrix components, such as collagen and glycoproteins, where the secretion of soluble mediators, particularly CXC chemokines, plays a significant role. AREAS COVERED We analyze the critical role of chemokines in glioblastoma tumorigenesis, proliferation, angiogenesis, tumor progression, and brain parenchyma invasiveness. Recent evidence highlights how chemokines and their receptors impact glioblastoma biology and represent potential therapeutic targets. Several studies show that chemokines modulate glioblastoma development by acting on glioma stem cell proliferation and self-renewal, promoting vasculogenic mimicry, and altering the extracellular matrix to facilitate tumor invasiveness. EXPERT OPINION There is clear evidence supporting CXC receptors (such as CXCR1, 2, 3, 4, and ACKR3/CXCR7) and their signaling pathways as promising pharmacological targets. This in-depth review of chemokine roles in glioblastoma development provides a critical evaluation of the possible clinical translation of innovative compounds targeting these ligand/receptor systems, leading to improved therapeutic outcomes for glioblastoma patients.
Collapse
Affiliation(s)
- Alessandro Corsaro
- Sezione di Farmacologia, Dipartimento di Medicina Interna, Università di Genova, Genova, Italy
| | - Beatrice Tremonti
- Sezione di Farmacologia, Dipartimento di Medicina Interna, Università di Genova, Genova, Italy
| | - Adriana Bajetto
- Sezione di Farmacologia, Dipartimento di Medicina Interna, Università di Genova, Genova, Italy
| | - Federica Barbieri
- Sezione di Farmacologia, Dipartimento di Medicina Interna, Università di Genova, Genova, Italy
- IRCCS Policlinico San Martino, Genova, Italy
| | - Stefano Thellung
- Sezione di Farmacologia, Dipartimento di Medicina Interna, Università di Genova, Genova, Italy
- IRCCS Policlinico San Martino, Genova, Italy
| | - Tullio Florio
- Sezione di Farmacologia, Dipartimento di Medicina Interna, Università di Genova, Genova, Italy
- IRCCS Policlinico San Martino, Genova, Italy
| |
Collapse
|
11
|
Klein C, Mebroukine S, Madéry M, Moisand A, Boyer T, Larmonier N, Robert G, Domblides C. Myeloid-Derived Suppressor Cells in Bladder Cancer: An Emerging Target. Cells 2024; 13:1779. [PMID: 39513886 PMCID: PMC11544784 DOI: 10.3390/cells13211779] [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: 10/03/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Bladder cancer remains a prevalent and challenging malignancy. Myeloid-derived suppressor cells (MDSCs) have emerged as key contributors to the immunosuppressive tumor microenvironment, facilitating tumor progression, immune evasion, and resistance to therapies. This review explores the role of MDSC in bladder cancer, highlighting their involvement in immune regulation; tumor progression; and resistance to therapies such as bacillus Calmette-Guérin (BCG) therapy, chemotherapy, and immune checkpoint inhibitors (ICIs). We also discuss their potential as biomarkers and therapeutic targets, with current evidence suggesting that targeting MDSCs, either alone or in combination with existing treatments such as BCG and ICIs, may enhance anti-tumor immunity and improve clinical outcomes. However,, challenges remain, particularly regarding the identification and therapeutic modulation of MDSC subpopulations. Further research is warranted to fully elucidate their role in bladder cancer and to optimize MDSC-targeted therapies.
Collapse
Affiliation(s)
- Clément Klein
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; (C.K.); (S.M.); (M.M.); (A.M.); (T.B.); (N.L.); (G.R.)
- Department of Urology, University Hospital of Bordeaux, 33000 Bordeaux, France
- Department of Biological and Medical Sciences, University of Bordeaux, 33000 Bordeaux, France
| | - Samy Mebroukine
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; (C.K.); (S.M.); (M.M.); (A.M.); (T.B.); (N.L.); (G.R.)
- Department of Urology, University Hospital of Bordeaux, 33000 Bordeaux, France
- Department of Biological and Medical Sciences, University of Bordeaux, 33000 Bordeaux, France
| | - Mathilde Madéry
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; (C.K.); (S.M.); (M.M.); (A.M.); (T.B.); (N.L.); (G.R.)
- Department of Biological and Medical Sciences, University of Bordeaux, 33000 Bordeaux, France
| | - Alexandra Moisand
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; (C.K.); (S.M.); (M.M.); (A.M.); (T.B.); (N.L.); (G.R.)
- Department of Biological and Medical Sciences, University of Bordeaux, 33000 Bordeaux, France
| | - Thomas Boyer
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; (C.K.); (S.M.); (M.M.); (A.M.); (T.B.); (N.L.); (G.R.)
- Department of Biological and Medical Sciences, University of Bordeaux, 33000 Bordeaux, France
| | - Nicolas Larmonier
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; (C.K.); (S.M.); (M.M.); (A.M.); (T.B.); (N.L.); (G.R.)
- Department of Biological and Medical Sciences, University of Bordeaux, 33000 Bordeaux, France
| | - Grégoire Robert
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; (C.K.); (S.M.); (M.M.); (A.M.); (T.B.); (N.L.); (G.R.)
- Department of Urology, University Hospital of Bordeaux, 33000 Bordeaux, France
- Department of Biological and Medical Sciences, University of Bordeaux, 33000 Bordeaux, France
| | - Charlotte Domblides
- CNRS UMR 5164, ImmunoConcEpT, Biological and Medical Sciences Department, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; (C.K.); (S.M.); (M.M.); (A.M.); (T.B.); (N.L.); (G.R.)
- Department of Biological and Medical Sciences, University of Bordeaux, 33000 Bordeaux, France
- Department of Medical Oncology, University Hospital of Bordeaux, 33000 Bordeaux, France
| |
Collapse
|
12
|
Yeon Kim S, Tang M, Lu T, Chih SY, Li W. Ferroptosis in glioma therapy: advancements in sensitizing strategies and the complex tumor-promoting roles. Brain Res 2024; 1840:149045. [PMID: 38821335 PMCID: PMC11323215 DOI: 10.1016/j.brainres.2024.149045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/03/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Ferroptosis, an iron-dependent form of non-apoptotic regulated cell death, is induced by the accumulation of lipid peroxides on cellular membranes. Over the past decade, ferroptosis has emerged as a crucial process implicated in various physiological and pathological systems. Positioned as an alternative modality of cell death, ferroptosis holds promise for eliminating cancer cells that have developed resistance to apoptosis induced by conventional therapeutics. This has led to a growing interest in leveraging ferroptosis for cancer therapy across diverse malignancies. Gliomas are tumors arising from glial or precursor cells, with glioblastoma (GBM) being the most common malignant primary brain tumor that is associated with a dismal prognosis. This review provides a summary of recent advancements in the exploration of ferroptosis-sensitizing methods, with a specific focus on their potential application in enhancing the treatment of gliomas. In addition to summarizing the therapeutic potential, this review also discusses the intricate interplay of ferroptosis and its potential tumor-promoting roles within gliomas. Recognizing these dual roles is essential, as they could potentially complicate the therapeutic benefits of ferroptosis. Exploring strategies aimed at circumventing these tumor-promoting roles could enhance the overall therapeutic efficacy of ferroptosis in the context of glioma treatment.
Collapse
Affiliation(s)
- Soo Yeon Kim
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Miaolu Tang
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Tong Lu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Stephen Y Chih
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA; Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
13
|
Luo M, He N, Xu Q, Wen Z, Wang Z, Zhao J, Liu Y. Roles of prostaglandins in immunosuppression. Clin Immunol 2024; 265:110298. [PMID: 38909972 DOI: 10.1016/j.clim.2024.110298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
Prostaglandins (PGs) play a crucial and multifaceted role in various physiological processes such as intercellular signaling, inflammation regulation, neurotransmission, vasodilation, vasoconstriction, and reproductive functions. The diversity and biological significance of these effects are contingent upon the specific types or subtypes of PGs, with each PG playing a crucial role in distinct physiological and pathological processes. Particularly within the immune system, PGs are essential in modulating the function of immune cells and the magnitude and orientation of immune responses. Hence, a comprehensive comprehension of the functions PG signaling pathways in immunosuppressive regulation holds substantial clinical relevance for disease prevention and treatment strategies. The manuscript provides a review of recent developments in PG signaling in immunosuppressive regulation. Furthermore, the potential clinical applications of PGs in immunosuppression are also discussed. While research into the immunosuppressive effects of PGs required further exploration, targeted therapies against their immunosuppressive pathways might open new avenues for disease prevention and treatment.
Collapse
Affiliation(s)
- Minjie Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Nina He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Qing Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Zhongchi Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Ziqin Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China.
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China.
| |
Collapse
|
14
|
Zhou Z, Wang J, Wang J, Yang S, Wang R, Zhang G, Li Z, Shi R, Wang Z, Lu Q. Deciphering the tumor immune microenvironment from a multidimensional omics perspective: insight into next-generation CAR-T cell immunotherapy and beyond. Mol Cancer 2024; 23:131. [PMID: 38918817 PMCID: PMC11201788 DOI: 10.1186/s12943-024-02047-2] [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: 03/25/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Tumor immune microenvironment (TIME) consists of intra-tumor immunological components and plays a significant role in tumor initiation, progression, metastasis, and response to therapy. Chimeric antigen receptor (CAR)-T cell immunotherapy has revolutionized the cancer treatment paradigm. Although CAR-T cell immunotherapy has emerged as a successful treatment for hematologic malignancies, it remains a conundrum for solid tumors. The heterogeneity of TIME is responsible for poor outcomes in CAR-T cell immunotherapy against solid tumors. The advancement of highly sophisticated technology enhances our exploration in TIME from a multi-omics perspective. In the era of machine learning, multi-omics studies could reveal the characteristics of TIME and its immune resistance mechanism. Therefore, the clinical efficacy of CAR-T cell immunotherapy in solid tumors could be further improved with strategies that target unfavorable conditions in TIME. Herein, this review seeks to investigate the factors influencing TIME formation and propose strategies for improving the effectiveness of CAR-T cell immunotherapy through a multi-omics perspective, with the ultimate goal of developing personalized therapeutic approaches.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jiahui Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Nephrology, Union Medical College Hospital, Chinese Academy of Medical Sciences, PekingBeijing, 100730, China
| | - Jiaojiao Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shuai Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruizhi Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
15
|
Hao X, Wang S, Wang L, Li J, Li Y, Liu J. Exosomes as drug delivery systems in glioma immunotherapy. J Nanobiotechnology 2024; 22:340. [PMID: 38890722 PMCID: PMC11184820 DOI: 10.1186/s12951-024-02611-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024] Open
Abstract
Recently, the significant benefits of cancer immunotherapy for most cancers have been demonstrated in clinical and preclinical studies. However, the efficacy of these immunotherapies for gliomas is limited, owing to restricted drug delivery and insufficient immune activation. As drug carriers, exosomes offer the advantages of low toxicity, good biocompatibility, and intrinsic cell targeting, which could enhance glioma immunotherapy efficacy. However, a review of exosome-based drug delivery systems for glioma immunotherapy has not been presented. This review introduces the current problems in glioma immunotherapy and the role of exosomes in addressing these issues. Meanwhile, preparation and application strategies of exosome-based drug delivery systems for glioma immunotherapy are discussed, especially for enhancing immunogenicity and reversing the immunosuppressive tumor microenvironment. Finally, we briefly describe the challenges of exosome-based drug delivery systems in clinical translation. We anticipate that this review will guide the use of exosomes as drug carriers for glioma immunotherapy.
