1
|
Gao Y, Zou Y, Wu C, Tao J, Nie Z, Yan J, Wang P, Huang X. Comparative evaluation of immunomodulatory cytokines for oncolytic therapy based on a high-efficient platform for oHSV1 reconstruction. Virol J 2025; 22:133. [PMID: 40325455 PMCID: PMC12054163 DOI: 10.1186/s12985-025-02758-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: 03/20/2024] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) presents significant therapeutic challenges due to its immunosuppressive tumor microenvironment (TME). Oncolytic herpes simplex virus type 1 (oHSV1) offers dual mechanisms of tumor lysis and immune activation, yet the optimal cytokine payloads for TNBC remain undefined. METHODS We developed a CRISPR/Cas9-mediated platform for high-efficiency oHSV1 engineering, replacing the ICP47 locus with murine IFN-γ, GM-CSF, or IL-15Rα/IL-15 fusion protein (IL15Fu). Constructs were validated for cytokine secretion, MHC modulation, and cytotoxicity in 4T1 TNBC and a panel of human cancer cell lines. Antitumor efficacy and immune remodeling were evaluated in a syngeneic 4T1 model using RNA sequencing and flow cytometry. RESULTS The CRISPR platform achieved 62.5-71.4% homologous recombination efficiency, enabling rapid virus construction. In vitro, OV-IFNG exhibited upregulated MHC I/II expression and potent cytotoxicity, while OV-GMCSF attenuated oncolysis in subsets of breast cancer cell lines. In the 4T1 model, OV-IL15Fu modestly improved tumor control and extended survival without apparent toxicity, while OV-IFNG induced early mortality associated with systemic toxicity. Transcriptomic profiling revealed divergent immune modulation: OV-IL15Fu enriched T cell/NK cytotoxicity pathways, OV-IFNG amplified cytokine/chemokine signaling, and OV-GMCSF paradoxically enhanced myeloid recruitment while inhibiting MHC-II pathways. Flow cytometry confirmed functional differences in immune activation: OV-IL15Fu expanding cytotoxic lymphocytes (CD8⁺ T/NK cells), OV-IFNG preferentially promote Th1 polarization and innate immune activation, and OV-GMCSF failed to activate T cells despite myeloid infiltration. CONCLUSIONS Our findings underscore the need for rational cytokine selection in oHSV1-based immunotherapy. While IFN-γ increased immunogenic markers, its systemic toxicity and myeloid effects may limit benefit. GM-CSF exacerbated immune suppression in this context, whereas IL15Fu showed favorable immunostimulatory properties without detectable toxicity. These data support IL15Fu as a contextually promising payload for further evaluation in TNBC-targeted oncolytic virotherapy.
Collapse
Affiliation(s)
- Yingzheng Gao
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yufang Zou
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Changjing Wu
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Juan Tao
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zuqing Nie
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jinyuan Yan
- Kunming Medical University, Kunming, Yunnan, China
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Pengfei Wang
- Kunming Medical University, Kunming, Yunnan, China.
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Xinwei Huang
- Kunming Medical University, Kunming, Yunnan, China.
- Key Laboratory of The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
2
|
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
|
3
|
Dadgar N, Arunachalam AK, Hong H, Phoon YP, Arpi-Palacios JE, Uysal M, Wehrle CJ, Aucejo F, Ma WW, Melenhorst JJ. Advancing Cholangiocarcinoma Care: Insights and Innovations in T Cell Therapy. Cancers (Basel) 2024; 16:3232. [PMID: 39335203 PMCID: PMC11429565 DOI: 10.3390/cancers16183232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a rare and aggressive malignancy originating from the bile ducts, with poor prognosis and limited treatment options. Traditional therapies, such as surgery, chemotherapy, and radiation, have shown limited efficacy, especially in advanced cases. Recent advancements in immunotherapy, particularly T cell-based therapies like chimeric antigen receptor T (CAR T) cells, tumor-infiltrating lymphocytes (TILs), and T cell receptor (TCR)-based therapies, have opened new avenues for improving outcomes in CCA. This review provides a comprehensive overview of the current state of T cell therapies for CCA, focusing on CAR T cell therapy. It highlights key challenges, including the complex tumor microenvironment and immune evasion mechanisms, and the progress made in preclinical and clinical trials. The review also discusses ongoing clinical trials targeting specific CCA antigens, such as MUC1, EGFR, and CD133, and the evolving role of precision immunotherapy in enhancing treatment outcomes. Despite significant progress, further research is needed to optimize these therapies for solid tumors like CCA. By summarizing the most recent clinical results and future directions, this review underscores the promising potential of T cell therapies in revolutionizing CCA treatment.
Collapse
Affiliation(s)
- Neda Dadgar
- Cleveland Clinic Foundation, Enterprise Cancer Institute, Translational Hematology & Oncology Research, Cleveland, OH 44114, USA;
| | - Arun K. Arunachalam
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Hanna Hong
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Yee Peng Phoon
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Jorge E. Arpi-Palacios
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Melis Uysal
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Chase J. Wehrle
- Cleveland Clinic Foundation, Digestive Diseases & Surgery Institute, Cleveland, OH 44195, USA; (C.J.W.); (F.A.)
| | - Federico Aucejo
- Cleveland Clinic Foundation, Digestive Diseases & Surgery Institute, Cleveland, OH 44195, USA; (C.J.W.); (F.A.)
| | - Wen Wee Ma
- Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44106, USA;
| | - Jan Joseph Melenhorst
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| |
Collapse
|
4
|
Aliazis K, Yenyuwadee S, Phikulsod P, Boussiotis VA. Emergency myelopoiesis in solid cancers. Br J Haematol 2024; 205:798-811. [PMID: 39044285 DOI: 10.1111/bjh.19656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
Cells of the innate and adaptive immune systems are the progeny of haematopoietic stem and progenitor cells (HSPCs). During steady-state myelopoiesis, HSPC undergo differentiation and proliferation but are called to respond directly and acutely to various signals that lead to emergency myelopoiesis, including bone marrow ablation, infections, and sterile inflammation. There is extensive evidence that many solid tumours have the potential to secrete classical myelopoiesis-promoting growth factors and other products able to mimic emergency haematopoiesis, and to aberrantly re-direct myeloid cell development into immunosuppressive cells with tumour promoting properties. Here, we summarize the current literature regarding the effects of solid cancers on HSPCs function and discuss how these effects might shape antitumour responses via a mechanism initiated at a site distal from the tumour microenvironment.
Collapse
Affiliation(s)
- Konstantinos Aliazis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Sasitorn Yenyuwadee
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ployploen Phikulsod
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vassiliki A Boussiotis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Calderon-Espinosa E, De Ridder K, Benoot T, Jansen Y, Vanhonacker D, Heestermans R, De Becker A, Van Riet I, Decoster L, Goyvaerts C. The crosstalk between lung cancer and the bone marrow niche fuels emergency myelopoiesis. Front Immunol 2024; 15:1397469. [PMID: 39148724 PMCID: PMC11324509 DOI: 10.3389/fimmu.2024.1397469] [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: 03/07/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
Modest response rates to immunotherapy observed in advanced lung cancer patients underscore the need to identify reliable biomarkers and targets, enhancing both treatment decision-making and efficacy. Factors such as PD-L1 expression, tumor mutation burden, and a 'hot' tumor microenvironment with heightened effector T cell infiltration have consistently been associated with positive responses. In contrast, the predictive role of the abundantly present tumor-infiltrating myeloid cell (TIMs) fraction remains somewhat uncertain, partly explained by their towering variety in terms of ontogeny, phenotype, location, and function. Nevertheless, numerous preclinical and clinical studies established a clear link between lung cancer progression and alterations in intra- and extramedullary hematopoiesis, leading to emergency myelopoiesis at the expense of megakaryocyte/erythroid and lymphoid differentiation. These observations affirm that a continuous crosstalk between solid cancers such as lung cancer and the bone marrow niche (BMN) must take place. However, the BMN, encompassing hematopoietic stem and progenitor cells, differentiated immune and stromal cells, remains inadequately explored in solid cancer patients. Subsequently, no clear consensus has been reached on the exact breadth of tumor installed hematopoiesis perturbing cues nor their predictive power for immunotherapy. As the current era of single-cell omics is reshaping our understanding of the hematopoietic process and the subcluster landscape of lung TIMs, we aim to present an updated overview of the hierarchical differentiation process of TIMs within the BMN of solid cancer bearing subjects. Our comprehensive overview underscores that lung cancer should be regarded as a systemic disease in which the cues governing the lung tumor-BMN crosstalk might bolster the definition of new biomarkers and druggable targets, potentially mitigating the high attrition rate of leading immunotherapies for NSCLC.
Collapse
Affiliation(s)
- Evelyn Calderon-Espinosa
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
- Laboratory for Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Chemistry, University of Warwick, Warwick, United Kingdom
| | - Kirsten De Ridder
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
- Laboratory for Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Benoot
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
- Laboratory for Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | - Yanina Jansen
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Domien Vanhonacker
- Department of Anesthesiology, Perioperative and Pain Medicine, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Robbe Heestermans
- Department of Hematology, Team Hematology and Immunology (HEIM), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Ann De Becker
- Department of Hematology, Team Hematology and Immunology (HEIM), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Ivan Van Riet
- Department of Hematology, Team Hematology and Immunology (HEIM), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Lore Decoster
- Department of Medical Oncology, Team Laboratory for Medical and Molecular Oncology (LMMO), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
- Laboratory for Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
6
|
Nepal MR, Shah S, Kang KT. Dual roles of myeloid-derived suppressor cells in various diseases: a review. Arch Pharm Res 2024; 47:597-616. [PMID: 39008186 DOI: 10.1007/s12272-024-01504-2] [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: 10/09/2023] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that originate from bone marrow stem cells. In pathological conditions, such as autoimmune disorders, allergies, infections, and cancer, normal myelopoiesis is altered to facilitate the formation of MDSCs. MDSCs were first shown to promote cancer initiation and progression by immunosuppression with the assistance of various chemokines and cytokines. Recently, various studies have demonstrated that MDSCs play two distinct roles depending on the physiological and pathological conditions. MDSCs have protective roles in autoimmune disorders (such as uveoretinitis, multiple sclerosis, rheumatoid arthritis, ankylosing spondylitis, type 1 diabetes, autoimmune hepatitis, inflammatory bowel disease, alopecia areata, and systemic lupus erythematosus), allergies, and organ transplantation. However, they play negative roles in infections and various cancers. Several immunosuppressive functions and mechanisms of MDSCs have been determined in different disease conditions. This review comprehensively discusses the associations between MDSCs and various pathological conditions and briefly describes therapeutic approaches.
Collapse
Affiliation(s)
- Mahesh Raj Nepal
- College of Pharmacy, Duksung Women's University, Seoul, South Korea
- Duksung Innovative Drug Center, Duksung Women's University, Seoul, South Korea
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Sajita Shah
- College of Pharmacy, Duksung Women's University, Seoul, South Korea
- Duksung Innovative Drug Center, Duksung Women's University, Seoul, South Korea
- The Comprehensive Cancer Center, Department of Radiation Oncology, Ohio State University, Columbus, OH, USA
| | - Kyu-Tae Kang
- College of Pharmacy, Duksung Women's University, Seoul, South Korea.
