1
|
Wang W, Zhai Y, Yang X, Ye L, Lu G, Shi X, Zhai G. Effective design of therapeutic nanovaccines based on tumor neoantigens. J Control Release 2025; 380:17-35. [PMID: 39892648 DOI: 10.1016/j.jconrel.2025.01.078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Neoantigen vaccines are among the most potent immunotherapies for personalized cancer treatment. Therapeutic vaccines containing tumor-specific neoantigens that elicit specific T cell responses offer the potential for long-term clinical benefits to cancer patients. Unlike immune-checkpoint inhibitors (ICIs), which rely on pre-existing specific T cell responses, personalized neoantigen vaccines not only promote existing specific T cell responses but importantly stimulate the generation of neoantigen-specific T cells, leading to the establishment of a persistent specific memory T cell pool. The review discusses the current state of clinical research on neoantigen nanovaccines, focusing on the application of vectors, adjuvants, and combinational strategies to address a range of challenges and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States of America
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
2
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
3
|
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
|
4
|
Wang J, Wang Y, Jiang X. Targeting anticancer immunity in melanoma tumour microenvironment: unleashing the potential of adjuvants, drugs, and phytochemicals. J Drug Target 2024; 32:1052-1072. [PMID: 39041142 DOI: 10.1080/1061186x.2024.2384071] [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: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Melanoma poses a challenge in oncology because of its aggressive nature and limited treatment modalities. The tumour microenvironment (TME) in melanoma contains unique properties such as an immunosuppressive and high-density environment, unusual vasculature, and a high number of stromal and immunosuppressive cells. In recent years, numerous experiments have focused on boosting the immune system to effectively remove malignant cells. Adjuvants, consisting of phytochemicals, toll-like receptor (TLR) agonists, and cytokines, have shown encouraging results in triggering antitumor immunity and augmenting the therapeutic effectiveness of anticancer therapy. These adjuvants can stimulate the maturation of dendritic cells (DCs) and infiltration of cytotoxic CD8+ T lymphocytes (CTLs). Furthermore, nanocarriers can help to deliver immunomodulators and antigens directly to the tumour stroma, thereby improving their efficacy against malignant cells. The remodelling of melanoma TME utilising phytochemicals, agonists, and other adjuvants can be combined with current modalities for improving therapy outcomes. This review article explores the potential of adjuvants, drugs, and their nanoformulations in enhancing the anticancer potency of macrophages, CTLs, and natural killer (NK) cells. Additionally, the capacity of these agents to repress the function of immunosuppressive components of melanoma TME, such as immunosuppressive subsets of macrophages, stromal and myeloid cells will be discussed.
Collapse
Affiliation(s)
- Jingping Wang
- Emergency Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Yaping Wang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Xiaofang Jiang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| |
Collapse
|
5
|
Paudel SN, Hutzen BJ, Miller KE, Garfinkle EAR, Chen CY, Wang PY, Glaspell AM, Currier MA, Ringwalt EM, Boon L, Mardis ER, Cairo MS, Ratner N, Dodd RD, Cassady KA, Cripe TP. Myelomodulatory treatments augment the therapeutic benefit of oncolytic viroimmunotherapy in murine models of malignant peripheral nerve sheath tumors. Front Immunol 2024; 15:1384623. [PMID: 39044819 PMCID: PMC11263800 DOI: 10.3389/fimmu.2024.1384623] [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: 02/10/2024] [Accepted: 06/10/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Malignant peripheral nerve sheath tumors (MPNST) pose a significant therapeutic challenge due to high recurrence rates after surgical resection and a largely ineffective response to traditional chemotherapy. An alternative treatment strategy is oncolytic viroimmunotherapy, which can elicit a durable and systemic antitumor immune response and is Food and Drug Administration (FDA)-approved for the treatment of melanoma. Unfortunately, only a subset of patients responds completely, underscoring the need to address barriers hindering viroimmunotherapy effectiveness. Methods Here we investigated the therapeutic utility of targeting key components of the MPNST immunosuppressive microenvironment to enhance viroimmunotherapy's antitumor efficacy in three murine models, one of which showed more immunogenic characteristics than the others. Results Myelomodulatory therapy with pexidartinib, a small molecule inhibitor of CSF1R tyrosine kinase, and the oncolytic herpes simplex virus T-VEC exhibited the most significant increase in median survival time in the highly immunogenic model. Additionally, targeting myeloid cells with the myelomodulatory therapy trabectedin, a small molecule activator of caspase-8 dependent apoptosis, augmented the survival benefit of T-VEC in a less immunogenic MPNST model. However, tumor regressions or shrinkages were not observed. Depletion experiments confirmed that the enhanced survival benefit relied on a T cell response. Furthermore, flow cytometry analysis following combination viroimmunotherapy revealed decreased M2 macrophages and myeloid-derived suppressor cells and increased tumor-specific gp70+ CD8 T cells within the tumor microenvironment. Discussion In summary, our findings provide compelling evidence for the potential to leverage viroimmunotherapy with myeloid cell targeting against MPNST and warrant further investigation.