Collapse
Affiliation(s)
- Xinqing Hao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193 Lianhe Road, Dalian, Liaoning, 116011, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57 Xinda Road, Dalian, Liaoning, 116085, China
| | - Shiming Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, No. 193 Lianhe Road, Dalian, Liaoning, 116011, China
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193 Lianhe Road, Dalian, Liaoning, 116011, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57 Xinda Road, Dalian, Liaoning, 116085, China
| | - Jiaqi Li
- Reproductive Medicine Center, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116011, China
| | - Ying Li
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193 Lianhe Road, Dalian, Liaoning, 116011, China.
- Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57 Xinda Road, Dalian, Liaoning, 116085, China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193 Lianhe Road, Dalian, Liaoning, 116011, China.
- Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57 Xinda Road, Dalian, Liaoning, 116085, China.
| |
Collapse
|
16
|
Zhang X, Liang Q, Cao Y, Yang T, An M, Liu Z, Yang J, Liu Y. Dual depletion of myeloid-derived suppressor cells and tumor cells with self-assembled gemcitabine-celecoxib nano-twin drug for cancer chemoimmunotherapy. J Nanobiotechnology 2024; 22:319. [PMID: 38849938 PMCID: PMC11161946 DOI: 10.1186/s12951-024-02598-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: 11/25/2023] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) have played a significant role in facilitating tumor immune escape and inducing an immunosuppressive tumor microenvironment. Eliminating MDSCs and tumor cells remains a major challenge in cancer immunotherapy. A novel approach has been developed using gemcitabine-celecoxib twin drug-based nano-assembled carrier-free nanoparticles (GEM-CXB NPs) for dual depletion of MDSCs and tumor cells in breast cancer chemoimmunotherapy. The GEM-CXB NPs exhibit prolonged blood circulation, leading to the preferential accumulation and co-release of GEM and CXB in tumors. This promotes synergistic chemotherapeutic activity by the proliferation inhibition and apoptosis induction against 4T1 tumor cells. In addition, it enhances tumor immunogenicity by immunogenic cell death induction and MDSC-induced immunosuppression alleviation through the depletion of MDSCs. These mechanisms synergistically activate the antitumor immune function of cytotoxic T cells and natural killer cells, inhibit the proliferation of regulatory T cells, and promote the M2 to M1 phenotype repolarization of tumor-associated macrophages, considerably enhancing the overall antitumor and anti-metastasis efficacy in BALB/c mice bearing 4T1 tumors. The simplified engineering of GEM-CXB NPs, with their dual depletion strategy targeting immunosuppressive cells and tumor cells, represents an advanced concept in cancer chemoimmunotherapy.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Qiangwei Liang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Yongjin Cao
- Department of Pharmacy, School of Nursing, Wuxi Taihu University, Wuxi, 214064, China
| | - Ting Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Min An
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Zihan Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiayu Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
17
|
Montoya M, Collins SA, Chuntova P, Patel TS, Nejo T, Yamamichi A, Kasahara N, Okada H. IRF8-driven reprogramming of the immune microenvironment enhances anti-tumor adaptive immunity and reduces immunosuppression in murine glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587608. [PMID: 38617245 PMCID: PMC11014587 DOI: 10.1101/2024.04.02.587608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Background Glioblastoma (GBM) has a highly immunosuppressive tumor immune microenvironment (TIME), largely mediated by myeloid-derived suppressor cells (MDSCs). Here, we utilized a retroviral replicating vector (RRV) to deliver Interferon Regulatory Factor 8 (IRF8), a master regulator of type 1 conventional dendritic cell (cDC1) development, in a syngeneic murine GBM model. We hypothesized that RRV-mediated delivery of IRF8 could "reprogram" intratumoral MDSCs into antigen-presenting cells (APCs) and thereby restore T-cell responses. Methods Effects of RRV-IRF8 on survival and tumor growth kinetics were examined in the SB28 murine GBM model. Immunophenotype was analyzed by flow cytometry and gene expression assays. We assayed functional immunosuppression and antigen presentation by ex vivo T-cell-myeloid co-culture. Results Mice with RRV-IRF8 pre-transduced intracerebral tumors had significantly longer survival and slower tumor growth compared to controls. RRV-IRF8 treated tumors exhibited significant enrichment of cDC1s and CD8+ T-cells. Additionally, myeloid cells derived from RRV-IRF8 tumors showed decreased expression of the immunosuppressive markers Arg1 and IDO1 and demonstrated reduced suppression of naïve T-cell proliferation in ex vivo co-culture, compared to controls. Furthermore, DCs from RRV-IRF8 tumors showed increased antigen presentation compared to those from control tumors. In vivo treatment with azidothymidine (AZT), a viral replication inhibitor, showed that IRF8 transduction in both tumor and non-tumor cells is necessary for survival benefit, associated with a reprogrammed, cDC1- and CD8 T-cell-enriched TIME. Conclusions Our results indicate that reprogramming of glioma-infiltrating myeloid cells by in vivo expression of IRF8 may reduce immunosuppression and enhance antigen presentation, achieving improved tumor control.
Collapse
Affiliation(s)
- Megan Montoya
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Sara A Collins
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Pavlina Chuntova
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Trishna S Patel
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Takahide Nejo
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Akane Yamamichi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Noriyuki Kasahara
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Hideho Okada
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; The Parker Institute for Cancer Immunotherapy
| |
Collapse
|
18
|
Yang EL, Sun ZJ. Nanomedicine Targeting Myeloid-Derived Suppressor Cells Enhances Anti-Tumor Immunity. Adv Healthc Mater 2024; 13:e2303294. [PMID: 38288864 DOI: 10.1002/adhm.202303294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/27/2023] [Indexed: 02/13/2024]
Abstract
Cancer immunotherapy, a field within immunology that aims to enhance the host's anti-cancer immune response, frequently encounters challenges associated with suboptimal response rates. The presence of myeloid-derived suppressor cells (MDSCs), crucial constituents of the tumor microenvironment (TME), exacerbates this issue by fostering immunosuppression and impeding T cell differentiation and maturation. Consequently, targeting MDSCs has emerged as crucial for immunotherapy aimed at enhancing anti-tumor responses. The development of nanomedicines specifically designed to target MDSCs aims to improve the effectiveness of immunotherapy by transforming immunosuppressive tumors into ones more responsive to immune intervention. This review provides a detailed overview of MDSCs in the TME and current strategies targeting these cells. Also the benefits of nanoparticle-assisted drug delivery systems, including design flexibility, efficient drug loading, and protection against enzymatic degradation, are highlighted. It summarizes advances in nanomedicine targeting MDSCs, covering enhanced treatment efficacy, safety, and modulation of the TME, laying the groundwork for more potent cancer immunotherapy.
Collapse
Affiliation(s)
- En-Li Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, Hubei, 430079, China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, Hubei, 430079, China
| |
Collapse
|
19
|
Tripathy DK, Panda LP, Biswal S, Barhwal K. Insights into the glioblastoma tumor microenvironment: current and emerging therapeutic approaches. Front Pharmacol 2024; 15:1355242. [PMID: 38523646 PMCID: PMC10957596 DOI: 10.3389/fphar.2024.1355242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/07/2024] [Indexed: 03/26/2024] Open
Abstract
Glioblastoma (GB) is an intrusive and recurrent primary brain tumor with low survivability. The heterogeneity of the tumor microenvironment plays a crucial role in the stemness and proliferation of GB. The tumor microenvironment induces tumor heterogeneity of cancer cells by facilitating clonal evolution and promoting multidrug resistance, leading to cancer cell progression and metastasis. It also plays an important role in angiogenesis to nourish the hypoxic tumor environment. There is a strong interaction of neoplastic cells with their surrounding microenvironment that comprise several immune and non-immune cellular components. The tumor microenvironment is a complex network of immune components like microglia, macrophages, T cells, B cells, natural killer (NK) cells, dendritic cells and myeloid-derived suppressor cells, and non-immune components such as extracellular matrix, endothelial cells, astrocytes and neurons. The prognosis of GB is thus challenging, making it a difficult target for therapeutic interventions. The current therapeutic approaches target these regulators of tumor micro-environment through both generalized and personalized approaches. The review provides a summary of important milestones in GB research, factors regulating tumor microenvironment and promoting angiogenesis and potential therapeutic agents widely used for the treatment of GB patients.