- Duksung Innovative Drug Center, Duksung Women's University, Seoul, South Korea.
| |
Collapse
|
7
|
Lian GY, Wang QM, Mak TSK, Huang XR, Yu XQ, Lan HY. Disrupting Smad3 potentiates immunostimulatory function of NK cells against lung carcinoma by promoting GM-CSF production. Cell Mol Life Sci 2024; 81:262. [PMID: 38878186 PMCID: PMC11335298 DOI: 10.1007/s00018-024-05290-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 04/28/2024] [Accepted: 05/23/2024] [Indexed: 06/29/2024]
Abstract
Through Smad3-dependent signalings, transforming growth factor-β (TGF-β) suppresses the development, maturation, cytokine productions and cytolytic functions of NK cells in cancer. Silencing Smad3 remarkably restores the cytotoxicity of NK-92 against cancer in TGF-β-rich microenvironment, but its effects on the immunoregulatory functions of NK cells remain obscure. In this study, we identified Smad3 functioned as a transcriptional repressor for CSF2 (GM-CSF) in NK cells. Therefore, disrupting Smad3 largely mitigated TGF-β-mediated suppression on GM-CSF production by NK cells. Furthermore, silencing GM-CSF in Smad3 knockout NK cells substantially impaired their anti-lung carcinoma effects. In-depth study demonstrated that NK-derived GM-CSF strengthened T cell immune responses by stimulating dendritic cell differentiation and M1 macrophage polarization. Meanwhile, NK-derived GM-CSF promoted the survival of neutrophils, which in turn facilitated the terminal maturation of NK cells, and subsequently boosted NK-cell mediated cytotoxicity against lung carcinoma. Thus, Smad3-silenced NK-92 (NK-92-S3KD) may serve as a promising immunoadjuvant therapy with clinical translational value given its robust cytotoxicity against malignant cells and immunostimulatory functions to reinforce the therapeutic effects of other immunotherapies.
Collapse
Affiliation(s)
- Guang-Yu Lian
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Departments of Pathology and Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qing-Ming Wang
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Thomas Shiu-Kwong Mak
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao-Ru Huang
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Departments of Pathology and Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xue-Qing Yu
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Departments of Pathology and Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Hui-Yao Lan
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Departments of Pathology and Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
8
|
Wang MJ, Xia Y, Gao QL. DNA Damage-driven Inflammatory Cytokines: Reprogramming of Tumor Immune Microenvironment and Application of Oncotherapy. Curr Med Sci 2024; 44:261-272. [PMID: 38561595 DOI: 10.1007/s11596-024-2859-1] [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: 12/15/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
DNA damage occurs across tumorigenesis and tumor development. Tumor intrinsic DNA damage can not only increase the risk of mutations responsible for tumor generation but also initiate a cellular stress response to orchestrate the tumor immune microenvironment (TIME) and dominate tumor progression. Accumulating evidence documents that multiple signaling pathways, including cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) and ataxia telangiectasia-mutated protein/ataxia telangiectasia and Rad3-related protein (ATM/ATR), are activated downstream of DNA damage and they are associated with the secretion of diverse cytokines. These cytokines possess multifaced functions in the anti-tumor immune response. Thus, it is necessary to deeply interpret the complex TIME reshaped by damaged DNA and tumor-derived cytokines, critical for the development of effective tumor therapies. This manuscript comprehensively reviews the relationship between the DNA damage response and related cytokines in tumors and depicts the dual immunoregulatory roles of these cytokines. We also summarize clinical trials targeting signaling pathways and cytokines associated with DNA damage and provide future perspectives on emerging technologies.
Collapse
Affiliation(s)
- Meng-Jie Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Xia
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Qing-Lei Gao
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
9
|
Kuziel G, Moore BN, Haugstad GP, Xiong Y, Williams AE, Arendt LM. Alterations in the mammary gland and tumor microenvironment of formerly obese mice. BMC Cancer 2023; 23:1183. [PMID: 38041006 PMCID: PMC10693119 DOI: 10.1186/s12885-023-11688-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Obesity is a risk factor for breast cancer, and women with obesity that develop breast cancer have a worsened prognosis. Within the mammary gland, obesity causes chronic, macrophage-driven inflammation and adipose tissue fibrosis. Weight loss is a recommended intervention to resolve obesity, but the impact of weight loss on the mammary gland microenvironment and in tumors has not been well identified. METHODS To examine the effects of weight loss following obesity, mice were fed a high-fat diet for 16 weeks to induce obesity, then switched to a low-fat diet for 6 weeks. We examined changes in immune cells, including fibrocytes, which are myeloid lineage cells that have attributes of both macrophages and myofibroblasts, and collagen deposition within the mammary glands of non-tumor-bearing mice and within the tumors of mice that were transplanted with estrogen receptor alpha positive TC2 tumor cells. RESULTS In formerly obese mice, we observed reduced numbers of crown-like structures and fibrocytes in mammary glands, while collagen deposition was not resolved with weight loss. Following transplant of TC2 tumor cells into the mammary glands of lean, obese, and formerly obese mice, diminished collagen deposition and cancer-associated fibroblasts were observed in tumors from formerly obese mice compared to obese mice. Within tumors of obese mice, increased myeloid-derived suppressor cells and diminished CD8+ T cells were identified, while the microenvironment of tumors of formerly obese mice were more similar to tumors from lean mice. When TC2 tumor cells were mixed with CD11b+CD34+ myeloid progenitor cells, which are the cells of origin for fibrocytes, and transplanted into mammary glands of lean and obese mice, collagen deposition within the tumors of both lean and obese was significantly greater than when tumor cells were mixed with CD11b+CD34- monocytes or total CD45+ immune cells. CONCLUSIONS Overall, these studies demonstrate that weight loss resolved some of the microenvironmental conditions within the mammary gland that may contribute to tumor progression. Additionally, fibrocytes may contribute to early collagen deposition in mammary tumors of obese mice leading to the growth of desmoplastic tumors.
Collapse
Affiliation(s)
- Genevra Kuziel
- Cancer Biology Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Brittney N Moore
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Grace P Haugstad
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Yue Xiong
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Abbey E Williams
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Lisa M Arendt
- Cancer Biology Program, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- School of Veterinary Medicine, 2015 Linden Drive Rm 4354A, Madison, WI, 53706, USA.
| |
Collapse
|
10
|
Blanco E, Escors D, Kochan G. Assessment of myeloid-derived suppressor cell differentiation ex vivo. Methods Cell Biol 2023; 184:85-96. [PMID: 38555160 DOI: 10.1016/bs.mcb.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) are major promoters of progression and metastasis in cancer. MDSCs inhibit the anti-tumor immune response through multiple mechanisms. The main MDSC functions in cancer are related to the inactivation of T cells and the establishment of an immunosuppressive tumor microenvironment (TME) through the production of pro-inflammatory cytokines, among other mechanisms. MDSCs are phenotypically similar to conventional myeloid cells, so their identification is challenging. Moreover, they infiltrate the tumors in limited numbers, and their purification from within the tumors is technically difficult and makes their study a challenge. Therefore, several ex vivo differentiation methods have been established. Our differentiation method leads to MDSCs that closely model tumor-infiltrating counterparts. In this protocol, MDSCs are differentiated from bone marrow precursors by incubation in differentiation medium produced by murine tumor cell lines engineered to constitutively express granulocyte-monocyte colony stimulating factor (GM-CSF). These ex vivo-generated MDSC subsets show high fidelity compared to their natural tumor-infiltrated counterparts. Moreover, the high yields of purification from these ex vivo differentiated MDSC enable their use for validation of new treatments in high-throughput assays. In this chapter we describe the engineering of a stable cell line overexpressing GM-CSF, followed by production and collection of conditioned media supporting MDSC differentiation. Finally, we detail the isolation procedure of bone marrow cells and the specific MDSC differentiation protocol.
Collapse
Affiliation(s)
- Ester Blanco
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain.
| | - David Escors
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Grazyna Kochan
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
11
|
Elanany MM, Mostafa D, Hamdy NM. Remodeled tumor immune microenvironment (TIME) parade via natural killer cells reprogramming in breast cancer. Life Sci 2023; 330:121997. [PMID: 37536617 DOI: 10.1016/j.lfs.2023.121997] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Breast cancer (BC) is the main cause of cancer-related mortality among women globally. Despite substantial advances in the identification and management of primary tumors, traditional therapies including surgery, chemotherapy, and radiation cannot completely eliminate the danger of relapse and metastatic illness. Metastasis is controlled by microenvironmental and systemic mechanisms, including immunosurveillance. This led to the evolvement of immunotherapies that has gained much attention in the recent years for cancer treatment directed to the innate immune system. The long forgotten innate immune cells known as natural killer (NK) cells have emerged as novel targets for more effective therapeutics for BC. Normally, NK cells has the capacity to identify and eradicate tumor cells either directly or by releasing cytotoxic granules, chemokines and proinflammatory cytokines. Yet, NK cells are exposed to inhibitory signals by cancer cells, which causes them to become dysfunctional in the immunosuppressive tumor microenvironment (TME) in BC, supporting tumor escape and spread. Potential mechanisms of NK cell dysfunction in BC metastasis have been recently identified. Understanding these immunologic pathways driving BC metastasis will lead to improvements in the current immunotherapeutic strategies. In the current review, we highlight how BC evades immunosurveillance by rendering NK cells dysfunctional and we shed the light on novel NK cell- directed therapies.
Collapse
Affiliation(s)
- Mona M Elanany
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Dina Mostafa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| | - Nadia M Hamdy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| |
Collapse
|
12
|
Ren R, Xiong C, Ma R, Wang Y, Yue T, Yu J, Shao B. The recent progress of myeloid-derived suppressor cell and its targeted therapies in cancers. MedComm (Beijing) 2023; 4:e323. [PMID: 37547175 PMCID: PMC10397484 DOI: 10.1002/mco2.323] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 08/08/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are an immature group of myeloid-derived cells generated from myeloid cell precursors in the bone marrow. MDSCs appear almost exclusively in pathological conditions, such as tumor progression and various inflammatory diseases. The leading function of MDSCs is their immunosuppressive ability, which plays a crucial role in tumor progression and metastasis through their immunosuppressive effects. Since MDSCs have specific molecular features, and only a tiny amount exists in physiological conditions, MDSC-targeted therapy has become a promising research direction for tumor treatment with minimal side effects. In this review, we briefly introduce the classification, generation and maturation process, and features of MDSCs, and detail their functions under various circumstances. The present review specifically demonstrates the environmental specificity of MDSCs, highlighting the differences between MDSCs from cancer and healthy individuals, as well as tumor-infiltrating MDSCs and circulating MDSCs. Then, we further describe recent advances in MDSC-targeted therapies. The existing and potential targeted drugs are divided into three categories, monoclonal antibodies, small-molecular inhibitors, and peptides. Their targeting mechanisms and characteristics have been summarized respectively. We believe that a comprehensive in-depth understanding of MDSC-targeted therapy could provide more possibilities for the treatment of cancer.