Collapse
Affiliation(s)
- Siddhi N. Paudel
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Brian J. Hutzen
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Katherine E. Miller
- Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University Wexner College of Medicine, Columbus, OH, United States
| | - Elizabeth A. R. Garfinkle
- Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Chun-Yu Chen
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Pin-Yi Wang
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Andrea M. Glaspell
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Mark A. Currier
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Emily M. Ringwalt
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | | | - Elaine R. Mardis
- Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University Wexner College of Medicine, Columbus, OH, United States
| | - Mitchell S. Cairo
- Department of Pediatrics, Medicine, Pathology, Microbiology and Immunology, and Cell Biology, New York Medical College, Valhalla, NY, United States
| | - Nancy Ratner
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Rebecca D. Dodd
- Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Kevin A. Cassady
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University Wexner College of Medicine, Columbus, OH, United States
| | - Timothy P. Cripe
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University Wexner College of Medicine, Columbus, OH, United States
- Division of Pediatric Hematology/Oncology/BMT, Nationwide Children’s Hospital, Columbus, OH, United States
| |
Collapse
|
6
|
Song Y, Liao Y, Liu T, Chen Y, Wang F, Zhou Z, Zhang W, Li J. Microglial repopulation restricts ocular inflammation and choroidal neovascularization in mice. Front Immunol 2024; 15:1366841. [PMID: 38711521 PMCID: PMC11070532 DOI: 10.3389/fimmu.2024.1366841] [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: 01/07/2024] [Accepted: 04/04/2024] [Indexed: 05/08/2024] Open
Abstract
Introduction Age-related macular degeneration (AMD) is a prevalent, chronic and progressive retinal degenerative disease characterized by an inflammatory response mediated by activated microglia accumulating in the retina. In this study, we demonstrate the therapeutically effects and the underlying mechanisms of microglial repopulation in the laser-induced choroidal neovascularization (CNV) model of exudative AMD. Methods The CSF1R inhibitor PLX3397 was used to establish a treatment paradigm for microglial repopulation in the retina. Neovascular leakage and neovascular area were examined by fundus fluorescein angiography (FFA) and immunostaining of whole-mount RPE-choroid-sclera complexes in CNV mice receiving PLX3397. Altered cellular senescence was measured by beta-galactosidase (SA-β-gal) activity and p16INK4a expression. The effect and mechanisms of repopulated microglia on leukocyte infiltration and the inflammatory response in CNV lesions were analyzed. Results We showed that ten days of the CSF1R inhibitor PLX3397 treatment followed by 11 days of drug withdrawal was sufficient to stimulate rapid repopulation of the retina with new microglia. Microglial repopulation attenuated pathological choroid neovascularization and dampened cellular senescence in CNV lesions. Repopulating microglia exhibited lower levels of activation markers, enhanced phagocytic function and produced fewer cytokines involved in the immune response, thereby ameliorating leukocyte infiltration and attenuating the inflammatory response in CNV lesions. Discussion The microglial repopulation described herein are therefore a promising strategy for restricting inflammation and choroidal neovascularization, which are important players in the pathophysiology of AMD.