Collapse
Affiliation(s)
- Dev Kumar Tripathy
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Lakshmi Priya Panda
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Suryanarayan Biswal
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Kalpana Barhwal
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| |
Collapse
|
20
|
Yokota S, Yonezawa T, Momoi Y, Maeda S. Myeloid derived suppressor cells in peripheral blood can be a prognostic factor in canine transitional cell carcinoma. Vet Immunol Immunopathol 2024; 269:110716. [PMID: 38308864 DOI: 10.1016/j.vetimm.2024.110716] [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: 11/14/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/05/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature cells with immunosuppressive properties found in the tumor microenvironment. MDSCs are divided into two major subsets: polymorphonuclear MDSCs (PMN-MDSCs) and monocytic MDSCs (M-MDSCs). Both MDSC subsets contribute to the creation of an immunosuppressive environment for tumor progression. In humans, patients with high levels of MDSCs show worse outcomes for several types of cancers. However, the association between MDSCs and clinical features has rarely been investigated in canine studies. In the present study, we measured the proportion of PMN-MDSCs and M-MDSCs in the peripheral blood and tumor tissue of dogs with hepatocellular carcinoma (HCC), prostate cancer (PC), transitional cell carcinoma (TCC), lymphoma, and pulmonary adenocarcinoma. Additionally, we examined immunosuppressive ability of PMN-MDSCs and M-MDSCs in peripheral blood mononuclear cells of TCC case on CD4+, CD8+ and interferon-γ+ cells and investigated the relationships of MDSCs with clinical features and outcomes. PMN-MDSCs increased in HCC, PC, TCC, and lymphoma. In contrast, M-MDSCs increased in the TCC. Both PMN-MDSCs and M-MDSCs exhibited immunosuppressive effects on CD8+, CD4+ and interferon-γ+ cells. In dogs with TCC, lymph node metastasis was associated with high level of PMN-MDSCs but not with M-MDSCs. High levels of both PMN-MDSCs and M-MDSCs were related to advanced tumor stage. Kaplan-Meier analysis revealed that high levels of both PMN-MDSCs and M-MDSCs were significantly associated with shorter overall survival. In addition, the Cox proportional hazard regression model showed that M-MDSCs and the tumor stage were independent prognostic factors for TCC. These results suggest that PMN-MDSCs and M-MDSCs may be involved in tumor progression and could be prognostic factors and promising therapeutic targets in dogs with TCC.
Collapse
Affiliation(s)
- Shohei Yokota
- Department of Veterinary Clinical Pathobiology, Guraduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Tomohiro Yonezawa
- Department of Veterinary Clinical Pathobiology, Guraduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yasuyuki Momoi
- Department of Veterinary Clinical Pathobiology, Guraduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Shingo Maeda
- Department of Veterinary Clinical Pathobiology, Guraduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| |
Collapse
|
21
|
Cuenca-Escalona J, Subtil B, Garcia-Perez A, Cambi A, de Vries IJM, Flórez-Grau G. EP2 and EP4 blockade prevents tumor-induced suppressive features in human monocytic myeloid-derived suppressor cells. Front Immunol 2024; 15:1355769. [PMID: 38343540 PMCID: PMC10853404 DOI: 10.3389/fimmu.2024.1355769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/12/2024] [Indexed: 02/15/2024] Open
Abstract
Tumors educate their environment to prime the occurrence of suppressive cell subsets, which enable tumor evasion and favors tumor progression. Among these, there are the myeloid-derived suppressor cells (MDSCs), their presence being associated with the poor clinical outcome of cancer patients. Tumor-derived prostaglandin E2 (PGE2) is known to mediate MDSC differentiation and the acquisition of pro-tumor features. In myeloid cells, PGE2 signaling is mediated via E-prostanoid receptor type 2 (EP2) and EP4. Although the suppressive role of PGE2 is well established in MDSCs, the role of EP2/4 on human MDSCs or whether EP2/4 modulation can prevent MDSCs suppressive features upon exposure to tumor-derived PGE2 is poorly defined. In this study, using an in vitro model of human monocytic-MDSCs (M-MDSCs) we demonstrate that EP2 and EP4 signaling contribute to the induction of a pro-tumor phenotype and function on M-MDSCs. PGE2 signaling via EP2 and EP4 boosted M-MDSC ability to suppress T and NK cell responses. Combined EP2/4 blockade on M-MDSCs during PGE2 exposure prevented the occurrence of these suppressive features. Additionally, EP2/4 blockade attenuated the suppressive phenotype of M-MDSCs in a 3D coculture with colorectal cancer patient-derived organoids. Together, these results identify the role of tumor-derived PGE2 signaling via EP2 and EP4 in this human M-MDSC model, supporting the therapeutic value of targeting PGE2-EP2/4 axis in M-MDSCs to alleviate immunosuppression and facilitate the development of anti-tumor immunity.
Collapse
|
22
|
Chan TYH, Wong JSY, Kiang KMY, Sun CWY, Leung GKK. The duality of CXCR3 in glioblastoma: unveiling autocrine and paracrine mechanisms for novel therapeutic approaches. Cell Death Dis 2023; 14:835. [PMID: 38104126 PMCID: PMC10725418 DOI: 10.1038/s41419-023-06354-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023]
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor associated with limited therapeutic options and a poor prognosis. CXCR3, a chemokine receptor, serves dual autocrine-paracrine functions in cancer. Despite gaps in our understanding of the functional role of the CXCR3 receptor in GBM, it has been shown to hold promise as a therapeutic target for the treatment of GBM. Existing clinical therapeutics and vaccines targeting CXCR3 ligand expression associated with the CXCR3 axes have also shown anti-tumorigenic effects in GBM. This review summarizes existing evidence on the oncogenic function of CXCR3 and its ligands CXCL9, CXCL10, and CXCL11, in GBM, and examines the controversies concerning the immunomodulatory functions of the CXCR3 receptor, including immune T cell recruitment, polarization, and positioning. The mechanisms underlying monotherpies and combination therapies targeting the CXCR3 pathways are discussed. A better understanding of the CXCR3 axes may lead to the development of strategies for overcoming the limitations of existing immunotherapies for GBM.
Collapse
Affiliation(s)
- Travis Yui Hei Chan
- Division of Neurosurgery, Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jenny Sum Yee Wong
- Division of Vascular Surgery, Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Karrie Mei-Yee Kiang
- Division of Neurosurgery, Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cherry Won Yuet Sun
- Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Republic of Ireland
| | - Gilberto Ka-Kit Leung
- Division of Neurosurgery, Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
23
|
Cheung TS, Giacomini C, Cereda M, Avivar-Valderas A, Capece D, Bertolino GM, delaRosa O, Hicks R, Ciccocioppo R, Franzoso G, Galleu A, Ciccarelli FD, Dazzi F. Apoptosis in mesenchymal stromal cells activates an immunosuppressive secretome predicting clinical response in Crohn's disease. Mol Ther 2023; 31:3531-3544. [PMID: 37805713 PMCID: PMC10727969 DOI: 10.1016/j.ymthe.2023.10.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/30/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023] Open
Abstract
In vivo apoptosis of human mesenchymal stromal cells (MSCs) plays a critical role in delivering immunomodulation. Yet, caspase activity not only mediates the dying process but also death-independent functions that may shape the immunogenicity of apoptotic cells. Therefore, a better characterization of the immunological profile of apoptotic MSCs (ApoMSCs) could shed light on their mechanistic action and therapeutic applications. We analyzed the transcriptomes of MSCs undergoing apoptosis and identified several immunomodulatory factors and chemokines dependent on caspase activation following Fas stimulation. The ApoMSC secretome inhibited human T cell proliferation and activation, and chemoattracted monocytes in vitro. Both immunomodulatory activities were dependent on the cyclooxygenase2 (COX2)/prostaglandin E2 (PGE2) axis. To assess the clinical relevance of ApoMSC signature, we used the peripheral blood mononuclear cells (PBMCs) from a cohort of fistulizing Crohn's disease (CD) patients who had undergone MSC treatment (ADMIRE-CD). Compared with healthy donors, MSCs exposed to patients' PBMCs underwent apoptosis and released PGE2 in a caspase-dependent manner. Both PGE2 and apoptosis were significantly associated with clinical responses to MSCs. Our findings identify a new mechanism whereby caspase activation delivers ApoMSC immunosuppression. Remarkably, such molecular signatures could implicate translational tools for predicting patients' clinical responses to MSC therapy in CD.
Collapse
Affiliation(s)
- Tik Shing Cheung
- School of Cancer and Pharmacological Sciences, King's College London, London, UK
| | - Chiara Giacomini
- School of Cancer and Pharmacological Sciences, King's College London, London, UK; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Matteo Cereda
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; Italian Institute for Genomic Medicine, c/o IRCCS, Str. Prov.le 142, km 3.95, 10060 Candiolo, TO, Italy
| | | | - Daria Capece
- Centre for Molecular Immunology and Inflammation, Department of Immunology and Inflammation, Imperial College London, London, UK
| | | | - Olga delaRosa
- Takeda Madrid, Cell Therapy Technology Center, Tres Cantos, Spain
| | - Ryan Hicks
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK; BioPharmaceuticals R&D Cell Therapy, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy
| | - Guido Franzoso
- Centre for Molecular Immunology and Inflammation, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Antonio Galleu
- School of Cancer and Pharmacological Sciences, King's College London, London, UK
| | - Francesca D Ciccarelli
- School of Cancer and Pharmacological Sciences, King's College London, London, UK; Cancer Systems Biology Laboratory, The Francis Crick Institute, London, UK
| | - Francesco Dazzi
- School of Cancer and Pharmacological Sciences, King's College London, London, UK; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK; BioPharmaceuticals R&D Cell therapy, AstraZeneca, Cambridge, UK.
| |
Collapse
|
24
|
Rajgopal S, Nakano K, Cook LM. Beyond the horizon: Neutrophils leading the way in the evolution of immunotherapy. Cancer Med 2023; 12:21885-21904. [PMID: 38062888 PMCID: PMC10757139 DOI: 10.1002/cam4.6761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/07/2023] [Accepted: 11/16/2023] [Indexed: 12/31/2023] Open
Abstract
Cancer is a complex and dynamic disease, initiated by a multitude of intrinsic mutations and progressed with the assistance of the tissue microenvironment, encompassed by stromal cells including immune cell infiltration. The novel finding that tumors can evade anti-cancer immune functions shaped the field of immunotherapy, which has been a revolutionary approach for the treatment of cancers. However, the development of predominantly T cell-targeted immunotherapy approaches, such as immune checkpoint inhibition, also brought about an accumulation of evidence demonstrating other immune cell drivers of tumor progression, such as innate immune cells and notably, neutrophils. In the past decade, neutrophils have emerged to be primary mediators of multiple cancer types and even in recent years, are gaining attention for their potential use in the next generation of immunotherapies. Here, we review current immunotherapy strategies and thoroughly discuss the roles of neutrophils in cancer and novel neutrophil-targeted methods for treating cancer.