Collapse
Affiliation(s)
- Ruiyang Ren
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Chenyi Xiong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Runyu Ma
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Yixuan Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Tianyang Yue
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Jiayun Yu
- Department of RadiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Bin Shao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
- State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
13
|
Rodriguez BL, Chen L, Li Y, Miao S, Peng DH, Fradette JJ, Diao L, Konen JM, Alvarez FRR, Solis LM, Yi X, Padhye A, Gibson LA, Ochieng JK, Zhou X, Wang J, Gibbons DL. Targeting immunosuppressive Ly6C+ classical monocytes reverses anti-PD-1/CTLA-4 immunotherapy resistance. Front Immunol 2023; 14:1161869. [PMID: 37449205 PMCID: PMC10336223 DOI: 10.3389/fimmu.2023.1161869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Despite significant clinical advancement with the use of immune checkpoint blockade (ICB) in non-small cell lung cancer (NSCLC) there are still a major subset of patients that develop adaptive/acquired resistance. Understanding resistance mechanisms to ICB is critical to developing new therapeutic strategies and improving patient survival. The dynamic nature of the tumor microenvironment and the mutational load driving tumor immunogenicity limit the efficacy to ICB. Recent studies indicate that myeloid cells are drivers of ICB resistance. In this study we sought to understand which immune cells were contributing to resistance and if we could modify them in a way to improve response to ICB therapy. Results Our results show that combination anti-PD-1/CTLA-4 produces an initial antitumor effect with evidence of an activated immune response. Upon extended treatment with anti-PD-1/CTLA-4 acquired resistance developed with an increase of the immunosuppressive populations, including T-regulatory cells, neutrophils and monocytes. Addition of anti-Ly6C blocking antibody to anti-PD-1/CTLA-4 was capable of completely reversing treatment resistance and restoring CD8 T cell activity in multiple KP lung cancer models and in the autochthonous lung cancer KrasLSL-G12D/p53fl/fl model. We found that there were higher classical Ly6C+ monocytes in anti-PD-1/CTLA-4 combination resistant tumors. B7 blockade illustrated the importance of dendritic cells for treatment efficacy of anti-Ly6C/PD-1/CTLA-4. We further determined that classical Ly6C+ monocytes in anti-PD-1/CTLA-4 resistant tumors are trafficked into the tumor via IFN-γ and the CCL2-CCR2 axis. Mechanistically we found that classical monocytes from ICB resistant tumors were unable to differentiate into antigen presenting cells and instead differentiated into immunosuppressive M2 macrophages or myeloid-derived suppressor cells (MDSC). Classical Ly6C+ monocytes from ICB resistant tumors had a decrease in both Flt3 and PU.1 expression that prevented differentiation into dendritic cells/macrophages. Conclusions Therapeutically we found that addition of anti-Ly6C to the combination of anti-PD-1/CTLA-4 was capable of complete tumor eradication. Classical Ly6C+ monocytes differentiate into immunosuppressive cells, while blockade of classical monocytes drives dendritic cell differentiation/maturation to reinvigorate the anti-tumor T cell response. These findings support that immunotherapy resistance is associated with infiltrating monocytes and that controlling the differentiation process of monocytes can enhance the therapeutic potential of ICB.
Collapse
Affiliation(s)
- B. Leticia Rodriguez
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Limo Chen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yanli Li
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Shucheng Miao
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- United of Texas (UT) Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - David H. Peng
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jared J. Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lixia Diao
- Department Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jessica M. Konen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Frank R. Rojas Alvarez
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Luisa M. Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xiaohui Yi
- Bellicum Pharmaceuticals, Inc., Houston, TX, United States
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aparna Padhye
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- United of Texas (UT) Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Laura A. Gibson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joshua K. Ochieng
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xiaofei Zhou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jing Wang
- Department Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
14
|
Kuziel G, Moore BN, Haugstad GP, Xiong Y, Williams AE, Arendt LM. Alterations in the Mammary Gland and Tumor Microenvironment of Formerly Obese Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.14.545000. [PMID: 37398468 PMCID: PMC10312750 DOI: 10.1101/2023.06.14.545000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Obesity is a risk factor for breast cancer, and women with obesity that develop breast cancer have a worsened prognosis. Within the mammary gland, obesity causes chronic, macrophage-driven inflammation and adipose tissue fibrosis. To examine the impact of weight loss on the mammary microenvironment, mice were fed high-fat diet to induce obesity, then switched to a low-fat diet. In formerly obese mice, we observed reduced numbers of crown-like structures and fibrocytes in mammary glands, while collagen deposition was not resolved with weight loss. Following transplant of TC2 tumor cells into the mammary glands of lean, obese, and formerly obese mice, diminished collagen deposition and cancer-associated fibroblasts were observed in tumors from formerly obese mice compared to obese mice. When TC2 tumor cells were mixed with CD11b+CD34+ myeloid progenitor cells, collagen deposition within the tumors was significantly greater compared to when tumor cells were mixed with CD11b+CD34- monocytes, suggesting that fibrocytes contribute to early collagen deposition in mammary tumors of obese mice. Overall, these studies show that weight loss resolved some of the microenvironmental conditions within the mammary gland that may contribute to tumor progression.
Collapse
Affiliation(s)
- Genevra Kuziel
- Cancer Biology Program, University of Wisconsin-Madison, Madison WI 53705, U.S.A
| | - Brittney N. Moore
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison WI 53706, U.S.A
| | - Grace P. Haugstad
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison WI 53706, U.S.A
| | - Yue Xiong
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison WI 53706, U.S.A
| | - Abbey E. Williams
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison WI 53706, U.S.A
| | - Lisa M. Arendt
- Cancer Biology Program, University of Wisconsin-Madison, Madison WI 53705, U.S.A
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison WI 53706, U.S.A
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison WI 53706, U.S.A
| |
Collapse
|
15
|
Andrés CMC, Pérez de la Lastra JM, Juan CA, Plou FJ, Pérez-Lebeña E. Myeloid-Derived Suppressor Cells in Cancer and COVID-19 as Associated with Oxidative Stress. Vaccines (Basel) 2023; 11:218. [PMID: 36851096 PMCID: PMC9966263 DOI: 10.3390/vaccines11020218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Myeloid-derived suppressor cells MDSCs are a heterogeneous population of cells that expand beyond their physiological regulation during pathologies such as cancer, inflammation, bacterial, and viral infections. Their key feature is their remarkable ability to suppress T cell and natural killer NK cell responses. Certain risk factors for severe COVID-19 disease, such as obesity and diabetes, are associated with oxidative stress. The resulting inflammation and oxidative stress can negatively impact the host. Similarly, cancer cells exhibit a sustained increase in intrinsic ROS generation that maintains the oncogenic phenotype and drives tumor progression. By disrupting endoplasmic reticulum calcium channels, intracellular ROS accumulation can disrupt protein folding and ultimately lead to proteostasis failure. In cancer and COVID-19, MDSCs consist of the same two subtypes (PMN-MSDC and M-MDSC). While the main role of polymorphonuclear MDSCs is to dampen the response of T cells and NK killer cells, they also produce reactive oxygen species ROS and reactive nitrogen species RNS. We here review the origin of MDSCs, their expansion mechanisms, and their suppressive functions in the context of cancer and COVID-19 associated with the presence of superoxide anion •O2- and reactive oxygen species ROS.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén 7, 47011 Valladolid, Spain
| | - Celia Andrés Juan
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | - Francisco J. Plou
- Institute of Catalysis and Petrochemistry, CSIC-Spanish Research Council, 28049 Madrid, Spain
| | | |
Collapse
|
16
|
Abstract
BACKGROUND Esophageal cancer (EC) originates in the setting of chronic inflammation. Although previous studies have sought to understand the role of inflammatory signaling in EC, the effect of these immunologic changes on patient outcomes remains understudied. This study's objective was to identify relationships between cytokine levels and prognosis in a mixed cohort of esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC) patients. STUDY DESIGN A total of 37 serum cytokines were profiled at the time of resection using multiplex ELISA in 47 patients (42 esophageal adenocarcinoma, 5 esophageal squamous cell carcinoma). Cytokine levels were median-binarized and assessed using Cox regression models. Findings were validated at the RNA level using The Cancer Genome Atlas EC cohort (81 esophageal adenocarcinoma, 81 esophageal squamous cell carcinoma). RESULTS Univariable analysis revealed high serum interleukin 4 (IL4) and granulocyte-macrophage colony-stimulating factor (GMCSF) were negatively associated with overall survival (p = 0.046, p = 0.040). Multivariable analysis determined both high serum IL4 or high serum GMCSF were negatively associated with survival independent of important clinical factors (hazard ratio [HR] 7.55, p < 0.001; HR 5.24, p = 0.001). These findings were validated at the RNA level in The Cancer Genome Atlas EC cohort, where multivariable analysis identified high IL4 expression, high CSF2 expression (encodes GMCSF), and advanced pathologic stage as independent negative predictors of survival when controlled for clinical factors (HR 2.35, p = 0.012; HR 1.97, p = 0.040). CONCLUSIONS These results show that high IL4/GMCSF levels are negatively associated with survival in EC. These relationships are independent of pathologic stage and are identified across modalities, histologic subtypes, and the presence/absence of neoadjuvant therapy.
Collapse
|
17
|
Pre-Existing HSV-1 Immunity Enhances Anticancer Efficacy of a Novel Immune-Stimulating Oncolytic Virus. Viruses 2022; 14:v14112327. [PMID: 36366425 PMCID: PMC9693100 DOI: 10.3390/v14112327] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/03/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Oncolytic viruses (OVs) can specifically replicate in the host and cause cancer cell lysis while inducing an antitumor immune response. The aim of this study is to investigate the impact of either pre-existing immunity against herpes simplex virus type-1 (HSV-1) or multicycle treatment with OVs on anticancer efficacy of VG161, an HSV-1 OV in phase 2 clinical trial. VG161 efficacy was tested in CT26 mouse models by comparing the efficacy and immune response in naïve mice or in mice that were immunized with VG161. Moreover, VG161 efficacy in HLA-matched CD34+ humanized intrahepatic cholangiocarcinoma (ICC) patient-derived xenograft (PDX) models was also tested in multicycle treatment and was compared to standard chemotherapy for this type of cancer (gemcitabine). The HSV-1-immunized mice significantly inhibited tumor growth in VG161-treated mice compared to control naïve treated mice. RNA expression profiling and ELISPOT analyses indicated changes in the tumor's immune profile in the immunized and treated group compared to naïve and treated mice, as well as enhanced T cell function depicted by higher numbers of tumor specific lymphocytes, which was enhanced by immunization. In the ICC PDX model, repeated treatment of VG161 with 2 or 3 cycles seemed to increase the anticancer efficacy of VG161. In conclusion, the anticancer efficacy of VG161 can be enhanced by pre-immunization with HSV-1 and multicycle administration when the virus is given intratumorally, indicating that pre-existing antiviral immunity might enhance OV-induced antitumor immunity. Our results suggest potential clinical benefits of HSV-1-based OV therapy in HSV-1-seropositive patients and multicycle administration of VG161 for long-term maintenance treatment.
Collapse
|
18
|
van Geffen C, Heiss C, Deißler A, Kolahian S. Pharmacological modulation of myeloid-derived suppressor cells to dampen inflammation. Front Immunol 2022; 13:933847. [PMID: 36110844 PMCID: PMC9468781 DOI: 10.3389/fimmu.2022.933847] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous cell population with potent suppressive and regulative properties. MDSCs’ strong immunosuppressive potential creates new possibilities to treat chronic inflammation and autoimmune diseases or induce tolerance towards transplantation. Here, we summarize and critically discuss different pharmacological approaches which modulate the generation, activation, and recruitment of MDSCs in vitro and in vivo, and their potential role in future immunosuppressive therapy.
Collapse
|
19
|
Chen C, Huang R, Zhou J, Guo L, Xiang S. Formation of pre-metastatic bone niche in prostate cancer and regulation of traditional chinese medicine. Front Pharmacol 2022; 13:897942. [PMID: 36059977 PMCID: PMC9428453 DOI: 10.3389/fphar.2022.897942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Prostate cancer with bone metastasis has a high cancer-specific mortality. Thus, it is essential to delineate the mechanism of bone metastasis. Pre-metastatic niche (PMN) is a concept in tumor metastasis, which is characterized by tumor-secreted factors, reprogramming of stromal cells, and immunosuppression by myeloid-derived suppressor cells (MDSC), which is induced by bone marrow-derived cells (BMDC) in the target organ. However, PMN does not explain the predilection of prostate cancer towards bone metastasis. In this review, we discuss the initiation of bone metastasis of prostate cancer from the perspective of PMN and tumor microenvironment in a step-wise manner. Furthermore, we present a new concept called pre-metastatic bone niche, featuring inherent BMDC, to interpret bone metastasis. Moreover, we illustrate the regulation of traditional Chinese medicine on PMN.