Collapse
Affiliation(s)
- Yinting Song
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Yuefeng Liao
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Tong Liu
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Yanxian Chen
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
- Experimental Ophthalmology, School of Optometry, The Hong Kong Polytechnic University, HongKong, Hong Kong SAR, China
| | - Fei Wang
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Zixia Zhou
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Weili Zhang
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Jinying Li
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Banuelos A, Zhang A, Berouti H, Baez M, Yılmaz L, Georgeos N, Marjon KD, Miyanishi M, Weissman IL. CXCR2 inhibition in G-MDSCs enhances CD47 blockade for melanoma tumor cell clearance. Proc Natl Acad Sci U S A 2024; 121:e2318534121. [PMID: 38261615 PMCID: PMC10835053 DOI: 10.1073/pnas.2318534121] [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: 10/24/2023] [Accepted: 12/16/2023] [Indexed: 01/25/2024] Open
Abstract
The use of colony-stimulating factor-1 receptor (CSF1R) inhibitors has been widely explored as a strategy for cancer immunotherapy due to their robust depletion of tumor-associated macrophages (TAMs). While CSF1R blockade effectively eliminates TAMs from the solid tumor microenvironment, its clinical efficacy is limited. Here, we use an inducible CSF1R knockout model to investigate the persistence of tumor progression in the absence of TAMs. We find increased frequencies of granulocytic myeloid-derived suppressor cells (G-MDSCs) in the bone marrow, throughout circulation, and in the tumor following CSF1R deletion and loss of TAMs. We find that G-MDSCs are capable of suppressing macrophage phagocytosis, and the elimination of G-MDSCs through CXCR2 inhibition increases macrophage capacity for tumor cell clearance. Further, we find that combination therapy of CXCR2 inhibition and CD47 blockade synergize to elicit a significant anti-tumor response. These findings reveal G-MDSCs as key drivers of tumor immunosuppression and demonstrate their inhibition as a potent strategy to increase macrophage phagocytosis and enhance the anti-tumor efficacy of CD47 blockade in B16-F10 melanoma.
Collapse
Affiliation(s)
- Allison Banuelos
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University, Stanford, CA94305
| | - Allison Zhang
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University, Stanford, CA94305
| | - Hala Berouti
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University, Stanford, CA94305
| | - Michelle Baez
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University, Stanford, CA94305
| | - Leyla Yılmaz
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University, Stanford, CA94305
| | - Nardin Georgeos
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University, Stanford, CA94305
| | - Kristopher D. Marjon
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University, Stanford, CA94305
| | - Masanori Miyanishi
- Hematopoietic Stem Cell Biology and Medical Innovation (HSCBMI), Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe650-0047, Japan
| | - Irving L. Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University, Stanford, CA94305
- Department of Pathology, Stanford University, Stanford, CA94305
| |
Collapse
|
8
|
Chang CH, Chen CJ, Yu CF, Tsai HY, Chen FH, Chiang CS. Targeting M-MDSCs enhances the therapeutic effect of BNCT in the 4-NQO-induced murine head and neck squamous cell carcinoma model. Front Oncol 2023; 13:1263873. [PMID: 37886177 PMCID: PMC10598372 DOI: 10.3389/fonc.2023.1263873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023] Open
Abstract
Purpose Malignant head and neck squamous cell carcinoma (HNSCC) is characterized by a poor prognosis and resistance to conventional radiotherapy. Infiltrating myeloid-derived suppressive cells (MDSCs) is prominent in HNSCC and is linked to immune suppression and tumor aggressiveness. This study aimed to investigate the impact of boron neutron capture therapy (BNCT) on the MDSCs in the tumor microenvironment and peripheral blood and to explore the potential for MDSCs depletion combined with BNCT to reactivate antitumor immunity. Methods and materials Carcinogen, 4-NQO, -induced oral tumors were irradiated with a total physical dose of 2 Gy BNCT in Tsing Hua Open Reactor (THOR). Flow cytometry and immunohistochemistry accessed the dynamics of peripheral MDSCs and infiltrated MDSCs within the tumor microenvironment. Mice were injected with an inhibitor of CSF-1 receptor (CSF-1R), PLX3397, to determine whether modulating M-MDSCs could affect mice survival after BNCT. Results Peripheral CD11b+Ly6ChighLy6G- monocytic-MDSCs (M-MDSCs), but not CD11b+Ly6CloLy6Ghigh polymorphonuclear-MDSCs (PMN-MDSCs), increased as tumor progression. After BNCT treatment, there were temporarily decreased and persistent increases of M-MDSCs thereafter, either in peripheral blood or in tumors. The administration of PLX-3397 hindered BNCT-caused M-MDSCs infiltration, prolonged mice survival, and activated tumor immunity by decreasing tumor-associated macrophages (TAMs) and increasing CD8+ T cells. Conclusion M-MDSCs were recruited into 4-NQO-induced tumors after BNCT, and their number was also increased in peripheral blood. Assessment of M-MDSCs levels in peripheral blood could be an index to determine the optimal intervention window. Their temporal alteration suggests an association with tumor recurrence after BNCT, making M-MDSCs a potential intervention target. Our preliminary results showed that PLX-3397 had strong M-MDSCs, TAMs, and TIL (tumor-infiltrating lymphocyte) modulating effects that could synergize tumor control when combined with BNCT.