Collapse
Affiliation(s)
- Sanjana Rajgopal
- Department of Pathology and MicrobiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Genetics, Cell Biology, and AnatomyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Kosuke Nakano
- Department of Pathology and MicrobiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Leah M. Cook
- Department of Pathology and MicrobiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterOmahaNebraskaUSA
| |
Collapse
|
25
|
Yi M, Li T, Niu M, Mei Q, Zhao B, Chu Q, Dai Z, Wu K. Exploiting innate immunity for cancer immunotherapy. Mol Cancer 2023; 22:187. [PMID: 38008741 PMCID: PMC10680233 DOI: 10.1186/s12943-023-01885-w] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/28/2023] Open
Abstract
Immunotherapies have revolutionized the treatment paradigms of various types of cancers. However, most of these immunomodulatory strategies focus on harnessing adaptive immunity, mainly by inhibiting immunosuppressive signaling with immune checkpoint blockade, or enhancing immunostimulatory signaling with bispecific T cell engager and chimeric antigen receptor (CAR)-T cell. Although these agents have already achieved great success, only a tiny percentage of patients could benefit from immunotherapies. Actually, immunotherapy efficacy is determined by multiple components in the tumor microenvironment beyond adaptive immunity. Cells from the innate arm of the immune system, such as macrophages, dendritic cells, myeloid-derived suppressor cells, neutrophils, natural killer cells, and unconventional T cells, also participate in cancer immune evasion and surveillance. Considering that the innate arm is the cornerstone of the antitumor immune response, utilizing innate immunity provides potential therapeutic options for cancer control. Up to now, strategies exploiting innate immunity, such as agonists of stimulator of interferon genes, CAR-macrophage or -natural killer cell therapies, metabolic regulators, and novel immune checkpoint blockade, have exhibited potent antitumor activities in preclinical and clinical studies. Here, we summarize the latest insights into the potential roles of innate cells in antitumor immunity and discuss the advances in innate arm-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ming Yi
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
| | - Bin Zhao
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
26
|
Rocha Pinheiro SL, Lemos FFB, Marques HS, Silva Luz M, de Oliveira Silva LG, Faria Souza Mendes dos Santos C, da Costa Evangelista K, Calmon MS, Sande Loureiro M, Freire de Melo F. Immunotherapy in glioblastoma treatment: Current state and future prospects. World J Clin Oncol 2023; 14:138-159. [PMID: 37124134 PMCID: PMC10134201 DOI: 10.5306/wjco.v14.i4.138] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023] Open
Abstract
Glioblastoma remains as the most common and aggressive malignant brain tumor, standing with a poor prognosis and treatment prospective. Despite the aggressive standard care, such as surgical resection and chemoradiation, median survival rates are low. In this regard, immunotherapeutic strategies aim to become more attractive for glioblastoma, considering its recent advances and approaches. In this review, we provide an overview of the current status and progress in immunotherapy for glioblastoma, going through the fundamental knowledge on immune targeting to promising strategies, such as Chimeric antigen receptor T-Cell therapy, immune checkpoint inhibitors, cytokine-based treatment, oncolytic virus and vaccine-based techniques. At last, it is discussed innovative methods to overcome diverse challenges, and future perspectives in this area.
Collapse
Affiliation(s)
- Samuel Luca Rocha Pinheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabian Fellipe Bueno Lemos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Hanna Santos Marques
- Campus Vitória da Conquista, Universidade Estadual do Sudoeste da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Marcel Silva Luz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | | | | | - Mariana Santos Calmon
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Matheus Sande Loureiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
27
|
Perioperative escape from dormancy of spontaneous micro-metastases: A role for malignant secretion of IL-6, IL-8, and VEGF, through adrenergic and prostaglandin signaling. Brain Behav Immun 2023; 109:175-187. [PMID: 36646396 DOI: 10.1016/j.bbi.2023.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/08/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
We recently showed that a minimally-invasive removal of MDA-MB-231HM primary tumors (PTs) and elimination of their secreted factors (including IL-6, IL-8, VEGF, EGF, PDGF-aa, MIF, SerpinE1, and M-CSF), caused regression of spontaneous micro-metastases into a non-growing dormant state. To explore the underlying mechanisms and potential clinical ramifications of this phenomenon, we herein used the MDA-MB-231HM human breast cancer cell-line, in-vitro, and in vivo following orthotopic implantation in immune-deficient BALB/C nu/nu mice. Employing bioluminescence imaging, we found that adding laparotomy to minimally-invasive removal of the PT caused an outbreak of micro-metastases. However, perioperative β-adrenergic and COX-2 inhibition, using propranolol + etodolac, maintained metastatic dormancy following laparotomy. In-vitro, β-adrenergic agonists (epinephrine or metaproterenol) and prostaglandin-E2 markedly increased MDA-MB-231HM secretion of the pro-metastatic factors IL-6, IL-8, and VEGF, whereas cortisol reduced their secretion, effects that were maintained even 12 h after the washout of these agonists. In-vivo, laparotomy elevated IL-6 and IL-8 levels in both plasma and ex-vivo PT spontaneous secretion, whereas perioperative propranolol + etodolac administration blocked these effects. Similar trends were evident for EGF and MIF. Promoter-based bioinformatics analyses of excised PT transcriptomes implicated elevated NF-kB activity and reduced IRF1 activity in the gene regulatory effects of laparotomy, and these effects were inhibited by pre-surgical propranolol + etodolac. Taken together, our findings suggest a novel mechanism of post-operative metastatic outbreak, where surgery-induced adrenergic and prostanoid signaling increase the secretion of pro-metastatic factors, including IL-6, IL-8, and VEGF, from PT and possibly residual malignant tissue, and thereby prevent residual disease from entering dormancy.
Collapse
|
28
|
Wen J, Xuan B, Gao Y, Liu Y, Wang L, He L, Meng X, Zhou T, Tao Y, Guo K, Wang Y. Lnc-17Rik promotes the immunosuppressive function of Myeloid-Derived suppressive cells in esophageal cancer. Cell Immunol 2023; 385:104676. [PMID: 36780770 DOI: 10.1016/j.cellimm.2023.104676] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a population of immature bone marrow cells that accumulate in large numbers in the spleen, peripheral blood, bone marrow, lymph nodes, and local and metastatic foci of tumors. C/EBP homologous protein (CHOP) and CCAAT/enhancer binding protein β (C/EBPβ) play key roles in regulating the immunosuppressive function and differentiation of MDSCs. Our study revealed that the long noncoding RNA Lnc-17Rik was able to promote immunosuppression in tumors by facilitating the activation and expression of key genes involved in MDSC differentiation. Lnc-17Rik was shown to directly interact with CHOP and C/EBPβ LIP to facilitate their dissociation from the transcriptional repressor complex involving C/EBP LAP/LIP/CHOP. Moreover, Lnc-17Rik increased the association of WD repeat-containing protein 5 (WDR5) with C/EBP LAP, promoting H3K4me3 enrichment in the promoter regions of arginase 1 (Arg-1), cyclooxygenase 2 (COX2), nitric oxide synthase 2 (NOS2) and NADPH oxidase 2 (NOX2) to enhance the expression of these genes. Furthermore, using a CD45 chimeric model we confirmed that Lnc-17Rik promoted the differentiation of monocytic (M)-MDSCs in vivo with the introduction of Lnc-17Rik-overexpressing MDSCs shown to promote tumor growth as a result of enhancing their immunosuppressive function. Notably, human Lnc-17Rik is highly homologous to mouse Lnc-17Rik and fulfills similar functions in human MDSC-like cells. In addition, we also found a high level of Lnc-17Rik in peripheral blood MDSC of patients with esophageal cancer. These findings suggest that Lnc-17Rik plays an important role in controlling the immunosuppressive function of MDSCs in the tumor environment and may further serve as a potential therapeutic target for patients with esophageal cancer.
Collapse
Affiliation(s)
- Jiexia Wen
- Department of Central Laboratory, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Bin Xuan
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Yunhuan Gao
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Yang Liu
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Liwei Wang
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Li He
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Xiangcai Meng
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Tao Zhou
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Yang Tao
- Department of Central Laboratory, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Kening Guo
- Department of Central Laboratory, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Yimin Wang
- Department of Central Laboratory, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China; Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China.
| |
Collapse
|
29
|
Korbecki J, Rębacz-Maron E, Kupnicka P, Chlubek D, Baranowska-Bosiacka I. Synthesis and Significance of Arachidonic Acid, a Substrate for Cyclooxygenases, Lipoxygenases, and Cytochrome P450 Pathways in the Tumorigenesis of Glioblastoma Multiforme, Including a Pan-Cancer Comparative Analysis. Cancers (Basel) 2023; 15:cancers15030946. [PMID: 36765904 PMCID: PMC9913267 DOI: 10.3390/cancers15030946] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive gliomas. New and more effective therapeutic approaches are being sought based on studies of the various mechanisms of GBM tumorigenesis, including the synthesis and metabolism of arachidonic acid (ARA), an omega-6 polyunsaturated fatty acid (PUFA). PubMed, GEPIA, and the transcriptomics analysis carried out by Seifert et al. were used in writing this paper. In this paper, we discuss in detail the biosynthesis of this acid in GBM tumors, with a special focus on certain enzymes: fatty acid desaturase (FADS)1, FADS2, and elongation of long-chain fatty acids family member 5 (ELOVL5). We also discuss ARA metabolism, particularly its release from cell membrane phospholipids by phospholipase A2 (cPLA2, iPLA2, and sPLA2) and its processing by cyclooxygenases (COX-1 and COX-2), lipoxygenases (5-LOX, 12-LOX, 15-LOX-1, and 15-LOX-2), and cytochrome P450. Next, we discuss the significance of lipid mediators synthesized from ARA in GBM cancer processes, including prostaglandins (PGE2, PGD2, and 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2)), thromboxane A2 (TxA2), oxo-eicosatetraenoic acids, leukotrienes (LTB4, LTC4, LTD4, and LTE4), lipoxins, and many others. These lipid mediators can increase the proliferation of GBM cancer cells, cause angiogenesis, inhibit the anti-tumor response of the immune system, and be responsible for resistance to treatment.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Ewa Rębacz-Maron
- Department of Ecology and Anthropology, Institute of Biology, University of Szczecin, Wąska 13, 71-415 Szczecin, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Correspondence: ; Tel.: +48-914-661-515
| |
Collapse
|
30
|
Mizutani T, Ano T, Yoshioka Y, Mizuta S, Takemoto K, Ouchi Y, Morita D, Kitano S, Miyachi H, Tsuruyama T, Fujiwara N, Sugita M. Neutrophil S100A9 supports M2 macrophage niche formation in granulomas. iScience 2023; 26:106081. [PMID: 36843852 PMCID: PMC9947307 DOI: 10.1016/j.isci.2023.106081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/02/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Mycobacterium infection gives rise to granulomas predominantly composed of inflammatory M1-like macrophages, with bacteria-permissive M2 macrophages also detected in deep granulomas. Our histological analysis of Mycobacterium bovis bacillus Calmette-Guerin-elicited granulomas in guinea pigs revealed that S100A9-expressing neutrophils bordered a unique M2 niche within the inner circle of concentrically multilayered granulomas. We evaluated the effect of S100A9 on macrophage M2 polarization based on guinea pig studies. S100A9-deficient mouse neutrophils abrogated M2 polarization, which was critically dependent on COX-2 signaling in neutrophils. Mechanistic evidence suggested that nuclear S100A9 interacts with C/EBPβ, which cooperatively activates the Cox-2 promoter and amplifies prostaglandin E2 production, followed by M2 polarization in proximal macrophages. Because the M2 populations in guinea pig granulomas were abolished via treatment with celecoxib, a selective COX-2 inhibitor, we propose the S100A9/Cox-2 axis as a major pathway driving M2 niche formation in granulomas.