Collapse
|
20
|
Kumar A, Taghi Khani A, Sanchez Ortiz A, Swaminathan S. GM-CSF: A Double-Edged Sword in Cancer Immunotherapy. Front Immunol 2022; 13:901277. [PMID: 35865534 PMCID: PMC9294178 DOI: 10.3389/fimmu.2022.901277] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/06/2022] [Indexed: 12/23/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine that drives the generation of myeloid cell subsets including neutrophils, monocytes, macrophages, and dendritic cells in response to stress, infections, and cancers. By modulating the functions of innate immune cells that serve as a bridge to activate adaptive immune responses, GM-CSF globally impacts host immune surveillance under pathologic conditions. As with other soluble mediators of immunity, too much or too little GM-CSF has been found to promote cancer aggressiveness. While too little GM-CSF prevents the appropriate production of innate immune cells and subsequent activation of adaptive anti-cancer immune responses, too much of GM-CSF can exhaust immune cells and promote cancer growth. The consequences of GM-CSF signaling in cancer progression are a function of the levels of GM-CSF, the cancer type, and the tumor microenvironment. In this review, we first discuss the secretion of GM-CSF, signaling downstream of the GM-CSF receptor, and GM-CSF’s role in modulating myeloid cell homeostasis. We then outline GM-CSF’s anti-tumorigenic and pro-tumorigenic effects both on the malignant cells and on the non-malignant immune and other cells in the tumor microenvironment. We provide examples of current clinical and preclinical strategies that harness GM-CSF’s anti-cancer potential while minimizing its deleterious effects. We describe the challenges in achieving the Goldilocks effect during administration of GM-CSF-based therapies to patients with cancer. Finally, we provide insights into how technologies that map the immune microenvironment spatially and temporally may be leveraged to intelligently harness GM-CSF for treatment of malignancies.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Adeleh Taghi Khani
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Ashly Sanchez Ortiz
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Srividya Swaminathan
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
- Department of Hematological Malignancies, Beckman Research Institute of City of Hope, Monrovia, CA, United States
- *Correspondence: Srividya Swaminathan,
| |
Collapse
|
21
|
Li T, Bou-Dargham MJ, Fultang N, Li X, Pear WS, Sun H, Chen YH. c-Rel-dependent monocytes are potent immune suppressor cells in cancer. J Leukoc Biol 2022; 112:845-859. [PMID: 35694784 PMCID: PMC9530019 DOI: 10.1002/jlb.1ma0422-518rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 04/25/2022] [Indexed: 11/08/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of leukocytes that are important for tumorigenesis and tumor immunotherapy. They comprise up to 10% of leukocytes in the blood of tumor patients and their depletion may be required for successful tumor immunotherapy. However, the identity of MDSCs remains obscure, primarily due to their heterogeneity and lack of a known lineage-specific transcription factor specifying their differentiation. Using single-cell transcriptomics and gene knockout approaches, we now describe a subset of murine and human myeloid suppressor cells, named rel-dependent monocytes (rMos), which are programmed by the transcription factor c-Rel of the NF-κB family. Unlike MDSCs described previously, the c-Rel-dependent monocytes expressed a high amount of the proinflammatory cytokine IL-1β together with a low level of suppressive molecule arginase 1. Both in vitro and in tumor-bearing mice, these c-Rel+ IL-1βhi Arg1- monocytes promoted tumor growth by potently suppressing T cell function and showed a strong migratory phenotype, all of which were impaired by c-Rel deficiency or inhibition. Mechanistic studies revealed that c-Rel controlled the expression of monocyte signature genes through a unique transcriptional complex called the c-Rel enhanceosome, and IL-1β-CCL2 crosstalk between tumor cells and the rel-dependent monocytes maintained the suppressive tumor microenvironment. Thus, c-Rel specifies the development of a suppressive monocyte population and could be selectively targeted for treating cancer.
Collapse
Affiliation(s)
- Ting Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mayassa J Bou-Dargham
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Norman Fultang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xinyuan Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Warren S Pear
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Honghong Sun
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Faculty of Pharmaceutical Sciences, CAS Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong, China
| |
Collapse
|
22
|
He K, Liu X, Hoffman RD, Shi RZ, Lv GY, Gao JL. G-CSF/GM-CSF-induced hematopoietic dysregulation in the progression of solid tumors. FEBS Open Bio 2022; 12:1268-1285. [PMID: 35612789 PMCID: PMC9249339 DOI: 10.1002/2211-5463.13445] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 11/06/2022] Open
Abstract
There are two types of abnormal hematopoiesis in solid tumor occurrence and treatment: pathological hematopoiesis, and myelosuppression induced by radiotherapy and chemotherapy. In this review, we primarily focus on the abnormal pathological hematopoietic differentiation in cancer induced by tumor-released granulocyte colony stimulating factor (G-CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF). As key factors in hematopoietic development, G-CSF/GM-CSF are well-known facilitators of myelopoiesis and mobilization of hematopoietic stem cells (HSCs). In addition, these two cytokines can also promote or inhibit tumors, dependent on tumor type. In multiple cancer types, hematopoiesis is greatly enhanced and abnormal lineage differentiation is induced by these two cytokines. Here, dysregulated hematopoiesis induced by G-CSF/GM-CSF in solid tumors and its mechanism are summarized, and the prognostic value of G-CSF/GM-CSF-associated dysregulated hematopoiesis for tumor metastasis is also briefly highlighted.
Collapse
Affiliation(s)
- Kai He
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Xi Liu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Robert D Hoffman
- Yo San University of Traditional Chinese Medicine, Los Angeles, CA, 90066, USA
| | - Rong-Zhen Shi
- Tangqi Branch of Traditional Chinese Medicine Hospital of Yuhang District, Hangzhou, Zhejiang, 311106, China
| | - Gui-Yuan Lv
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University Hangzhou, Zhejiang, 310053, China
| | - Jian-Li Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University Hangzhou, Zhejiang, 310053, China
| |
Collapse
|
23
|
Liu JQ, Zhang C, Zhang X, Yan J, Zeng C, Talebian F, Lynch K, Zhao W, Hou X, Du S, Kang DD, Deng B, McComb DW, Bai XF, Dong Y. Intratumoral delivery of IL-12 and IL-27 mRNA using lipid nanoparticles for cancer immunotherapy. J Control Release 2022; 345:306-313. [PMID: 35301053 PMCID: PMC9133152 DOI: 10.1016/j.jconrel.2022.03.021] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 02/05/2022] [Accepted: 03/10/2022] [Indexed: 11/21/2022]
Abstract
Cytokines are important immunotherapeutics with approved drugs for the treatment of human cancers. However, systemic administration of cytokines often fails to achieve adequate concentrations to immune cells in tumors due to dose-limiting toxicity. Thus, developing localized therapy that directly delivers immune-stimulatory cytokines to tumors may improve the therapeutic efficacy. In this study, we generated novel lipid nanoparticles (LNPs) encapsulated with mRNAs encoding cytokines including IL-12, IL-27 and GM-CSF, and tested their anti-tumor activity. We first synthesized ionizable lipid materials containing di-amino groups with various head groups (DALs). The novel DAL4-LNP effectively delivered different mRNAs in vitro to tumor cells and in vivo to tumors. Intratumoral injection of DAL4-LNP loaded with IL-12 mRNA was most potent in inhibiting B16F10 melanoma tumor growth compared to IL-27 or GM-CSF mRNAs in monotherapy. Furthermore, intratumoral injection of dual DAL4-LNP-IL-12 mRNA and IL-27 mRNA showed a synergistic effect in suppressing tumor growth without causing systematic toxicity. Most importantly, intratumoral delivery of IL-12 and IL-27 mRNAs induced robust infiltration of immune effector cells, including IFN-γ and TNF-α producing NK and CD8+ T cells into tumors. Thus, intratumoral administration of DAL-LNP loaded with IL-12 and IL-27 mRNA provides a new treatment strategy for cancer.
Collapse
Affiliation(s)
- Jin-Qing Liu
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Chengxiang Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Xinfu Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Jingyue Yan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Chunxi Zeng
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States; Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Fatemeh Talebian
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Kimberly Lynch
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Weiyu Zhao
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Xucheng Hou
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Shi Du
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Diana D Kang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Binbin Deng
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, United States
| | - David W McComb
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, United States; Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States
| | - Xue-Feng Bai
- Department of Pathology, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States; Department of Radiation Oncology, Department of Biomedical Engineering, The Center for Clinical and Translational Science, The Comprehensive Cancer Center, Dorothy M. Davis Heart & Lung Research Institute, Center for Cancer Engineering, Center for Cancer Metabolism, Pelotonia Institute for Immune-Oncology, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
24
|
Deng H, Muthupalani S, Erdman S, Liu H, Niu Z, Wang TC, Fox JG. Translocation of Helicobacter hepaticus synergizes with myeloid-derived suppressor cells and contributes to breast carcinogenesis. Oncoimmunology 2022; 11:2057399. [PMID: 35371619 PMCID: PMC8966989 DOI: 10.1080/2162402x.2022.2057399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Microbial dysbiosis plays an important role in the development of intestinal diseases. Recent studies suggest a link between intestinal bacteria and mammary cancer. Here, we report that female ApcMin/+ mice infected with Helicobacter hepaticus exhibited an increased mammary and small/large intestine tumor burden compared with uninfected littermates. H. hepaticus DNA was detected in small/large intestine, mammary tumors, and adjacent lymph nodes, suggesting a migration pathway. CD11b+Gr1+ myeloid-derived suppressor cells (MDSCs) infiltrated and expressed high levels of Wnts, likely enhancing tumorigenesis through activation of Wnt/β-catenin pathway. Our previous studies indicated that histidine decarboxylase (Hdc) marks a population of myeloid-biased hematopoietic stem cells and granulocytic MDSCs. Cytokines/chemokines secreted by IL-17-expressing mast cells and tumor tissues promoted Hdc+ MDSCs expansion and trafficking toward mammary tumors. Adoptive transfer of MDSCs isolated from H. hepaticus-infected mice increased MDSCs frequencies in peripheral blood, mesenteric lymph nodes, mammary gland, and lymph nodes in recipient ApcMin/+ mice. The adoptive transfer of H. hepaticus primed MDSCs also increased the size and number of mammary tumors. Our results demonstrate that H. hepaticus can translocate from the intestine to mammary tissues to promote mammary tumorigenesis with MDSCs. Targeting bacteria and MDSCs may be useful for the prevention and therapy of extraintestinal cancers. Abbreviations: Helicobacter hepaticus, Hh; myeloid-derived suppressor cell, MDSC; histidine decarboxylase, Hdc; Breast cancer, BC; T regulatory, TR; inflammatory bowel disease, IBD; fluorescence in situ hybridization, FISH; myeloid-biased hematopoietic stem cells, MB-HSCs; granulocytic MDSCs, PMN-MDSCs; Lipopolysaccharide, LPS; Toll-like receptors, TLRs; Mast cells, MCs; Granulocyte-macrophage colony-stimulating factor, GM-CSF; epithelial–mesenchymal transition, EMT; Intestinal epithelial cells, IECs.