Collapse
Affiliation(s)
- Chun-Hsiang Chang
- Department of Biomedical Engineering and Environment Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Jui Chen
- Department of Biomedical Engineering and Environment Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Fang Yu
- Institute for Radiological Research, Chang Gung University, Taoyuan, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan, Taiwan
| | - Hui-Yu Tsai
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Fang-Hsin Chen
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environment Sciences, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
9
|
Matos AI, Peres C, Carreira B, Moura LIF, Acúrcio RC, Vogel T, Wegener E, Ribeiro F, Afonso MB, Santos FMF, Martínez‐Barriocanal Á, Arango D, Viana AS, Góis PMP, Silva LC, Rodrigues CMP, Graca L, Jordan R, Satchi‐Fainaro R, Florindo HF. Polyoxazoline-Based Nanovaccine Synergizes with Tumor-Associated Macrophage Targeting and Anti-PD-1 Immunotherapy against Solid Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300299. [PMID: 37434063 PMCID: PMC10477894 DOI: 10.1002/advs.202300299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/22/2023] [Indexed: 07/13/2023]
Abstract
Immune checkpoint blockade reaches remarkable clinical responses. However, even in the most favorable cases, half of these patients do not benefit from these therapies in the long term. It is hypothesized that the activation of host immunity by co-delivering peptide antigens, adjuvants, and regulators of the transforming growth factor (TGF)-β expression using a polyoxazoline (POx)-poly(lactic-co-glycolic) acid (PLGA) nanovaccine, while modulating the tumor-associated macrophages (TAM) function within the tumor microenvironment (TME) and blocking the anti-programmed cell death protein 1 (PD-1) can constitute an alternative approach for cancer immunotherapy. POx-Mannose (Man) nanovaccines generate antigen-specific T-cell responses that control tumor growth to a higher extent than poly(ethylene glycol) (PEG)-Man nanovaccines. This anti-tumor effect induced by the POx-Man nanovaccines is mediated by a CD8+ -T cell-dependent mechanism, in contrast to the PEG-Man nanovaccines. POx-Man nanovaccine combines with pexidartinib, a modulator of the TAM function, restricts the MC38 tumor growth, and synergizes with PD-1 blockade, controlling MC38 and CT26 tumor growth and survival. This data is further validated in the highly aggressive and poorly immunogenic B16F10 melanoma mouse model. Therefore, the synergistic anti-tumor effect induced by the combination of nanovaccines with the inhibition of both TAM- and PD-1-inducing immunosuppression, holds great potential for improving immunotherapy outcomes in solid cancer patients.