Collapse
Affiliation(s)
- Tatsuaki Mizutani
- Laboratory of Cell Regulation, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan,Laboratory of Cell Regulation and Molecular Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan,Corresponding author
| | - Toshiaki Ano
- Laboratory of Cell Regulation, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan,Laboratory of Cell Regulation and Molecular Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yuya Yoshioka
- Laboratory of Cell Regulation, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan,Laboratory of Cell Regulation and Molecular Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Satoshi Mizuta
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Keiko Takemoto
- Laboratory of Immune Regulation, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yuki Ouchi
- Laboratory of Cell Regulation, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan,Laboratory of Cell Regulation and Molecular Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Daisuke Morita
- Laboratory of Cell Regulation, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan,Laboratory of Cell Regulation and Molecular Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Satsuki Kitano
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hitoshi Miyachi
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Tatsuaki Tsuruyama
- Department of Drug Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Masahiko Sugita
- Laboratory of Cell Regulation, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan,Laboratory of Cell Regulation and Molecular Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
31
|
Jin K, Qian C, Lin J, Liu B. Cyclooxygenase-2-Prostaglandin E2 pathway: A key player in tumor-associated immune cells. Front Oncol 2023; 13:1099811. [PMID: 36776289 PMCID: PMC9911818 DOI: 10.3389/fonc.2023.1099811] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/12/2023] [Indexed: 01/29/2023] Open
Abstract
Cyclooxygenases-2 (COX-2) and Prostaglandin E2 (PGE2), which are important in chronic inflammatory diseases, can increase tumor incidence and promote tumor growth and metastasis. PGE2 binds to various prostaglandin E receptors to activate specific downstream signaling pathways such as PKA pathway, β-catenin pathway, NF-κB pathway and PI3K/AKT pathway, all of which play important roles in biological and pathological behavior. Nonsteroidal anti-inflammatory drugs (NSAIDs), which play as COX-2 inhibitors, and EP antagonists are important in anti-tumor immune evasion. The COX-2-PGE2 pathway promotes tumor immune evasion by regulating myeloid-derived suppressor cells, lymphocytes (CD8+ T cells, CD4+ T cells and natural killer cells), and antigen presenting cells (macrophages and dendritic cells). Based on conventional treatment, the addition of COX-2 inhibitors or EP antagonists may enhance immunotherapy response in anti-tumor immune escape. However, there are still a lot of challenges in cancer immunotherapy. In this review, we focus on how the COX-2-PGE2 pathway affects tumor-associated immune cells.
Collapse
Affiliation(s)
- Kaipeng Jin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chao Qian
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jinti Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China,*Correspondence: Bing Liu, ; Jinti Lin,
| | - Bing Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China,*Correspondence: Bing Liu, ; Jinti Lin,
| |
Collapse
|
32
|
Menter DG, Bresalier RS. An Aspirin a Day: New Pharmacological Developments and Cancer Chemoprevention. Annu Rev Pharmacol Toxicol 2023; 63:165-186. [PMID: 36202092 DOI: 10.1146/annurev-pharmtox-052020-023107] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Chemoprevention refers to the use of natural or synthetic agents to reverse, suppress, or prevent the progression or recurrence of cancer. A large body of preclinical and clinical data suggest the ability of aspirin to prevent precursor lesions and cancers, but much of the clinical data are inferential and based on descriptive epidemiology, case control, and cohort studies or studies designed to answer other questions (e.g., cardiovascular mortality). Multiple pharmacological, clinical, and epidemiologic studies suggest that aspirin can prevent certain cancers but may also cause other effects depending on the tissue or disease and organ site in question. The best-known biological targets of aspirin are cyclooxygenases, which drive a wide variety of functions, including hemostasis, inflammation, and immune modulation. Newly recognized molecular and cellular interactions suggest additional modifiable functional targets, and the existence of consensus molecular cancer subtypes suggests that aspirin may have differential effects based on tumor heterogeneity. This review focuses on new pharmacological developments and innovations in biopharmacology that clarify the potential role of aspirin in cancer chemoprevention.
Collapse
Affiliation(s)
- David G Menter
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert S Bresalier
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA;
| |
Collapse
|
33
|
Akter Z, Salamat N, Ali MY, Zhang L. The promise of targeting heme and mitochondrial respiration in normalizing tumor microenvironment and potentiating immunotherapy. Front Oncol 2023; 12:1072739. [PMID: 36686754 PMCID: PMC9851275 DOI: 10.3389/fonc.2022.1072739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Cancer immunotherapy shows durable treatment responses and therapeutic benefits compared to other cancer treatment modalities, but many cancer patients display primary and acquired resistance to immunotherapeutics. Immunosuppressive tumor microenvironment (TME) is a major barrier to cancer immunotherapy. Notably, cancer cells depend on high mitochondrial bioenergetics accompanied with the supply of heme for their growth, proliferation, progression, and metastasis. This excessive mitochondrial respiration increases tumor cells oxygen consumption, which triggers hypoxia and irregular blood vessels formation in various regions of TME, resulting in an immunosuppressive TME, evasion of anti-tumor immunity, and resistance to immunotherapeutic agents. In this review, we discuss the role of heme, heme catabolism, and mitochondrial respiration on mediating immunosuppressive TME by promoting hypoxia, angiogenesis, and leaky tumor vasculature. Moreover, we discuss the therapeutic prospects of targeting heme and mitochondrial respiration in alleviating tumor hypoxia, normalizing tumor vasculature, and TME to restore anti-tumor immunity and resensitize cancer cells to immunotherapy.
Collapse
|
34
|
Friedmann-Morvinski D, Hambardzumyan D. Monocyte-neutrophil entanglement in glioblastoma. J Clin Invest 2023; 133:163451. [PMID: 36594465 PMCID: PMC9797336 DOI: 10.1172/jci163451] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Glioblastoma (GBM) is the most belligerent and frequent brain tumor in adults. Research over the past two decades has provided increased knowledge of the genomic and molecular landscape of GBM and highlighted the presence of a high degree of inter- and intratumor heterogeneity within the neoplastic compartment. It is now appreciated that GBMs are composed of multiple distinct and impressionable neoplastic and non-neoplastic cell types that form the unique brain tumor microenvironment (TME). Non-neoplastic cells in the TME form reciprocal interactions with neoplastic cells to promote tumor growth and invasion, and together they influence the tumor response to standard-of-care therapies as well as emerging immunotherapies. One of the most prevalent non-neoplastic cell types in the GBM TME are myeloid cells, the most abundant of which are of hematopoietic origin, including monocytes/monocyte-derived macrophages. Less abundant, although still a notable presence, are neutrophils of hematopoietic origin and intrinsic brain-resident microglia. In this Review we focus on neutrophils and monocytes that infiltrate tumors from the blood circulation, their heterogeneity, and their interactions with neoplastic cells and other non-neoplastic cells in the TME. We conclude with an overview of challenges in targeting these cells and discuss avenues for therapeutic exploitation to improve the dismal outcomes of patients with GBM.
Collapse
Affiliation(s)
- Dinorah Friedmann-Morvinski
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, and,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, Tisch Cancer Institute, and,Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
35
|
Anwar MM, Albanese C, Hamdy NM, Sultan AS. Rise of the natural red pigment 'prodigiosin' as an immunomodulator in cancer. Cancer Cell Int 2022; 22:419. [PMID: 36577970 PMCID: PMC9798661 DOI: 10.1186/s12935-022-02815-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/29/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer is a heterogeneous disease with multifaceted drug resistance mechanisms (e.g., tumour microenvironment [TME], tumour heterogeneity, and immune evasion). Natural products are interesting repository of bioactive molecules, especially those with anticancer activities. Prodigiosin, a red pigment produced by Serratia marcescens, possesses inherent anticancer characteristics, showing interesting antitumour activities in different cancers (e.g., breast, gastric) with low or without harmful effects on normal cells. The present review discusses the potential role of prodigiosin in modulating and reprogramming the metabolism of the various immune cells in the TME, such as T and B lymphocytes, tumour-associated macrophages (TAMs), natural killer (NK) cells, and tumour-associated dendritic cells (TADCs), and myeloid-derived suppressor cells (MDSCs) which in turn might introduce as an immunomodulator in cancer therapy.
Collapse
Affiliation(s)
- Mohammed Moustapha Anwar
- grid.7155.60000 0001 2260 6941Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt
| | - Chris Albanese
- grid.516085.f0000 0004 0606 3221Oncology and Radiology Departments, Lombardi Comprehensive Cancer Center, Washington, D.C. USA
| | - Nadia M. Hamdy
- Department of Biochemistry, Ain Shams Faculty of Pharmacy, Cairo, Egypt
| | - Ahmed S. Sultan
- grid.7155.60000 0001 2260 6941Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
36
|
Shibata M, Nanno K, Yoshimori D, Nakajima T, Takada M, Yazawa T, Mimura K, Inoue N, Watanabe T, Tachibana K, Muto S, Momma T, Suzuki Y, Kono K, Endo S, Takenoshita S. Myeloid-derived suppressor cells: Cancer, autoimmune diseases, and more. Oncotarget 2022; 13:1273-1285. [PMID: 36395389 PMCID: PMC9671473 DOI: 10.18632/oncotarget.28303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although cancer immunotherapy using immune checkpoint inhibitors (ICIs) has been recognized as one of the major treatment modalities for malignant diseases, the clinical outcome is not uniform in all cancer patients. Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells that possess various strong immunosuppressive activities involving multiple immunocompetent cells that are significantly accumulated in patients who did not respond well to cancer immunotherapies. We reviewed the perspective of MDSCs with emerging evidence in this review. Many studies on MDSCs were performed in malignant diseases. Substantial studies on the participation of MDSCs on non-malignant diseases such as chronic infection and autoimmune diseases, and physiological roles in obesity, aging, pregnancy and neonates have yet to be reported. With the growing understanding of the roles of MDSCs, variable therapeutic strategies and agents targeting MDSCs are being investigated, some of which have been used in clinical trials. More studies are required in order to develop more effective strategies against MDSCs.