Collapse
Affiliation(s)
- Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | | | - Susan Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Haibo Liu
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhengchuan Niu
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
25
|
Mirshahi F, Aqbi HF, Isbell M, Manjili SH, Guo C, Saneshaw M, Dozmorov M, Khosla A, Wack K, Carrasco-Zevallos OM, Idowu MO, Wang XY, Sanyal AJ, Manjili MH, Bandyopadhyay D. Distinct hepatic immunological patterns are associated with the progression or inhibition of hepatocellular carcinoma. Cell Rep 2022; 38:110454. [PMID: 35235789 PMCID: PMC9028248 DOI: 10.1016/j.celrep.2022.110454] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 12/30/2022] Open
Abstract
To discover distinct immune responses promoting or inhibiting hepatocellular carcinoma (HCC), we perform a three-dimensional analysis of the immune cells, correlating immune cell types, interactions, and changes over time in an animal model displaying gender disparity in nonalcoholic fatty liver disease (NAFLD)-associated HCC. In response to a Western diet (WD), animals mount acute and chronic patterns of inflammatory cytokines, respectively. Tumor progression in males and females is associated with a predominant CD8+ > CD4+, Th1 > Th17 > Th2, NKT > NK, M1 > M2 pattern in the liver. A complete rescue of females from HCC is associated with an equilibrium Th1 = Th17 = Th2, NKT = NK, M1 = M2 pattern, while a partial rescue of males from HCC is associated with an equilibrium CD8+ = CD4+, NKT = NK and a semi-equilibrium Th1 = Th17 > Th2 but a sustained M1 > M2 pattern in the liver. Our data suggest that immunological pattern-recognition can explain immunobiology of HCC and guide immune modulatory interventions for the treatment of HCC in a gender-specific manner. Mirshahi et al. performed a three-dimensional analysis of hepatic and splenic immune cells, correlating the immune cell types, their interactions and proportions, and changes over time. They discover gender-associated immunological patterns determining tumor progression, as well as partial or complete inhibition of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Faridoddin Mirshahi
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA 23298, USA
| | - Hussein F Aqbi
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA 23298, USA; VCU Massey Cancer Center, 401 College Street, Richmond, VA 23298, USA; College of Science, Mustansiriyah University, Baghdad, Iraq
| | - Madison Isbell
- Department of Microbiology & Immunology, VCU School of Medicine, Richmond, VA 23298, USA
| | - Saeed H Manjili
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA 23298, USA
| | - Chunqing Guo
- Department of Human & Molecular Genetics, VCU School of Medicine, Richmond, VA 23298, USA
| | - Mulugeta Saneshaw
- VCU Massey Cancer Center, 401 College Street, Richmond, VA 23298, USA
| | - Mikhail Dozmorov
- VCU Massey Cancer Center, 401 College Street, Richmond, VA 23298, USA; Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | | - Michael O Idowu
- VCU Massey Cancer Center, 401 College Street, Richmond, VA 23298, USA; Department of Pathology, VCU School of Medicine, Richmond, VA 23298, USA
| | - Xiang-Yang Wang
- VCU Massey Cancer Center, 401 College Street, Richmond, VA 23298, USA; Department of Human & Molecular Genetics, VCU School of Medicine, Richmond, VA 23298, USA; Hunter Holmes McGuire VA Medical Center, Richmond, VA 23298, USA
| | - Arun J Sanyal
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA 23298, USA; VCU Massey Cancer Center, 401 College Street, Richmond, VA 23298, USA.
| | - Masoud H Manjili
- Department of Microbiology & Immunology, VCU School of Medicine, Richmond, VA 23298, USA; VCU Massey Cancer Center, 401 College Street, Richmond, VA 23298, USA; Department of Pathology, VCU School of Medicine, Richmond, VA 23298, USA.
| | - Dipankar Bandyopadhyay
- VCU Massey Cancer Center, 401 College Street, Richmond, VA 23298, USA; Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
26
|
Bayik D, Lee J, Lathia JD. The Role of Myeloid-Derived Suppressor Cells in Tumor Growth and Metastasis. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:189-217. [PMID: 35165865 DOI: 10.1007/978-3-030-91311-3_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature bone marrow-derived suppressive cells that are an important component of the pathological immune response associated with cancer. Expansion of MDSCs has been linked to poor disease outcome and therapeutic resistance in patients with various malignancies, making these cells potential targets for next-generation treatment strategies. MDSCs are classified into monocytic (M-MDSC) and polymorphonuclear/granulocytic (PMN-MDSC) subtypes that undertake distinct and numerous roles in the tumor microenvironment or systemically to drive disease progression. In this chapter, we will discuss how MDSC subsets contribute to the growth of primary tumors and induce metastatic spread by suppressing the antitumor immune response, supporting cancer stem cell (CSC)/epithelial-to-mesenchymal transition (EMT) phenotypes and promoting angiogenesis. We will also summarize the signaling networks involved in the crosstalk between cancer cells and MDSCs that could represent putative immunotherapy targets.
Collapse
Affiliation(s)
- Defne Bayik
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Euclid, OH, USA
| | - Juyeun Lee
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Justin D Lathia
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA. .,Case Comprehensive Cancer Center, Euclid, OH, USA.
| |
Collapse
|
27
|
Ma T, Renz BW, Ilmer M, Koch D, Yang Y, Werner J, Bazhin AV. Myeloid-Derived Suppressor Cells in Solid Tumors. Cells 2022; 11:cells11020310. [PMID: 35053426 PMCID: PMC8774531 DOI: 10.3390/cells11020310] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/03/2022] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are one of the main suppressive cell population of the immune system. They play a pivotal role in the establishment of the tumor microenvironment (TME). In the context of cancers or other pathological conditions, MDSCs can differentiate, expand, and migrate in large quantities during circulation, inhibiting the cytotoxic functions of T cells and NK cells. This process is regulated by ROS, iNOS/NO, arginase-1, and multiple soluble cytokines. The definition of MDSCs and their phenotypes in humans are not as well represented as in other organisms such as mice, owing to the absence of the cognate molecule. However, a comprehensive understanding of the differences between different species and subsets will be beneficial for clarifying the immunosuppressive properties and potential clinical values of these cells during tumor progression. Recently, experimental evidence and clinical investigations have demonstrated that MDSCs have a close relationship with poor prognosis and drug resistance, which is considered to be a leading marker for practical applications and therapeutic methods. In this review, we summarize the remarkable position of MDSCs in solid tumors, explain their classifications in different models, and introduce new treatment approaches to target MDSCs to better understand the advancement of new approaches to cancer treatment.
Collapse
Affiliation(s)
- Tianmiao Ma
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (T.M.); (B.W.R.); (M.I.); (D.K.); (J.W.)
| | - Bernhard W. Renz
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (T.M.); (B.W.R.); (M.I.); (D.K.); (J.W.)
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Matthias Ilmer
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (T.M.); (B.W.R.); (M.I.); (D.K.); (J.W.)
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Dominik Koch
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (T.M.); (B.W.R.); (M.I.); (D.K.); (J.W.)
| | - Yuhui Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China;
| | - Jens Werner
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (T.M.); (B.W.R.); (M.I.); (D.K.); (J.W.)
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (T.M.); (B.W.R.); (M.I.); (D.K.); (J.W.)
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Correspondence:
| |
Collapse
|
28
|
Lin HJ, Liu Y, Lofland D, Lin J. Breast Cancer Tumor Microenvironment and Molecular Aberrations Hijack Tumoricidal Immunity. Cancers (Basel) 2022; 14:cancers14020285. [PMID: 35053449 PMCID: PMC8774102 DOI: 10.3390/cancers14020285] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Immune therapy is designed to stimulate tumoricidal effects in a variety of solid tumors including breast carcinomas. However, the emergence of resistant clones leads to treatment failure. Understanding the molecular, cellular, and microenvironmental aberrations is crucial to uncovering underlying mechanisms and developing advanced strategies for preventing or combating these resistant malignancies. This review will summarize research findings revealing various mechanisms employed to hijack innate and adaptive immune surveillance mechanisms, develop hypoxic and tumor promoting metabolism, and foster an immune tolerance microenvironment. In addition, it will highlight potential targets for therapeutic approaches. Abstract Breast cancer is the most common malignancy among females in western countries, where women have an overall lifetime risk of >10% for developing invasive breast carcinomas. It is not a single disease but is composed of distinct subtypes associated with different clinical outcomes and is highly heterogeneous in both the molecular and clinical aspects. Although tumor initiation is largely driven by acquired genetic alterations, recent data suggest microenvironment-mediated immune evasion may play an important role in neoplastic progression. Beyond surgical resection, radiation, and chemotherapy, additional therapeutic options include hormonal deactivation, targeted-signaling pathway treatment, DNA repair inhibition, and aberrant epigenetic reversion. Yet, the fatality rate of metastatic breast cancer remains unacceptably high, largely due to treatment resistance and metastases to brain, lung, or bone marrow where tumor bed penetration of therapeutic agents is limited. Recent studies indicate the development of immune-oncological therapy could potentially eradicate this devastating malignancy. Evidence suggests tumors express immunogenic neoantigens but the immunity towards these antigens is frequently muted. Established tumors exhibit immunological tolerance. This tolerance reflects a process of immune suppression elicited by the tumor, and it represents a critical obstacle towards successful antitumor immunotherapy. In general, immune evasive mechanisms adapted by breast cancer encompasses down-regulation of antigen presentations or recognition, lack of immune effector cells, obstruction of anti-tumor immune cell maturation, accumulation of immunosuppressive cells, production of inhibitory cytokines, chemokines or ligands/receptors, and up-regulation of immune checkpoint modulators. Together with altered metabolism and hypoxic conditions, they constitute a permissive tumor microenvironment. This article intends to discern representative incidents and to provide potential innovative therapeutic regimens to reinstate tumoricidal immunity.
Collapse
Affiliation(s)
- Huey-Jen Lin
- Department of Medical & Molecular Sciences, University of Delaware, Willard Hall Education Building, 16 West Main Street, Newark, DE 19716, USA
- Correspondence: ; Tel.: +1-302-831-7576; Fax: +1-302-831-4180
| | - Yingguang Liu
- Department of Molecular and Cellular Sciences, College of Osteopathic Medicine, Liberty University, 306 Liberty View Lane, Lynchburg, VA 24502, USA;
| | - Denene Lofland
- Department of Microbiology and Immunology, Tower Campus, Drexel University College of Medicine, 50 Innovation Way, Wyomissing, PA 19610, USA;
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, Molecular Medicine Graduate Program, University of Maryland School of Medicine and Greenebaum Comprehensive Cancer Center, 108 N. Greene Street, Baltimore, MD 21201, USA;
| |
Collapse
|
29
|
Nalawade SA, Shafer P, Bajgain P, McKenna MK, Ali A, Kelly L, Joubert J, Gottschalk S, Watanabe N, Leen A, Parihar R, Vera Valdes JF, Hoyos V. Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer. J Immunother Cancer 2021; 9:jitc-2021-003237. [PMID: 34815355 PMCID: PMC8611441 DOI: 10.1136/jitc-2021-003237] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Successful targeting of solid tumors such as breast cancer (BC) using chimeric antigen receptor (CAR) T cells has proven challenging, largely attributed to the immunosuppressive tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs) inhibit CAR T cell function and persistence within the breast TME. To overcome this challenge, we have developed CAR T cells targeting tumor-associated mucin 1 (MUC1) with a novel chimeric costimulatory receptor that targets tumor necrosis factor-related apoptosis-inducing ligand receptor 2 (TR2) expressed on MDSCs. METHODS The function of the TR2.41BB costimulatory receptor was assessed by exposing non-transduced (NT) and TR2.41BB transduced T cells to recombinant TR2, after which nuclear translocation of NFκB was measured by ELISA and western blot. The cytolytic activity of CAR.MUC1/TR2.41BB T cells was measured in a 5-hour cytotoxicity assay using MUC1+ tumor cells as targets in the presence or absence of MDSCs. In vivo antitumor activity was assessed using MDSC-enriched tumor-bearing mice treated with CAR T cells with or without TR2.41BB. RESULTS Nuclear translocation of NFκB in response to recombinant TR2 was detected only in TR2.41BB T cells. The presence of MDSCs diminished the cytotoxic potential of CAR.MUC1 T cells against MUC1+ BC cell lines by 25%. However, TR2.41BB expression on CAR.MUC1 T cells induced MDSC apoptosis, thereby restoring the cytotoxic activity of CAR.MUC1 T cells against MUC1+ BC lines. The presence of MDSCs resulted in an approximately twofold increase in tumor growth due to enhanced angiogenesis and fibroblast accumulation compared with mice with tumor alone. Treatment of these MDSC-enriched tumors with CAR.MUC1.TR2.41BB T cells led to superior tumor cell killing and significant reduction in tumor growth (24.54±8.55 mm3) compared with CAR.MUC1 (469.79±81.46 mm3) or TR2.41BB (434.86±64.25 mm3) T cells alone. CAR.MUC1.TR2.41BB T cells also demonstrated improved T cell proliferation and persistence at the tumor site, thereby preventing metastases. We observed similar results using CAR.HER2.TR2.41BB T cells in a HER2+ BC model. CONCLUSIONS Our findings demonstrate that CAR T cells that coexpress the TR2.4-1BB receptor exhibit superior antitumor potential against breast tumors containing immunosuppressive and tumor promoting MDSCs, resulting in TME remodeling and improved T cell proliferation at the tumor site.