Collapse
Affiliation(s)
- Ana I. Matos
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Carina Peres
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Barbara Carreira
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Liane I. F. Moura
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Rita C. Acúrcio
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Theresa Vogel
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Erik Wegener
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Filipa Ribeiro
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Marta B. Afonso
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Fábio M. F. Santos
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Águeda Martínez‐Barriocanal
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d'Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
- Group of Molecular OncologyLleida Biomedical Research Institute (IRBLleida)Lleida25198Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d'Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
- Group of Molecular OncologyLleida Biomedical Research Institute (IRBLleida)Lleida25198Spain
| | - Ana S. Viana
- Centro de Química EstruturalDepartamento de Química e BioquímicaInstitute of Molecular SciencesFaculty of SciencesUniversidade de LisboaLisbon1749‐016Portugal
| | - Pedro M. P. Góis
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Liana C. Silva
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Cecília M. P. Rodrigues
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Luis Graca
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Rainer Jordan
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Ronit Satchi‐Fainaro
- Department of Physiology and PharmacologyFaculty of MedicineSagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
| | - Helena F. Florindo
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| |
Collapse
|
10
|
van Elsas MJ, Labrie C, Etzerodt A, Charoentong P, van Stigt Thans JJC, Van Hall T, van der Burg SH. Invasive margin tissue-resident macrophages of high CD163 expression impede responses to T cell-based immunotherapy. J Immunother Cancer 2023; 11:jitc-2022-006433. [PMID: 36914207 PMCID: PMC10016286 DOI: 10.1136/jitc-2022-006433] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Primary and secondary resistance is a major hurdle in cancer immunotherapy. Therefore, a better understanding of the underlying mechanisms involved in immunotherapy resistance is of pivotal importance to improve therapy outcome. METHOD Here, two mouse models with resistance against therapeutic vaccine-induced tumor regression were studied. Exploration of the tumor microenvironment by high dimensional flow cytometry in combination with therapeutic in vivo settings allowed for the identification of immunological factors driving immunotherapy resistance. RESULTS Comparison of the tumor immune infiltrate during early and late regression revealed a change from tumor-rejecting toward tumor-promoting macrophages. In concert, a rapid exhaustion of tumor-infiltrating T cells was observed. Perturbation studies identified a small but discernible CD163hi macrophage population, with high expression of several tumor-promoting macrophage markers and a functional anti-inflammatory transcriptome profile, but not other macrophages, to be responsible. In-depth analyses revealed that they localize at the tumor invasive margins and are more resistant to Csf1r inhibition when compared with other macrophages. In vivo studies validated the activity of heme oxygenase-1 as an underlying mechanism of immunotherapy resistance. The transcriptomic profile of CD163hi macrophages is highly similar to a human monocyte/macrophage population, indicating that they represent a target to improve immunotherapy efficacy. CONCLUSIONS In this study, a small population of CD163hi tissue-resident macrophages is identified to be responsible for primary and secondary resistance against T-cell-based immunotherapies. While these CD163hi M2 macrophages are resistant to Csf1r-targeted therapies, in-depth characterization and identification of the underlying mechanisms driving immunotherapy resistance allows the specific targeting of this subset of macrophages, thereby creating new opportunities for therapeutic intervention with the aim to overcome immunotherapy resistance.
Collapse
Affiliation(s)
- Marit J van Elsas
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Camilla Labrie
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Pornpimol Charoentong
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jordi J C van Stigt Thans
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Thorbald Van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
11
|
Kazakova A, Sudarskikh T, Kovalev O, Kzhyshkowska J, Larionova I. Interaction of tumor‑associated macrophages with stromal and immune components in solid tumors: Research progress (Review). Int J Oncol 2023; 62:32. [PMID: 36660926 PMCID: PMC9851132 DOI: 10.3892/ijo.2023.5480] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023] Open
Abstract
Tumor‑associated macrophages (TAMs) are crucial cells of the tumor microenvironment (TME), which belong to the innate immune system and regulate primary tumor growth, immunosuppression, angiogenesis, extracellular matrix remodeling and metastasis. The review discusses current knowledge of essential cell‑cell interactions of TAMs within the TME of solid tumors. It summarizes the mechanisms of stromal cell (including cancer‑associated fibroblasts and endothelial cells)‑mediated monocyte recruitment and regulation of differentiation, as well as pro‑tumor and antitumor polarization of TAMs. Additionally, it focuses on the perivascular TAM subpopulations that regulate angiogenesis and lymphangiogenesis. It describes the possible mechanisms of reciprocal interactions of TAMs with other immune cells responsible for immunosuppression. Finally, it highlights the perspectives for novel therapeutic approaches to use combined cellular targets that include TAMs and other stromal and immune cells in the TME. The collected data demonstrated the importance of understanding cell‑cell interactions in the TME to prevent distant metastasis and reduce the risk of tumor recurrence.