Collapse
Affiliation(s)
- Masahiko Shibata
- 1Department of Comprehensive Cancer Treatment and Research at Aizu, Fukushima Medical University, Fukushima, Japan,2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan,Correspondence to:Masahiko Shibata, email:
| | - Kotaro Nanno
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,5Department of Surgery, Nippon Medical School, Tokyo, Japan
| | - Daigo Yoshimori
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,5Department of Surgery, Nippon Medical School, Tokyo, Japan
| | - Takahiro Nakajima
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan
| | - Makoto Takada
- 4Aizu Oncology Consortium, Fukushima, Japan,6Department of Surgery, Bange Kousei General Hospital, Fukushima, Japan
| | - Takashi Yazawa
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan
| | - Kousaku Mimura
- 3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan
| | - Norio Inoue
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan
| | - Takafumi Watanabe
- 7Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima, Japan
| | | | - Satoshi Muto
- 9Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Tomoyuki Momma
- 3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan
| | - Yoshiyuki Suzuki
- 1Department of Comprehensive Cancer Treatment and Research at Aizu, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan,10Department of Radiation Oncology, Fukushima Medical University, Fukushima, Japan
| | - Koji Kono
- 1Department of Comprehensive Cancer Treatment and Research at Aizu, Fukushima Medical University, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan
| | - Shungo Endo
- 11Department of Colorectoanal Surgery, Aizu Medical Center, Fukushima Medical University, Fukushima, Japan
| | | |
Collapse
|
37
|
Cheng X, Zhang H, Hamad A, Huang H, Tsung A. Surgery-mediated tumor-promoting effects on the immune microenvironment. Semin Cancer Biol 2022; 86:408-419. [PMID: 35066156 PMCID: PMC11770836 DOI: 10.1016/j.semcancer.2022.01.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023]
Abstract
Surgical resection continues to be the mainstay treatment for solid cancers even though chemotherapy and immunotherapy have significantly improved patient overall survival and progression-free survival. Numerous studies have shown that surgery induces the dissemination of circulating tumor cells (CTCs) and that the resultant inflammatory response promotes occult tumor growth and the metastatic process by forming a supportive tumor microenvironment (TME). Surgery-induced platelet activation is one of the initial responses to a wound and the formation of fibrin clots can provide the scaffold for recruited inflammatory cells. Activated platelets can also shield CTCs to protect them from blood shear forces and promote CTCs evasion of immune destruction. Similarly, neutrophils are recruited to the fibrin clot and enhance cancer metastatic dissemination and progression by forming neutrophil extracellular traps (NETs). Activated macrophages are also recruited to surgical sites to facilitate the metastatic spread. More importantly, the body's response to surgical insult results in the recruitment and expansion of immunosuppressive cell populations (i.e. myeloid-derived suppressor cells and regulatory T cells) and in the suppression of natural killer (NK) cells that contribute to postoperative cancer recurrence and metastasis. In this review, we seek to provide an overview of the pro-tumorigenic mechanisms resulting from surgery's impact on these cells in the TME. Further understanding of these events will allow for the development of perioperative therapeutic strategies to prevent surgery-associated metastasis.
Collapse
Affiliation(s)
- Xiang Cheng
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Hongji Zhang
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Ahmad Hamad
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Hai Huang
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Allan Tsung
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| |
Collapse
|
38
|
Wang T, Lin M, Mao J, Tian L, Gan H, Hu X, Yan L, Long H, Cai J, Zheng X, Xiao Y, Li D, Shuai X, Pang P. Inflammation-Regulated Nanodrug Sensitizes Hepatocellular Carcinoma to Checkpoint Blockade Therapy by Reprogramming the Tumor Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:49542-49554. [PMID: 36314479 DOI: 10.1021/acsami.2c14448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Immune checkpoint blockade (ICB) utilizing programmed death ligand-1 (PD-L1) antibody is a promising treatment strategy in solid tumors. However, in fact, more than half of hepatocellular carcinoma (HCC) patients are unresponsive to PD-L1-based ICB treatment due to multiple immune evasion mechanisms such as the hyperactivation of inflammation pathway, excessive tumor-associated macrophages (TAMs) infiltration, and insufficient infiltration of T cells. Herein, an inflammation-regulated nanodrug was designed to codeliver NF-κB inhibitor curcumin and PD-L1 antibody to reprogram the tumor microenvironment (TME) and activate antitumor immunity. The nanodrug accumulated in TME by an enhanced permeability and retention effect, where it left antibody to block PD-L1 on the membrane of tumor cells and TAMs due to pH-responsiveness. Simultaneously, a new curcumin-encapsulated nanodrug was generated, which was easily absorbed by either tumor cells or TAMs to inhibit the nuclear factor kappa-B (NF-κB) signal and related immunosuppressive genes. The inflammation-regulated nanodrug possessed good biocompatibility. Simultaneously, it reprogrammed TME effectively and exhibited an effective anticancer effect in immunocompetent mice. Overall, this study provided a potent strategy to improve the efficiency of ICB-based treatment for HCC.
Collapse
Affiliation(s)
- TianCheng Wang
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - MinZhao Lin
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - JunJie Mao
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - LiRong Tian
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - HaiRun Gan
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - XinYan Hu
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - LeYe Yan
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - HaoYu Long
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - JianXun Cai
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - XiaoDi Zheng
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - YuDong Xiao
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha 410012, China
| | - Dan Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - XinTao Shuai
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - PengFei Pang
- Center of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| |
Collapse
|
39
|
Xu C, Xiao M, Li X, Xin L, Song J, Zhan Q, Wang C, Zhang Q, Yuan X, Tan Y, Fang C. Origin, activation, and targeted therapy of glioma-associated macrophages. Front Immunol 2022; 13:974996. [PMID: 36275720 PMCID: PMC9582955 DOI: 10.3389/fimmu.2022.974996] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/22/2022] [Indexed: 12/02/2022] Open
Abstract
The glioma tumor microenvironment plays a crucial role in the development, occurrence, and treatment of gliomas. Glioma-associated macrophages (GAMs) are the most widely infiltrated immune cells in the tumor microenvironment (TME) and one of the major cell populations that exert immune functions. GAMs typically originate from two cell types-brain-resident microglia (BRM) and bone marrow-derived monocytes (BMDM), depending on a variety of cytokines for recruitment and activation. GAMs mainly contain two functionally and morphologically distinct activation types- classically activated M1 macrophages (antitumor/immunostimulatory) and alternatively activated M2 macrophages (protumor/immunosuppressive). GAMs have been shown to affect multiple biological functions of gliomas, including promoting tumor growth and invasion, angiogenesis, energy metabolism, and treatment resistance. Both M1 and M2 macrophages are highly plastic and can polarize or interconvert under various malignant conditions. As the relationship between GAMs and gliomas has become more apparent, GAMs have long been one of the promising targets for glioma therapy, and many studies have demonstrated the therapeutic potential of this target. Here, we review the origin and activation of GAMs in gliomas, how they regulate tumor development and response to therapies, and current glioma therapeutic strategies targeting GAMs.
Collapse
Affiliation(s)
- Can Xu
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Menglin Xiao
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Xiang Li
- Hebei University School of Basic Medical Sciences, Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Lei Xin
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Jia Song
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
- Hebei University School of Basic Medical Sciences, Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Qi Zhan
- Tianjin Key Laboratory of Composite and Functional Materials, School of Material Science and Engineering, Tianjin University, Tianjin, China
| | - Changsheng Wang
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Qisong Zhang
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Xiaoye Yuan
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
- Hebei University School of Basic Medical Sciences, Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Yanli Tan
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
- Hebei University School of Basic Medical Sciences, Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
- *Correspondence: Chuan Fang, ; Yanli Tan,
| | - Chuan Fang
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
- *Correspondence: Chuan Fang, ; Yanli Tan,
| |
Collapse
|
40
|
Schaafsma E, Jiang C, Nguyen T, Zhu K, Cheng C. Microglia-Based Gene Expression Signature Highly Associated with Prognosis in Low-Grade Glioma. Cancers (Basel) 2022; 14:cancers14194802. [PMID: 36230719 PMCID: PMC9564275 DOI: 10.3390/cancers14194802] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Gliomas make up ~80% of malignant brain tumors in adults and are responsible for the majority of deaths from primary brain tumors. Consequently, a better understanding of the malignant features of the TME in glioma is pertinent. The aim of our study was to evaluate the expression of immune-related genes (IRGs) in glioma and their association with patient prognosis. We utilized several approaches to interrogate the glioma immune microenvironment. We found that immune genes are generally negatively associated with survival and that overall survival was significantly lower in those with a high level of microglia infiltration. The microglia abundance was significantly associated with common genomic aberrations. Lastly, we generated a 23-gene expression signature that is highly associated with patient prognosis, independent of clinical variables. These findings are relevant to investigators in the glioma field, those working in biomarker development, but also to individuals working on glioma therapeutics. Abstract Gliomas make up ~80% of malignant brain tumors in adults and are responsible for the majority of deaths from primary brain tumors. The glioma tumor microenvironment (TME) is a dynamic, heterogeneous mixture of extracellular matrix and malignant and non-malignant cells. Several ongoing clinical trials are evaluating the efficacy of therapies that target non-malignant cells, particularly immune cells. Consequently, a better understanding of the TME in glioma is pertinent. We utilized several gene expression datasets to evaluate the relationship between immune-related genes (IRGs) and patient prognosis. We generated microglia signatures using single-cell RNAseq data from human and mouse glioma cells to infer microglia abundance. Lastly, we built a LASSO Cox regression model that predicts patient survival. We found that 428 IRGs were negatively associated with survival in glioma patients. Overall survival was significantly lower in those with a high level of microglia infiltration. In addition, we also found that microglia abundance was significantly associated with several common genomic aberrations, including IDH2 and TP53 mutations. Furthermore, we found that patients with high risk scores had significantly worse overall survival than those with low risk scores in several independent datasets. Altogether, we characterized immune features predictive of overall survival in glioma and found that microglia abundance is negatively associated with survival. We developed a 23-gene risk score that can significantly stratify patients into low- and high-risk categories.