Collapse
Affiliation(s)
- Saisha A Nalawade
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Paul Shafer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Pradip Bajgain
- Mouse Cancer Genetics Program, National Cancer Institute at Frederick, Frederick, Maryland, USA
| | - Mary K McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Arushana Ali
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Lauren Kelly
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Jarrett Joubert
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Stephen Gottschalk
- Bone Marrow Transplant Department, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Ann Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Robin Parihar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | | | - Valentina Hoyos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
30
|
Jesser EA, Brady NJ, Huggins DN, Witschen PM, O'Connor CH, Schwertfeger KL. STAT5 is activated in macrophages by breast cancer cell-derived factors and regulates macrophage function in the tumor microenvironment. Breast Cancer Res 2021; 23:104. [PMID: 34743736 PMCID: PMC8573892 DOI: 10.1186/s13058-021-01481-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In breast cancer, complex interactions between tumor cells and cells within the surrounding stroma, such as macrophages, are critical for tumor growth, progression, and therapeutic response. Recent studies have highlighted the complex nature and heterogeneous populations of macrophages associated with both tumor-promoting and tumor-inhibiting phenotypes. Defining the pathways that drive macrophage function is important for understanding their complex phenotypes within the tumor microenvironment. Signal transducer and activator of transcription (STAT) transcription factors, such as STAT5, are key regulators of immune cell function. The studies described here investigate the functional contributions of STAT5 to tumor-associated macrophage function in breast cancer. METHODS Initial studies were performed using a panel of human breast cancer and mouse mammary tumor cell lines to determine the ability of tumor cell-derived factors to induce STAT5 activation in macrophages. Further studies used these models to identify soluble factors that activate STAT5 in macrophages. To delineate STAT5-specific contributions to macrophage function, a conditional model of myeloid STAT5 deletion was used for in vitro, RNA-sequencing, and in vivo studies. The effects of STAT5 deletion in macrophages on tumor cell migration and metastasis were evaluated using in vitro co-culture migration assays and an in vivo tumor cell-macrophage co-injection model. RESULTS We demonstrate here that STAT5 is robustly activated in macrophages by tumor cell-derived factors and that GM-CSF is a key cytokine stimulating this pathway. The analysis of RNA-seq studies reveals that STAT5 promotes expression of immune stimulatory genes in macrophages and that loss of STAT5 in macrophages results in increased expression of tissue remodeling factors. Finally, we demonstrate that loss of STAT5 in macrophages promotes tumor cell migration in vitro and mammary tumor metastasis in vivo. CONCLUSIONS Breast cancer cells produce soluble factors, such as GM-CSF, that activate the STAT5 pathway in macrophages and drive expression of inflammatory factors. STAT5 deletion in myeloid cells enhances metastasis, suggesting that STAT5 activation in tumor-associated macrophages protects against tumor progression. Understanding mechanisms that drive macrophage function in the tumor microenvironment will ultimately lead to new approaches that suppress tumor-promoting functions while enhancing their anti-tumor functions.
Collapse
Affiliation(s)
- Emily A Jesser
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, USA
| | - Nicholas J Brady
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Danielle N Huggins
- Department of Laboratory Medicine and Pathology, 6Th St SE, University of Minnesota, Minneapolis, MN, USA
| | - Patrice M Witschen
- Comparative and Molecular Biosciences Graduate Program, University of Minnesota, Minneapolis, USA
| | - Christine H O'Connor
- Department of Laboratory Medicine and Pathology, 6Th St SE, University of Minnesota, Minneapolis, MN, USA
- University of Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Kathryn L Schwertfeger
- Department of Laboratory Medicine and Pathology, 6Th St SE, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Center for Immunology, University of Minnesota, Minneapolis, USA.
| |
Collapse
|
31
|
Williams MM, Hafeez SA, Christenson JL, O’Neill KI, Hammond NG, Richer JK. Reversing an Oncogenic Epithelial-to-Mesenchymal Transition Program in Breast Cancer Reveals Actionable Immune Suppressive Pathways. Pharmaceuticals (Basel) 2021; 14:ph14111122. [PMID: 34832904 PMCID: PMC8622696 DOI: 10.3390/ph14111122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
Approval of checkpoint inhibitors for treatment of metastatic triple negative breast cancer (mTNBC) has opened the door for the use of immunotherapies against this disease. However, not all patients with mTNBC respond to current immunotherapy approaches such as checkpoint inhibitors. Recent evidence demonstrates that TNBC metastases are more immune suppressed than primary tumors, suggesting that combination or additional immunotherapy strategies may be required to activate an anti-tumor immune attack at metastatic sites. To identify other immune suppressive mechanisms utilized by mTNBC, our group and others manipulated oncogenic epithelial-to-mesenchymal transition (EMT) programs in TNBC models to reveal differences between this breast cancer subtype and its more epithelial counterpart. This review will discuss how EMT modulation revealed several mechanisms, including tumor cell metabolism, cytokine milieu and secretion of additional immune modulators, by which mTNBC cells may suppress both the innate and adaptive anti-tumor immune responses. Many of these pathways/proteins are under preclinical or clinical investigation as therapeutic targets in mTNBC and other advanced cancers to enhance their response to chemotherapy and/or checkpoint inhibitors.
Collapse
|
32
|
Liu C, Qiang J, Deng Q, Xia J, Deng L, Zhou L, Wang D, He X, Liu Y, Zhao B, Lv J, Yu Z, Lei QY, Shao ZM, Zhang XY, Zhang L, Liu S. ALDH1A1 activity in tumor-initiating cells remodels myeloid-derived suppressor cells to promote breast cancer progression. Cancer Res 2021; 81:5919-5934. [PMID: 34580061 DOI: 10.1158/0008-5472.can-21-1337] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/17/2021] [Accepted: 09/24/2021] [Indexed: 11/16/2022]
Abstract
Tumor-initiating cells (TIC) are associated with tumor initiation, growth, metastasis, and recurrence. Aldehyde dehydrogenase 1A1 (ALDH1A1) is a TIC marker in many cancers, including breast cancer. However the molecular mechanisms underlying ALDH1A1 functions in solid tumors remain largely unknown. Here we demonstrate that ALDH1A1 enzymatic activity facilitates breast tumor growth. Mechanistically, ALDH1A1 decreased the intracellular pH in breast cancer cells to promote phosphorylation of TAK1, activate NFκB signaling, and increase the secretion of granulocyte macrophage colony-stimulating factor (GM-CSF), which led to myeloid-derived suppressor cell (MDSC) expansion and immunosuppression. Furthermore, the ALDH1A1 inhibitor disulfiram and chemotherapeutic agent gemcitabine cooperatively inhibited breast tumor growth and tumorigenesis by purging ALDH+ TICs and activating T cell immunity. These findings elucidate how active ALDH1A1 modulates the immune system to promote tumor development, highlghting new therapeutic strategies for malignant breast cancer.
Collapse
Affiliation(s)
- Cuicui Liu
- Cancer Institute, Fudan University Shanghai Cancer Center
| | | | - Qiaodan Deng
- Cancer Institute, Fudan University Shanghai Cancer Center
| | - Jie Xia
- Cancer Institute, Fudan University Shanghai Cancer Center
| | - Lu Deng
- Stowers Institute for Medical Research
| | - Lei Zhou
- Cancer Institute, Fudan University Shanghai Cancer Center
| | | | - Xueyan He
- Life Science, University of Science and Technology of China
| | | | | | - Jinhui Lv
- Shanghai East Hospital, Tongji University School of Medicine
| | - Zuoren Yu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Life Sciences and Technology
| | - Qun-Ying Lei
- Department of Oncology, Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University
| | - Zhi-Ming Shao
- Breast Surgery, Fudan University Shanghai Cancer Center
| | - Xiao-Yong Zhang
- The Institute of Science and Technology for Brain-inspired Intelligence, Fudan University
| | | | - Suling Liu
- Cancer Institute, Fudan University Shanghai Cancer Center
| |
Collapse
|
33
|
Luong N, Lenz JA, Modiano JF, Olson JK. Extracellular Vesicles Secreted by Tumor Cells Promote the Generation of Suppressive Monocytes. Immunohorizons 2021; 5:647-658. [PMID: 34404719 DOI: 10.4049/immunohorizons.2000017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/14/2021] [Indexed: 11/19/2022] Open
Abstract
Monocytes are among the first cells to infiltrate the tumor microenvironment. The conversion of monocytes to suppressor cells in the tumor microenvironment is crucial in evasion of the immune response and tumor maintenance. Tumor cells may secrete products that promote the conversion of monocytes to suppressor cells. Cells secrete extracellular vesicles (EVs) containing cargos of genetic materials and proteins as a way to communicate with neighboring cells. During pathologic conditions like cancers, tumor cells increase their EVs production containing microRNA, RNA, and proteins that may affect the immune cell response, contributing to the immunosuppressive microenvironment. Our studies show that EVs secreted by a wide range of murine tumor cells, including osteosarcoma, glioma, colon carcinoma, sarcoma, and melanoma, can be taken up by bone marrow-derived monocytes. The monocytes that took up the EVs secreted by tumor cells matured toward an immune-suppressive phenotype by upregulating the expression of suppressive cytokines and effector molecules. The monocytes also downregulated MHC class II and costimulatory molecules while increasing the expression of PD-L1 on their surface after taking up EVs from tumor cells. Most importantly, monocytes exposed to EVs secreted by tumor cells suppressed activated Ag-specific CD4+ T cells. These results show that tumor cells from several different tumor types secrete EVs which promote the conversion of monocytes into suppressor cells, thus promoting immune evasion. These studies suggest that EVs secreted by tumors are potentially a new target for future cancer therapy.
Collapse
Affiliation(s)
- Nhungoc Luong
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN
| | - Jennifer A Lenz
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN
| | - Jaime F Modiano
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN; and
| | - Julie K Olson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN;
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN
| |
Collapse
|
34
|
Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther 2021; 6:263. [PMID: 34248142 PMCID: PMC8273155 DOI: 10.1038/s41392-021-00658-5] [Citation(s) in RCA: 1286] [Impact Index Per Article: 321.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/11/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.