Collapse
Affiliation(s)
- Anna Kazakova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk 634050, Russian Federation
| | - Tatiana Sudarskikh
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk 634050, Russian Federation
| | - Oleg Kovalev
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russian Federation
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk 634050, Russian Federation
- Institute of Transfusion Medicine and Immunology, Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk 634050, Russian Federation
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russian Federation
| |
Collapse
|
12
|
Lee S, Karns R, Shin S. Mechanism of paracrine communications between hepatic progenitor cells and endothelial cells. Cell Signal 2022; 100:110458. [PMID: 36055565 PMCID: PMC9971365 DOI: 10.1016/j.cellsig.2022.110458] [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: 05/31/2022] [Revised: 08/16/2022] [Accepted: 08/25/2022] [Indexed: 11/27/2022]
Abstract
Hepatic progenitor cells (HPCs) are facultative tissue-specific stem cells lining reactive ductules, which are ubiquitously observed in chronic liver diseases and cancer. Although previous research mainly focused on their contribution to liver regeneration, it turned out that in vivo differentiation of HPCs into hepatocytes only occurs after extreme injury. While recent correlative evidence implies the association of HPCs with disease progression, their exact role in pathogenesis remains largely unknown. Our previous research demonstrated that HPCs expressing angiogenic paracrine factors accumulate in the peritumoral area and are positively correlated with the extent of intratumoral cell proliferation and angiogenesis in the livers of patients with liver cancer. Given the crucial roles of angiogenesis in liver disease progression and carcinogenesis, we aimed to test the hypothesis that HPCs secrete paracrine factors to communicate with endothelial cells, to determine molecular mechanisms mediating HPCs-endothelial interactions, and to understand how the paracrine function of HPCs is regulated. HPCs promoted viability and tubulogenesis of human umbilical vein endothelial cells (HUVECs) and upregulated genes known to be involved in angiogenesis, endothelial cell function, and disease progression in a paracrine manner. The paracrine function of HPCs as well as expression of colony stimulating factor 1 (CSF1) were inhibited upon differentiation of HPCs toward hepatocytes. Inhibition of CSF1 receptor partly suppressed the paracrine effects of HPCs on HUVECs. Taken together, our study indicates that inhibition of the paracrine function of HPCs through modulation of their differentiation status and inhibition of CSF1 signaling is a promising strategy for inhibition of angiogenesis during pathological progression.
Collapse
Affiliation(s)
- Sanghoon Lee
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Rebekah Karns
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Soona Shin
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
13
|
Joshi S, Sharabi A. Targeting myeloid-derived suppressor cells to enhance natural killer cell-based immunotherapy. Pharmacol Ther 2022; 235:108114. [DOI: 10.1016/j.pharmthera.2022.108114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 12/09/2022]
|
14
|
Wang JC, Sun L. PD-1/PD-L1, MDSC Pathways, and Checkpoint Inhibitor Therapy in Ph(-) Myeloproliferative Neoplasm: A Review. Int J Mol Sci 2022; 23:5837. [PMID: 35628647 PMCID: PMC9143160 DOI: 10.3390/ijms23105837] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/23/2022] Open
Abstract
There has been significant progress in immune checkpoint inhibitor (CPI) therapy in many solid tumor types. However, only a single failed study has been published in treating Ph(-) myeloproliferative neoplasm (MPN). To make progress in CPI studies on this disease, herein, we review and summarize the mechanisms of activation of the PD-L1 promoter, which are as follows: (a) the extrinsic mechanism, which is activated by interferon gamma (IFN γ) by tumor infiltration lymphocytes (TIL) and NK cells; (b) the intrinsic mechanism of EGFR or PTEN loss resulting in the activation of the MAPK and AKT pathways and then stat 1 and 3 activation; and (c) 9p24 amplicon amplification, resulting in PD-L1 and Jak2 activation. We also review the literature and postulate that many of the failures of CPI therapy in MPN are likely due to excessive MDSC activities. We list all of the anti-MDSC agents, especially those with ruxolitinib, IMID compounds, and BTK inhibitors, which may be combined with CPI therapy in the future as part of clinical trials applying CPI therapy to Ph(-) MPN.