Collapse
Affiliation(s)
- Evelien Schaafsma
- Department of Microbiology and Immunology, Dartmouth College, Hanover, NH 03755, USA
| | - Chongming Jiang
- Department of Medicine, Baylor College of Medicine, Houston, TX 77054, USA
| | - Thinh Nguyen
- Department of Medicine, Baylor College of Medicine, Houston, TX 77054, USA
| | - Kenneth Zhu
- Medical School, UT Southwestern Medical Center, Dallas, TX 77054, USA
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX 77054, USA
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77054, USA
- L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77054, USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH 03755, USA
- Correspondence:
| |
Collapse
|
41
|
Yaping W, Zhe W, Zhuling C, Ruolei L, Pengyu F, Lili G, Cheng J, Bo Z, Liuyin L, Guangdong H, Yaoling W, Niuniu H, Rui L. The soldiers needed to be awakened: Tumor-infiltrating immune cells. Front Genet 2022; 13:988703. [PMID: 36246629 PMCID: PMC9558824 DOI: 10.3389/fgene.2022.988703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
In the tumor microenvironment, tumor-infiltrating immune cells (TIICs) are a key component. Different types of TIICs play distinct roles. CD8+ T cells and natural killer (NK) cells could secrete soluble factors to hinder tumor cell growth, whereas regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) release inhibitory factors to promote tumor growth and progression. In the meantime, a growing body of evidence illustrates that the balance between pro- and anti-tumor responses of TIICs is associated with the prognosis in the tumor microenvironment. Therefore, in order to boost anti-tumor response and improve the clinical outcome of tumor patients, a variety of anti-tumor strategies for targeting TIICs based on their respective functions have been developed and obtained good treatment benefits, including mainly immune checkpoint blockade (ICB), adoptive cell therapies (ACT), chimeric antigen receptor (CAR) T cells, and various monoclonal antibodies. In recent years, the tumor-specific features of immune cells are further investigated by various methods, such as using single-cell RNA sequencing (scRNA-seq), and the results indicate that these cells have diverse phenotypes in different types of tumors and emerge inconsistent therapeutic responses. Hence, we concluded the recent advances in tumor-infiltrating immune cells, including functions, prognostic values, and various immunotherapy strategies for each immune cell in different tumors.
Collapse
Affiliation(s)
- Wang Yaping
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Zhe
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Chu Zhuling
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
| | - Li Ruolei
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Fan Pengyu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Guo Lili
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ji Cheng
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhang Bo
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Liu Liuyin
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hou Guangdong
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Yaoling
- Department of Geriatrics, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hou Niuniu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| | - Ling Rui
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| |
Collapse
|
42
|
Wu Y, Yi M, Niu M, Mei Q, Wu K. Myeloid-derived suppressor cells: an emerging target for anticancer immunotherapy. Mol Cancer 2022; 21:184. [PMID: 36163047 PMCID: PMC9513992 DOI: 10.1186/s12943-022-01657-y] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/19/2022] [Indexed: 02/07/2023] Open
Abstract
The clinical responses observed following treatment with immune checkpoint inhibitors (ICIs) support immunotherapy as a potential anticancer treatment. However, a large proportion of patients cannot benefit from it due to resistance or relapse, which is most likely attributable to the multiple immunosuppressive cells in the tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs), a heterogeneous array of pathologically activated immature cells, are a chief component of immunosuppressive networks. These cells potently suppress T-cell activity and thus contribute to the immune escape of malignant tumors. New findings indicate that targeting MDSCs might be an alternative and promising target for immunotherapy, reshaping the immunosuppressive microenvironment and enhancing the efficacy of cancer immunotherapy. In this review, we focus primarily on the classification and inhibitory function of MDSCs and the crosstalk between MDSCs and other myeloid cells. We also briefly summarize the latest approaches to therapies targeting MDSCs.
Collapse
Affiliation(s)
- Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Ming Yi
- Department of Breast Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, 310003, China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China. .,Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
43
|
Abstract
The clinical responses observed following treatment with immune checkpoint inhibitors (ICIs) support immunotherapy as a potential anticancer treatment. However, a large proportion of patients cannot benefit from it due to resistance or relapse, which is most likely attributable to the multiple immunosuppressive cells in the tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs), a heterogeneous array of pathologically activated immature cells, are a chief component of immunosuppressive networks. These cells potently suppress T-cell activity and thus contribute to the immune escape of malignant tumors. New findings indicate that targeting MDSCs might be an alternative and promising target for immunotherapy, reshaping the immunosuppressive microenvironment and enhancing the efficacy of cancer immunotherapy. In this review, we focus primarily on the classification and inhibitory function of MDSCs and the crosstalk between MDSCs and other myeloid cells. We also briefly summarize the latest approaches to therapies targeting MDSCs.
Collapse
Affiliation(s)
- Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Ming Yi
- Department of Breast Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, 310003, China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
44
|
Franson A, McClellan BL, Varela ML, Comba A, Syed MF, Banerjee K, Zhu Z, Gonzalez N, Candolfi M, Lowenstein P, Castro MG. Development of immunotherapy for high-grade gliomas: Overcoming the immunosuppressive tumor microenvironment. Front Med (Lausanne) 2022; 9:966458. [PMID: 36186781 PMCID: PMC9515652 DOI: 10.3389/fmed.2022.966458] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023] Open
Abstract
The preclinical and clinical development of novel immunotherapies for the treatment of central nervous system (CNS) tumors is advancing at a rapid pace. High-grade gliomas (HGG) are aggressive tumors with poor prognoses in both adult and pediatric patients, and innovative and effective therapies are greatly needed. The use of cytotoxic chemotherapies has marginally improved survival in some HGG patient populations. Although several challenges exist for the successful development of immunotherapies for CNS tumors, recent insights into the genetic alterations that define the pathogenesis of HGG and their direct effects on the tumor microenvironment (TME) may allow for a more refined and targeted therapeutic approach. This review will focus on the TME in HGG, the genetic drivers frequently found in these tumors and their effect on the TME, the development of immunotherapy for HGG, and the practical challenges in clinical trials employing immunotherapy for HGG. Herein, we will discuss broadly the TME and immunotherapy development in HGG, with a specific focus on glioblastoma multiforme (GBM) as well as additional discussion in the context of the pediatric HGG diagnoses of diffuse midline glioma (DMG) and diffuse hemispheric glioma (DHG).
Collapse
Affiliation(s)
- Andrea Franson
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Brandon L. McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Immunology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria Luisa Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Mohammad Faisal Syed
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ziwen Zhu
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Nazareno Gonzalez
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pedro Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI, United States
- Biosciences Initiative in Brain Cancer, Biointerface Institute, University of Michigan, Ann Arbor, MI, United States
| | - Maria Graciela Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Biosciences Initiative in Brain Cancer, Biointerface Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
45
|
The function of myeloid-derived suppressor cells in COVID-19 lymphopenia. Int Immunopharmacol 2022; 112:109277. [PMID: 36206651 PMCID: PMC9513342 DOI: 10.1016/j.intimp.2022.109277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/02/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022]
Abstract
Coronavirus disease 2019 (COVID-19) has caused a global pandemic and presents a significant danger to public health. Lymphopenia is considered to be the defining characteristic of severe COVID-19, especially in elderly people. Lymphopenia has been suggested as a pivotal factor in disease severity. To minimize mortality in COVID-19 patients, it is essential to have a deeper understanding of the processes behind lymphocytopenia. Recently, myeloid-derived suppressor cells (MDSCs) have been confirmed as a key mediator of lymphopenia. MDSCs are characterized by their powerful capacity to suppress T cells and eventually contribute to the course of illness. Targeting these cells may improve the disease prognosis. In this article, we analyze the available research on MDSCs in lymphopenia and discuss their immunopathologic changes and prospective therapeutic targets in patients with COVID-19 lymphocytopenia.
Collapse
|
46
|
Pi C, Jing P, Li B, Feng Y, Xu L, Xie K, Huang T, Xu X, Gu H, Fang J. Reversing PD-1 Resistance in B16F10 Cells and Recovering Tumour Immunity Using a COX2 Inhibitor. Cancers (Basel) 2022; 14:cancers14174134. [PMID: 36077671 PMCID: PMC9455073 DOI: 10.3390/cancers14174134] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Immunotherapy is an effective method for tumour treatment. Anti-programmed cell death protein 1 (PD-1) and anti-programmed death-ligand 1 (PD-L1) monoclonal antibodies play a significant role in immunotherapy of most tumours; however, some patients develop drug resistance to PD-1/PD-L1 therapy. Cyclooxygenase-2 (COX2) is expressed in various solid tumours, and prostaglandin E2 (PGE2) drives the development of malignant tumours. We developed a drug-resistant B16F10 (B16F10-R) tumour mouse model through four rounds of selection in vivo. Subsequently, we investigated changes in PD-L1 expression and lymphocyte infiltration in B16F10-NR and B16F10-R tumours. Additionally, we explored the role of COX2 in acquired resistance to pembrolizumab, an anti-PD-1 treatment. Immune cell infiltration was significantly decreased in resistant tumours compared to B16F10-NR tumours; however, ptgs2 gene expression was significantly elevated in resistant tumours. Aspirin or celecoxib combined with pembrolizumab can effectively reverse tumour drug resistance. In addition, ptgs2 knockout or the use of the EP4 inhibitor E7046 abrogated drug resistance to anti-PD-1 treatment in B16F10-R tumour cells. Our study showed that inhibition of the COX2/PGE2/EP4 axis could increase the number of immune cells infiltrating the tumour microenvironment and recover drug-resistant tumour sensitivity to pembrolizumab. Thus, we highlight COX2 inhibition as a promising therapeutic target for drug-resistant tumours for future consideration.