Collapse
|
35
|
Liu G, Li H, Zhang W, Yu J, Zhang X, Wu R, Niu M, Liu X, Yu R. Csnk1a1 inhibition modulates the inflammatory secretome and enhances response to radiotherapy in glioma. J Cell Mol Med 2021; 25:7395-7406. [PMID: 34216174 PMCID: PMC8335695 DOI: 10.1111/jcmm.16767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/28/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM), a fatal brain tumour with no available targeted therapies, has a poor prognosis. At present, radiotherapy is one of the main methods to treat glioma, but it leads to an obvious increase in inflammatory factors in the tumour microenvironment, especially IL-6 and CXCL1, which plays a role in tumour to resistance radiotherapy and tumorigenesis. Casein kinase 1 alpha 1 (CK1α) (encoded on chromosome 5q by Csnk1a1) is considered an attractive target for Tp53 wild-type acute myeloid leukaemia (AML) treatment. In this study, we evaluated the anti-tumour effect of Csnk1a1 suppression in GBM cells in vitro and in vivo. We found that down-regulation of Csnk1a1 or inhibition by D4476, a Csnk1a1 inhibitor, reduced GBM cell proliferation efficiently in both Tp53 wild-type and Tp53-mutant GBM cells. On the contrary, overexpression of Csnk1a1 promoted cell proliferation and colony formation. Csnk1a1 inhibition improved the sensitivity to radiotherapy. Furthermore, down-regulation of Csnk1a1 reduced the production and secretion of pro-inflammatory factors. In the preclinical GBM model, treatment with D4476 significantly inhibited the increase in pro-inflammatory factors caused by radiotherapy and improved radiotherapy sensitivity, thus inhibiting tumour growth and prolonging animal survival time. These results suggest targeting Csnk1a1 exert an anti-tumour role as an inhibitor of inflammatory factors, providing a new strategy for the treatment of glioma.
Collapse
Affiliation(s)
- Guanzheng Liu
- Insititute of Nervous System Diseases, the Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Huan Li
- Insititute of Nervous System Diseases, the Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wanhong Zhang
- Insititute of Nervous System Diseases, the Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, Kaifeng Central hospital, Kaifeng, China
| | - Jiefeng Yu
- Insititute of Nervous System Diseases, the Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Xu Zhang
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Runqiu Wu
- Insititute of Nervous System Diseases, the Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Mingshan Niu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Xuejiao Liu
- Insititute of Nervous System Diseases, the Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Rutong Yu
- Insititute of Nervous System Diseases, the Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
36
|
Abstract
Immunotherapy has revolutionized cancer treatment, but efficacy remains limited in most clinical settings. Cancer is a systemic disease that induces many functional and compositional changes to the immune system as a whole. Immunity is regulated by interactions of diverse cell lineages across tissues. Therefore, an improved understanding of tumour immunology must assess the systemic immune landscape beyond the tumour microenvironment (TME). Importantly, the peripheral immune system is required to drive effective natural and therapeutically induced antitumour immune responses. In fact, emerging evidence suggests that immunotherapy drives new immune responses rather than the reinvigoration of pre-existing immune responses. However, new immune responses in individuals burdened with tumours are compromised even beyond the TME. Herein, we aim to comprehensively outline the current knowledge of systemic immunity in cancer.
Collapse
Affiliation(s)
- Kamir J Hiam-Galvez
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, San Francisco, CA, USA
| | - Breanna M Allen
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, San Francisco, CA, USA
| | - Matthew H Spitzer
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA.
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
37
|
Hargreaves A, Barry ST, Bigley A, Kendrew J, Price S. Tumors modulate fenestrated vascular beds and host endocrine status. J Appl Toxicol 2021; 41:1952-1965. [PMID: 33977518 DOI: 10.1002/jat.4176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 11/12/2022]
Abstract
Allograft and xenograft transplantation into a mouse host is frequently utilized to study cancer biology, tumor behavior, and response to treatment. Preclinical studies employing these models often focus solely upon the intra-tumoral effects of a given treatment, without consideration of systemic toxicity or tumor-host interaction, nor whether this latter relationship could modulate the toxicologic response to therapy. Here it is demonstrated that the implantation and growth of a range of human- and mouse-derived cell lines leads to structural vascular and, potentially, functional changes within peripheral endocrine tissues, a process that could conceivably ameliorate the severity of anti-angiogenic-induced fenestrated vessel attenuation. Observations suggest a multifactorial process, which may involve host- and tumor-derived cytokines/growth factors, and the liberation of myeloid-derived suppressor cells. Further investigation revealed a structurally comparable response to the administration of exogenous estrogen. These findings, in addition to providing insight into the development of clinical anti-angiogenic "adaptation," may be of significance within the "cancer-cachexia" and cancer-related anemia syndromes in man.
Collapse
|
38
|
Sharma V, Aggarwal A, Jacob J, Sahni D. Myeloid-derived suppressor cells: Bridging the gap between inflammation and pancreatic adenocarcinoma. Scand J Immunol 2021; 93:e13021. [PMID: 33455004 DOI: 10.1111/sji.13021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 01/09/2021] [Accepted: 01/10/2021] [Indexed: 12/29/2022]
Abstract
Pancreatic cancer has been identified as one of the deadliest malignancies because it remains asymptomatic and usually presents in the advanced stage. Tumour immune evasion is a well-known mechanism of tumorigenesis in various forms of human malignancies. Chronic inflammation via complex networking of various inflammatory cytokines in the local tissue microenvironment dysregulates the immune system and support tumour development. Pro-inflammatory mediators present in the tumour microenvironment increase the tumour burden by causing immune suppression through the generation of myeloid-derived suppressor cells (MDSCs) and T regulatory cells. These cells, along-with myofibroblasts, create a highly immunosuppressive and resistant tumour microenvironment and are thus considered as one of the culprits for the failure of anti-cancer chemotherapies in pancreatic adenocarcinoma patients. Targeting these MDSCs using various combinatorial approaches might have the potential for abrogating the resistance and suppressive nature of the pancreatic tumour microenvironment. Therefore, there is more curiosity in studying the crosstalk of MDSCs with other immune cells during pathological conditions and the underlying mechanisms of immunosuppression in the current scenario. In this article, the possible role of MDSCs in inflammation-mediated tumour progression of pancreatic adenocarcinoma has been discussed.
Collapse
Affiliation(s)
- Vinit Sharma
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Anjali Aggarwal
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Justin Jacob
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Daisy Sahni
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
39
|
Yan X, Xie Y, Yang F, Hua Y, Zeng T, Sun C, Yang M, Huang X, Wu H, Fu Z, Li W, Jiao S, Yin Y. Comprehensive description of the current breast cancer microenvironment advancements via single-cell analysis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:142. [PMID: 33906694 PMCID: PMC8077685 DOI: 10.1186/s13046-021-01949-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/15/2021] [Indexed: 02/07/2023]
Abstract
Breast cancer is a heterogeneous disease with a complex microenvironment consisting of tumor cells, immune cells, fibroblasts and vascular cells. These cancer-associated cells shape the tumor microenvironment (TME) and influence the progression of breast cancer and the therapeutic responses in patients. The exact composition of the intra-tumoral cells is mixed as the highly heterogeneous and dynamic nature of the TME. Recent advances in single-cell technologies such as single-cell DNA sequencing (scDNA-seq), single-cell RNA sequencing (scRNA-seq) and mass cytometry have provided new insights into the phenotypic and functional diversity of tumor-infiltrating cells in breast cancer. In this review, we have outlined the recent progress in single-cell characterization of breast tumor ecosystems, and summarized the phenotypic diversity of intra-tumoral cells and their potential prognostic relevance.
Collapse
Affiliation(s)
- Xueqi Yan
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yinghong Xie
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Fan Yang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yijia Hua
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tianyu Zeng
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chunxiao Sun
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Mengzhu Yang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiang Huang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hao Wu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ziyi Fu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wei Li
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Shiping Jiao
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210029, Jiangsu Province, China. .,Drum Tower Institute of clinical medicine, Nanjing University, Nanjing, 210029, Jiangsu Province, China.
| | - Yongmei Yin
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
40
|
van Wigcheren GF, Roelofs D, Figdor CG, Flórez-Grau G. Three distinct tolerogenic CD14 + myeloid cell types to actively manage autoimmune disease: Opportunities and challenges. J Autoimmun 2021; 120:102645. [PMID: 33901801 DOI: 10.1016/j.jaut.2021.102645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 01/18/2023]
Abstract
Current treatment for patients with autoimmune disorders including rheumatoid arthritis, multiple sclerosis and type 1 diabetes, often consists of long-term drug regimens that broadly dampen immune responses. These non-specific treatments are frequently associated with severe side effects creating an urgent need for safer and more effective therapy to promote peripheral tolerance in autoimmune diseases. Cell-based immunotherapy may offer an encouraging alternative, where tolerogenic CD14+ myeloid cells are infused to inhibit autoreactive effector cells. In this review, we compared in depth three promising tolerogenic CD14+ candidates for the treatment of autoimmune disease: 1) tolerogenic dendritic cells, 2) monocytic myeloid-derived suppressor cells and 3) CD14+ type 2 conventional dendritic cells. TolDC-based therapy has entered clinical testing whereas evidence from the latter two cell types m-MDSCs and CD14+ cDC2s is predominantly coming from cancer immunology research. These three cell types have distinct cellular properties and immunosuppressive mechanisms offering unique opportunities to be explored. However, these cells differ in stage of development towards immunotherapy each facing additional hurdles. Therefore, we speculate on the potential benefits and risks of these cell types as novel cell-based immunotherapies to control autoimmune disease in patients.
Collapse
Affiliation(s)
- Glenn F van Wigcheren
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands; Oncode Institute, the Netherlands
| | - Daphne Roelofs
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands; Oncode Institute, the Netherlands.
| | - Georgina Flórez-Grau
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands
| |
Collapse
|
41
|
Bejarano L, Jordāo MJC, Joyce JA. Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov 2021; 11:933-959. [PMID: 33811125 DOI: 10.1158/2159-8290.cd-20-1808] [Citation(s) in RCA: 870] [Impact Index Per Article: 217.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 01/10/2023]
Abstract
Strategies to therapeutically target the tumor microenvironment (TME) have emerged as a promising approach for cancer treatment in recent years due to the critical roles of the TME in regulating tumor progression and modulating response to standard-of-care therapies. Here, we summarize the current knowledge regarding the most advanced TME-directed therapies, which have either been clinically approved or are currently being evaluated in trials, including immunotherapies, antiangiogenic drugs, and treatments directed against cancer-associated fibroblasts and the extracellular matrix. We also discuss some of the challenges associated with TME therapies, and future perspectives in this evolving field. SIGNIFICANCE: This review provides a comprehensive analysis of the current therapies targeting the TME, combining a discussion of the underlying basic biology with clinical evaluation of different therapeutic approaches, and highlighting the challenges and future perspectives.
Collapse
Affiliation(s)
- Leire Bejarano
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Marta J C Jordāo
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
42
|
Metcalf KJ, Alazzeh A, Werb Z, Weaver VM. Leveraging microenvironmental synthetic lethalities to treat cancer. J Clin Invest 2021; 131:143765. [PMID: 33720045 PMCID: PMC7954586 DOI: 10.1172/jci143765] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Treatment resistance leads to cancer patient mortality. Therapeutic approaches that employ synthetic lethality to target mutational vulnerabilities in key tumor cell signaling pathways have proven effective in overcoming therapeutic resistance in some cancers. Yet, tumors are organs composed of malignant cells residing within a cellular and noncellular stroma. Tumor evolution and resistance to anticancer treatment are mediated through a dynamic and reciprocal dialogue with the tumor microenvironment (TME). Accordingly, expanding tumor cell synthetic lethality to encompass contextual synthetic lethality has the potential to eradicate tumors by targeting critical TME circuits that promote tumor progression and therapeutic resistance. In this Review, we summarize current knowledge about the TME and discuss its role in treatment. We outline the concept of tumor cell-specific synthetic lethality and describe therapeutic approaches to expand this paradigm to leverage TME synthetic lethality to improve cancer therapy.