Collapse
Affiliation(s)
- Jen-Chin Wang
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA;
| | | |
Collapse
|
15
|
Xu Y, Wang X, Liu L, Wang J, Wu J, Sun C. Role of macrophages in tumor progression and therapy (Review). Int J Oncol 2022; 60:57. [PMID: 35362544 PMCID: PMC8997338 DOI: 10.3892/ijo.2022.5347] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
The number and phenotype of macrophages are closely related to tumor growth and prognosis. Macrophages are recruited to (and polarized at) the tumor site thereby promoting tumor growth, stimulating tumor angiogenesis, facilitating tumor cell migration, and creating a favorable environment for subsequent colonization by (and survival of) tumor cells. These phenomena contribute to the formation of an immunosuppressive tumor microenvironment (TME) and therefore speed up tumor cell proliferation and metastasis and reduce the efficacy of antitumor factors and therapies. The ability of macrophages to remodel the TME through interactions with other cells and corresponding changes in their number, activity, and phenotype during conventional therapies, as well as the association between these changes and drug resistance, make tumor-associated macrophages a new target for antitumor therapies. In this review, advantages and limitations of the existing antitumor strategies targeting macrophages in Traditional Chinese and Western medicine were analyzed, starting with the effect of macrophages on tumors and their interactions with other cells and then the role of macrophages in conventional treatments was explored. Possible directions of future developments in this field from an all-around multitarget standpoint were also examined.
Collapse
Affiliation(s)
- Yiwei Xu
- Institute of Integrated Medicine, School of Medicine, Qingdao University, Qingdao, Shandong 266073, P.R. China
| | - Xiaomin Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261041, P.R. China
| | - Jia Wang
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
16
|
Deng J, Fleming JB. Inflammation and Myeloid Cells in Cancer Progression and Metastasis. Front Cell Dev Biol 2022; 9:759691. [PMID: 35127700 PMCID: PMC8814460 DOI: 10.3389/fcell.2021.759691] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Abstract
To date, the most immunotherapy drugs act upon T cell surface proteins to promote tumoricidal T cell activity. However, this approach has to date been unsuccessful in certain solid tumor types including pancreatic, prostate cancer and glioblastoma. Myeloid-related innate immunity can promote tumor progression through direct and indirect effects on T cell activity; improved understanding of this field may provide another therapeutic avenue for patients with these tumors. Myeloid cells can differentiate into both pro-inflammatory and anti-inflammatory mature form depending upon the microenvironment. Most cancer type exhibit oncogenic activating point mutations (ex. P53 and KRAS) that trigger cytokines production. In addition, tumor environment (ex. Collagen, Hypoxia, and adenosine) also regulated inflammatory signaling cascade. Both the intrinsic and extrinsic factor driving the tumor immune microenvironment and regulating the differentiation and function of myeloid cells, T cells activity and tumor progression. In this review, we will discuss the relationship between cancer cells and myeloid cells-mediated tumor immune microenvironment to promote cancer progression and immunotherapeutic resistance. Furthermore, we will describe how cytokines and chemokines produced by cancer cells influence myeloid cells within immunosuppressive environment. Finally, we will comment on the development of immunotherapeutic strategies with respect to myeloid-related innate immunity.
Collapse
Affiliation(s)
- Jenying Deng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jason B. Fleming
- H. Lee Moffitt Cancer Center, Department of Gastrointestinal Oncology, Tampa, FL, United States
- *Correspondence: Jason B. Fleming,
| |
Collapse
|
17
|
Chu H, He QX, Wang J, Hu Y, Wang YQ, Lin ZH. In silico design novel dihydropyrimio[4, 5-d]pyrimidine derivatives as inhibitors for colony-stimulating factor-1 receptor based on 3D-QSAR, molecular docking and molecular dynamics simulation. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
18
|
Yang Q, Guo N, Zhou Y, Chen J, Wei Q, Han M. The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy. Acta Pharm Sin B 2020; 10:2156-2170. [PMID: 33304783 PMCID: PMC7714989 DOI: 10.1016/j.apsb.2020.04.004] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Macrophages have a leading position in the tumor microenvironment (TME) which paves the way to carcinogenesis. Initially, monocytes and macrophages are recruited to the sites where the tumor develops. Under the guidance of different microenvironmental signals, macrophages would polarize into two functional phenotypes, named as classically activated macrophages (M1) and alternatively activated macrophages (M2). Contrary to the anti-tumor effect of M1, M2 exerts anti-inflammatory and tumorigenic characters. In progressive tumor, M2 tumor-associated macrophages (TAMs) are in the majority, being vital regulators reacting upon TME. This review elaborates on the role of TAMs in tumor progression. Furthermore, prospective macrophage-focused therapeutic strategies, including drugs not only in clinical trials but also at primary research stages, are summarized followed by a discussion about their clinical application values. Nanoparticulate systems with efficient drug delivery and improved antitumor effect are also summed up in this article.