Collapse
Affiliation(s)
- Chenyu Pi
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Ping Jing
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Bingyu Li
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- College of Medicine, Henan University of Science and Technology, Luoyang 471000, China
| | - Yan Feng
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Lijun Xu
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- College of Medicine, Henan University of Science and Technology, Luoyang 471000, China
| | - Kun Xie
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Tao Huang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaoqing Xu
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Hua Gu
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Correspondence: (H.G.); (J.F.); Tel.: +86-021-6598-2878 (H.G. & J.F.)
| | - Jianmin Fang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Biomedical Research Center, Suzhou 230031, China
- Shanghai Tongji Hospital, Shanghai 200065, China
- Correspondence: (H.G.); (J.F.); Tel.: +86-021-6598-2878 (H.G. & J.F.)
| |
Collapse
|
47
|
Papafragkos I, Grigoriou M, Boon L, Kloetgen A, Hatzioannou A, Verginis P. Ablation of NLRP3 inflammasome rewires MDSC function and promotes tumor regression. Front Immunol 2022; 13:889075. [PMID: 36032139 PMCID: PMC9407017 DOI: 10.3389/fimmu.2022.889075] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are myeloid precursors that exert potent immunosuppressive properties in cancer. Despite the extensive knowledge on mechanisms implicated in mobilization, recruitment, and function of MDSCs, their therapeutic targeting remains an unmet need in cancer immunotherapy, suggesting that unappreciated mechanisms of MDSC-mediated suppression exist. Herein, we demonstrate an important role of NLRP3 inflammasome in the functional properties of MDSCs in tumor-bearing hosts. Specifically, Nlrp3-deficient mice exhibited reduced tumor growth compared to wild-type animals and induction of robust anti-tumor immunity, accompanied by re-wiring of the MDSC compartment. Interestingly, both monocytic (M-MDSCs) and granulocytic (G-MDSCs) subsets from Nlrp3-/- mice displayed impaired suppressive activity and demonstrated significant transcriptomic alterations supporting the loss-of-function and associated with metabolic re-programming. Finally, therapeutic targeting of NLRP3 inhibited tumor development and re-programmed the MDSC compartment. These findings propose that targeting NLRP3 in MDSCs could overcome tumor-induced tolerance and may provide new checkpoints of cancer immunotherapy.
Collapse
Affiliation(s)
- Iosif Papafragkos
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, University of Crete Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
| | - Maria Grigoriou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | | | - Andreas Kloetgen
- Department of Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Aikaterini Hatzioannou
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Panayotis Verginis
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, University of Crete Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- *Correspondence: Panayotis Verginis,
| |
Collapse
|
48
|
Costa A, Thirant C, Kramdi A, Pierre-Eugène C, Louis-Brennetot C, Blanchard O, Surdez D, Gruel N, Lapouble E, Pierron G, Sitbon D, Brisse H, Gauthier A, Fréneaux P, Bohec M, Raynal V, Baulande S, Leclere R, Champenois G, Nicolas A, Meseure D, Bellini A, Marabelle A, Geoerger B, Mechta-Grigoriou F, Schleiermacher G, Menger L, Delattre O, Janoueix-Lerosey I. Single-cell transcriptomics reveals shared immunosuppressive landscapes of mouse and human neuroblastoma. J Immunother Cancer 2022; 10:jitc-2022-004807. [PMID: 36054452 PMCID: PMC9362821 DOI: 10.1136/jitc-2022-004807] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND High-risk neuroblastoma is a pediatric cancer with still a dismal prognosis, despite multimodal and intensive therapies. Tumor microenvironment represents a key component of the tumor ecosystem the complexity of which has to be accurately understood to define selective targeting opportunities, including immune-based therapies. METHODS We combined various approaches including single-cell transcriptomics to dissect the tumor microenvironment of both a transgenic mouse neuroblastoma model and a cohort of 10 biopsies from neuroblastoma patients, either at diagnosis or at relapse. Features of related cells were validated by multicolor flow cytometry and functional assays. RESULTS We show that the immune microenvironment of MYCN-driven mouse neuroblastoma is characterized by a low content of T cells, several phenotypes of macrophages and a population of cells expressing signatures of myeloid-derived suppressor cells (MDSCs) that are molecularly distinct from the various macrophage subsets. We document two cancer-associated fibroblasts (CAFs) subsets, one of which corresponding to CAF-S1, known to have immunosuppressive functions. Our data unravel a complex content in myeloid cells in patient tumors and further document a striking correspondence of the microenvironment populations between both mouse and human tumors. We show that mouse intratumor T cells exhibit increased expression of inhibitory receptors at the protein level. Consistently, T cells from patients are characterized by features of exhaustion, expressing inhibitory receptors and showing low expression of effector cytokines. We further functionally demonstrate that MDSCs isolated from mouse neuroblastoma have immunosuppressive properties, impairing the proliferation of T lymphocytes. CONCLUSIONS Our study demonstrates that neuroblastoma tumors have an immunocompromised microenvironment characterized by dysfunctional T cells and accumulation of immunosuppressive cells. Our work provides a new and precious data resource to better understand the neuroblastoma ecosystem and suggest novel therapeutic strategies, targeting both tumor cells and components of the microenvironment.
Collapse
Affiliation(s)
- Ana Costa
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Cécile Thirant
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Amira Kramdi
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Cécile Pierre-Eugène
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Caroline Louis-Brennetot
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Orphée Blanchard
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Didier Surdez
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Nadege Gruel
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,Department of Translational Research, Institut Curie, Paris, France
| | - Eve Lapouble
- Unité de Génétique Somatique, Institut Curie, Paris, France
| | - Gaëlle Pierron
- Unité de Génétique Somatique, Institut Curie, Paris, France
| | - Deborah Sitbon
- Unité de Génétique Somatique, Institut Curie, Paris, France
| | - Hervé Brisse
- Department of Imaging, PSL Research University, Institut Curie, Paris, France
| | | | - Paul Fréneaux
- Department of Biopathology, Institut Curie, Paris, France
| | - Mylène Bohec
- Genomics of Excellence (ICGex) Platform, Institut Curie, Paris, France
| | - Virginie Raynal
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,Genomics of Excellence (ICGex) Platform, Institut Curie, Paris, France
| | - Sylvain Baulande
- Genomics of Excellence (ICGex) Platform, Institut Curie, Paris, France
| | - Renaud Leclere
- Department of Biopathology, Institut Curie, Paris, France
| | | | - Andre Nicolas
- Department of Biopathology, Institut Curie, Paris, France
| | - Didier Meseure
- Department of Biopathology, Institut Curie, Paris, France
| | - Angela Bellini
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France.,Department of Translational Research, Institut Curie, Paris, France.,Laboratory Recherche Translationnelle en Oncologie Pédiatrique (RTOP), Laboratoire "Gilles Thomas", Institut Curie, Paris, France
| | - Aurelien Marabelle
- Inserm U1015 & CIC1428, Université Paris Saclay, Gustave Roussy, Villejuif, France
| | - Birgit Geoerger
- Inserm U1015, Department of Pediatric and Adolescent Oncology, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Fatima Mechta-Grigoriou
- Inserm U830, Equipe labelisée LNCC, Stress and Cancer Laboratory, PSL Research University, Institut Curie Research Centre, Paris, France
| | - Gudrun Schleiermacher
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France.,Department of Translational Research, Institut Curie, Paris, France.,Laboratory Recherche Translationnelle en Oncologie Pédiatrique (RTOP), Laboratoire "Gilles Thomas", Institut Curie, Paris, France
| | - Laurie Menger
- Inserm U932, PSL Research University, Institut Curie, Paris, France
| | - Olivier Delattre
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Isabelle Janoueix-Lerosey
- Inserm U830, Equipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Paris, France .,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| |
Collapse
|
49
|
Yan D, Li W, Liu Q, Yang K. Advances in Immune Microenvironment and Immunotherapy of Isocitrate Dehydrogenase Mutated Glioma. Front Immunol 2022; 13:914618. [PMID: 35769466 PMCID: PMC9234270 DOI: 10.3389/fimmu.2022.914618] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/16/2022] [Indexed: 11/21/2022] Open
Abstract
The tumor immune microenvironment and immunotherapy have become current important tumor research concerns. The unique immune microenvironment plays a crucial role in the malignant progression of isocitrate dehydrogenase (IDH) mutant gliomas. IDH mutations in glioma can inhibit tumor-associated immune system evasion of NK cell immune surveillance. Meanwhile, mutant IDH can inhibit classical and alternative complement pathways and directly inhibit T-cell responses by metabolizing isocitrate to D-2-Hydroxyglutaric acid (2-HG). IDH has shown clinically relevant efficacy as a potential target for immunotherapy. This article intends to summarize the research progress in the immunosuppressive microenvironment and immunotherapy of IDH-mutant glioma in recent years in an attempt to provide new ideas for the study of occurrence, progression, and treatment of IDH-mutant glioma.
Collapse
Affiliation(s)
- Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Weicheng Li
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qibing Liu
- Department of Pharmacology, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Qibing Liu, ; Kun Yang,
| | - Kun Yang
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Qibing Liu, ; Kun Yang,
| |
Collapse
|
50
|
Kim HJ, Ji YR, Lee YM. Crosstalk between angiogenesis and immune regulation in the tumor microenvironment. Arch Pharm Res 2022; 45:401-416. [PMID: 35759090 PMCID: PMC9250479 DOI: 10.1007/s12272-022-01389-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
Cancer creates a complex tumor microenvironment (TME) composed of immune cells, stromal cells, blood vessels, and various other cellular and extracellular elements. It is essential for the development of anti-cancer combination therapies to understand and overcome this high heterogeneity and complexity as well as the dynamic interactions between them within the TME. Recent treatment strategies incorporating immune-checkpoint inhibitors and anti-angiogenic agents have brought many changes and advances in clinical cancer treatment. However, there are still challenges for immune suppressive tumors, which are characterized by a lack of T cell infiltration and treatment resistance. In this review, we will investigate the crosstalk between immunity and angiogenesis in the TME. In addition, we will look at strategies designed to enhance anti-cancer immunity, to convert "immune suppressive tumors" into "immune activating tumors," and the mechanisms by which these strategies enhance effector immune cell infiltration.
Collapse
Affiliation(s)
- Hei Jung Kim
- Vessel-Organ Interaction Research Center, VOICE (MRC), Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Republic of Korea
| | - Young Rae Ji
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, USA
| | - You Mie Lee
- Vessel-Organ Interaction Research Center, VOICE (MRC), Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Republic of Korea.
- Department of Molecular Pathophysiology, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Republic of Korea.
| |
Collapse
|