Collapse
Affiliation(s)
| | | | - Zena Werb
- Department of Anatomy
- Helen Diller Family Comprehensive Cancer Center
| | - Valerie M. Weaver
- Department of Surgery
- Helen Diller Family Comprehensive Cancer Center
- Center for Bioengineering and Tissue Regeneration, and
- Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California, USA
| |
Collapse
|
43
|
Manjili MH, Payne KK. Cancer immunotherapy: Re-programming cells of the innate and adaptive immune systems. Oncoimmunology 2021; 1:201-204. [PMID: 22720242 PMCID: PMC3377002 DOI: 10.4161/onci.1.2.18113] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cancers utilize multiple mechanisms to overcome immune responses. Emerging evidence suggest that immunotherapy of cancer should focus on inducing and re-programming cells of the innate and adaptive immune systems rather than focusing solely on T cells. Recently, we have shown that such a multifaceted approach can improve immunotherapy of breast cancer.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology and Immunology; Virginia Commonwealth University Massey Cancer Center; Richmond, VA USA
| | | |
Collapse
|
44
|
Fultang N, Li X, Li T, Chen YH. Myeloid-Derived Suppressor Cell Differentiation in Cancer: Transcriptional Regulators and Enhanceosome-Mediated Mechanisms. Front Immunol 2021; 11:619253. [PMID: 33519825 PMCID: PMC7840597 DOI: 10.3389/fimmu.2020.619253] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/30/2020] [Indexed: 01/16/2023] Open
Abstract
Myeloid-derived Suppressor Cells (MDSCs) are a sub-population of leukocytes that are important for carcinogenesis and cancer immunotherapy. During carcinogenesis or severe infections, inflammatory mediators induce MDSCs via aberrant differentiation of myeloid precursors. Although several transcription factors, including C/EBPβ, STAT3, c-Rel, STAT5, and IRF8, have been reported to regulate MDSC differentiation, none of them are specifically expressed in MDSCs. How these lineage-non-specific transcription factors specify MDSC differentiation in a lineage-specific manner is unclear. The recent discovery of the c-Rel-C/EBPβ enhanceosome in MDSCs may help explain these context-dependent roles. In this review, we examine several transcriptional regulators of MDSC differentiation, and discuss the concept of non-modular regulation of MDSC signature gene expression by transcription factors such as c-Rel and C/EBPß.
Collapse
Affiliation(s)
- Norman Fultang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | | | - Youhai H. Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
45
|
An Overview of Advances in Cell-Based Cancer Immunotherapies Based on the Multiple Immune-Cancer Cell Interactions. Methods Mol Biol 2021; 2097:139-171. [PMID: 31776925 DOI: 10.1007/978-1-0716-0203-4_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumors have a complex ecosystem in which behavior and fate are determined by the interaction of diverse cancerous and noncancerous cells at local and systemic levels. A number of studies indicate that various immune cells participate in tumor development (Fig. 1). In this review, we will discuss interactions among T lymphocytes (T cells), B cells, natural killer (NK) cells, dendritic cells (DCs), tumor-associated macrophages (TAMs), neutrophils, and myeloid-derived suppressor cells (MDSCs). In addition, we will touch upon attempts to either use or block subsets of immune cells to target cancer.
Collapse
|
46
|
Antonioli L, Fornai M, Pellegrini C, D'Antongiovanni V, Turiello R, Morello S, Haskó G, Blandizzi C. Adenosine Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1270:145-167. [PMID: 33123998 DOI: 10.1007/978-3-030-47189-7_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adenosine, deriving from ATP released by dying cancer cells and then degradated in the tumor environment by CD39/CD73 enzyme axis, is linked to the generation of an immunosuppressed niche favoring the onset of neoplasia. Signals delivered by extracellular adenosine are detected and transduced by G-protein-coupled cell surface receptors, classified into four subtypes: A1, A2A, A2B, and A3. A critical role of this nucleoside is emerging in the modulation of several immune and nonimmune cells defining the tumor microenvironment, providing novel insights about the development of novel therapeutic strategies aimed at undermining the immune-privileged sites where cancer cells grow and proliferate.
Collapse
Affiliation(s)
- Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | | | - Roberta Turiello
- Department of Pharmacy, University of Salerno, Fisciano, Italy.,PhD Program in Drug discovery and Development, Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Silvana Morello
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
47
|
Arianfar E, Shahgordi S, Memarian A. Natural Killer Cell Defects in Breast Cancer: A Key Pathway for Tumor Evasion. Int Rev Immunol 2020; 40:197-216. [PMID: 33258393 DOI: 10.1080/08830185.2020.1845670] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
As the most important innate immune component cancers invader, natural killer (NK) cells have a magnificent role in antitumor immunity without any prior sensitization. Different subsets of NK cells have distinct responses during tumor cell exposure, according to their phenotypes and environments. Their function is induced mainly by the activity of both inhibitory and activating receptors against cancerous cells. Since the immunosuppression in the tumor microenvironment of breast cancer patients has directly deteriorated the phenotype and disturbed the function of NK cells, recruiting compensatory mechanisms indicate promising outcomes for immunotherapeutic approaches. These evidences accentuate the importance of NK cell distinct features in protection against breast tumors. In this review, we discuss the several mechanisms involved in NK cells suppression which consequently promote tumor progression and disease recurrence in patients with breast cancer.
Collapse
Affiliation(s)
- Elaheh Arianfar
- Student Research Committee, Faculty of Medicine, Department of Immunology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sanaz Shahgordi
- Student Research Committee, Faculty of Medicine, Department of Immunology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Memarian
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran.,Immunology department, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
48
|
Induction of Durable Antitumor Response by a Novel Oncolytic Herpesvirus Expressing Multiple Immunomodulatory Transgenes. Biomedicines 2020; 8:biomedicines8110484. [PMID: 33182232 PMCID: PMC7695276 DOI: 10.3390/biomedicines8110484] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/28/2020] [Accepted: 11/04/2020] [Indexed: 12/17/2022] Open
Abstract
Oncolytic virotherapy is a promising new tool for cancer treatment, but direct lytic destruction of tumor cells is not sufficient and must be accompanied by strong immune activation to elicit anti-tumor immunity. We report here the creation of a novel replication-competent recombinant oncolytic herpes simplex virus type 1 (VG161) that carries genes coding for IL-12, IL-15, and IL-15 receptor alpha subunit, along with a peptide fusion protein capable of disrupting PD-1/PD-L1 interactions. The VG161 virus replicates efficiently and exhibits robust cytotoxicity in multiple tumor cell lines. Moreover, the encoded cytokines and the PD-L1 blocking peptide work cooperatively to boost immune cell function. In vivo testing in syngeneic CT26 and A20 tumor models reveals superior efficacy when compared to a backbone virus that does not express exogenous genes. Intratumoral injection of VG161 induces abscopal responses in non-injected distal tumors and grants resistance to tumor re-challenge. The robust anti-tumor effect of VG161 is associated with T cell and NK cell tumor infiltration, expression of Th1 associated genes in the injection site, and increased frequency of splenic tumor-specific T cells. VG161 also displayed a superb safety profile in GLP acute and repeated injection toxicity studies performed using cynomolgus monkeys. Overall, we demonstrate that VG161 can induce robust oncolysis and stimulate a robust anti-tumor immune response without sacrificing safety.
Collapse
|
49
|
Dendritic Cells and Myeloid Derived Suppressor Cells Fully Responsive to Stimulation via Toll-Like Receptor 4 Are Rapidly Induced from Bone-Marrow Cells by Granulocyte-Macrophage Colony-Stimulating Factor. Vaccines (Basel) 2020; 8:vaccines8030522. [PMID: 32932705 PMCID: PMC7564202 DOI: 10.3390/vaccines8030522] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) are commonly generated from bone marrow (BM) progenitor cells with granulocyte-macrophage colony-stimulating factor (GM-CSF) alone or in combination with interleukin 4 (IL-4). These cells are often harvested post day 5, when they acquire maturation markers and can stimulate T cells. Apart from DCs, myeloid derived suppressor cells (MDSCs) are also found within these cultures. However, little is known about the functional characteristics of DCs and MDSCs before day 5. Herein, using a murine model, it is shown that early DCs and MDSCs, even in cultures with GM-CSF alone, upregulate fully maturation and activation surface molecules in response to the toll-like receptor 4 (TLR4) ligand lipopolysaccharide (LPS) stimulation. Despite initially displaying lower marker expression levels, these cells efficiently induced T cell stimulation and cytokine production. Interestingly, Gr-1int MDSCs increased their T cell co-stimulatory activity upon TLR4 stimulation. Additionally, early DCs and MDSCs exhibited differential endocytic capacity for viral sized nanoparticles and bacterial sized microparticles. DCs internalized both particle sizes, whilst MDSCs only internalized the larger microparticles, with reduced endocytic activity over time in the culture. These findings have unveiled an important role for the rapid initiation of productive immunity by GM-CSF, with promising implications for future vaccine and DC immunotherapy developments.
Collapse
|
50
|
Piseddu I, Röhrle N, Knott MML, Moder S, Eiber S, Schnell K, Vetter V, Meyer B, Layritz P, Kühnemuth B, Wiedemann GM, Gruen J, Perleberg C, Rapp M, Endres S, Anz D. Constitutive Expression of CCL22 Is Mediated by T Cell-Derived GM-CSF. THE JOURNAL OF IMMUNOLOGY 2020; 205:2056-2065. [PMID: 32907996 DOI: 10.4049/jimmunol.2000004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022]
Abstract
CCL22 is a key mediator of leukocyte trafficking in inflammatory immune responses, allergy, and cancer. It acts by attracting regulatory T cells and Th2 cells via their receptor CCR type 4 (CCR4). Beyond its role in inflammation, CCL22 is constitutively expressed at high levels in lymphoid organs during homeostasis, where it controls immunity by recruiting regulatory T cells to dendritic cells (DCs). In this study, we aimed to identify the mechanisms responsible for constitutive CCL22 expression. We confirmed that CD11c+ DCs are the exclusive producers of CCL22 in secondary lymphatic organs during homeostasis. We show that in vitro both murine splenocytes and human PBMCs secrete CCL22 spontaneously without any further stimulation. Interestingly, isolated DCs alone, however, are unable to produce CCL22, but instead require T cell help. In vitro, only the coculture of DCs with T cells or their supernatants resulted in CCL22 secretion, and we identified T cell-derived GM-CSF as the major inducer of DC-derived CCL22 expression. In vivo, Rag1 -/- mice, which lack functional T cells, have low CCL22 levels in lymphoid organs, and this can be restored by adoptive transfer of wild-type T cells or administration of GM-CSF. Taken together, we uncover T cell-derived GM-CSF as a key inducer of the chemokine CCL22 and thus, to our knowledge, identify a novel role for this cytokine as a central regulator of immunity in lymphatic organs. This knowledge could contribute to the development of new therapeutic interventions in cancer and autoimmunity.
Collapse
Affiliation(s)
- Ignazio Piseddu
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Natascha Röhrle
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Maximilian Martin Ludwig Knott
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Stefan Moder
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Stephan Eiber
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Konstantin Schnell
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Viola Vetter
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Bastian Meyer
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Patrick Layritz
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Benjamin Kühnemuth
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Gabriela Maria Wiedemann
- Department of Medicine II, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; and
| | - Juliane Gruen
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Carolin Perleberg
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Moritz Rapp
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany
| | - David Anz
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, Department of Internal Medicine IV, University Hospital of Munich, 80337 Munich, Germany; .,Department of Internal Medicine II (Gastroenterology and Hepatology), University Hospital of Munich, 81377 Munich, Germany
| |
Collapse
|