Collapse
Affiliation(s)
- Qiyao Yang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ningning Guo
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Zhou
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiejian Chen
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qichun Wei
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
19
|
Wu K, Lin K, Li X, Yuan X, Xu P, Ni P, Xu D. Redefining Tumor-Associated Macrophage Subpopulations and Functions in the Tumor Microenvironment. Front Immunol 2020; 11:1731. [PMID: 32849616 PMCID: PMC7417513 DOI: 10.3389/fimmu.2020.01731] [Citation(s) in RCA: 438] [Impact Index Per Article: 87.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/29/2020] [Indexed: 12/25/2022] Open
Abstract
The immunosuppressive status of the tumor microenvironment (TME) remains poorly defined due to a lack of understanding regarding the function of tumor-associated macrophages (TAMs), which are abundant in the TME. TAMs are crucial drivers of tumor progression, metastasis, and resistance to therapy. Intra- and inter-tumoral spatial heterogeneities are potential keys to understanding the relationships between subpopulations of TAMs and their functions. Antitumor M1-like and pro-tumor M2-like TAMs coexist within tumors, and the opposing effects of these M1/M2 subpopulations on tumors directly impact current strategies to improve antitumor immune responses. Recent studies have found significant differences among monocytes or macrophages from distinct tumors, and other investigations have explored the existence of diverse TAM subsets at the molecular level. In this review, we discuss emerging evidence highlighting the redefinition of TAM subpopulations and functions in the TME and the possibility of separating macrophage subsets with distinct functions into antitumor M1-like and pro-tumor M2-like TAMs during the development of tumors. Such redefinition may relate to the differential cellular origin and monocyte and macrophage plasticity or heterogeneity of TAMs, which all potentially impact macrophage biomarkers and our understanding of how the phenotypes of TAMs are dictated by their ontogeny, activation status, and localization. Therefore, the detailed landscape of TAMs must be deciphered with the integration of new technologies, such as multiplexed immunohistochemistry (mIHC), mass cytometry by time-of-flight (CyTOF), single-cell RNA-seq (scRNA-seq), spatial transcriptomics, and systems biology approaches, for analyses of the TME.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
20
|
Hashimoto A, Fukumoto T, Zhang R, Gabrilovich D. Selective targeting of different populations of myeloid-derived suppressor cells by histone deacetylase inhibitors. Cancer Immunol Immunother 2020; 69:1929-1936. [PMID: 32435850 DOI: 10.1007/s00262-020-02588-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are widely implicated in negative regulation of immune responses in cancer. Inhibition of class I histone deacetylases (HDAC) with entinostat has anti-MDSC activity. However, as single agent, it did not delay tumor growth in EL4 and LLC tumor models. Here, we found that entinostat reduced immune suppressive activity of only one type of MDSC-polymorphonuclear, PMN-MDSC, whereas it had no effect on monocytic M-MDSC or macrophages. M-MDSC had high amount of class II HDAC-HDAC6, which was further increased after the treatment of mice with entinostat. Inhibition of HDAC6 with ricolinostat reduced suppressive activity of M-MDSC, but did not affect PMN-MDSC or delayed tumor growth. However, combination of entinostat and ricolinostat abrogated suppressive activity of both populations of MDSC and substantially delayed tumor progression. Thus, inactivation of MDSC required targeting of both major subsets of these cells via inhibitors of class I and class II HDAC.
Collapse
Affiliation(s)
- Ayumi Hashimoto
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Rm. 118, 3601 Spruce Str., Philadelphia, PA, 19104, USA
| | - Takeshi Fukumoto
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Rugang Zhang
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Dmitry Gabrilovich
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Rm. 118, 3601 Spruce Str., Philadelphia, PA, 19104, USA.
| |
Collapse
|
21
|
Li X, Fu Y, Yang B, Guo E, Wu Y, Huang J, Zhang X, Xiao R, Li K, Wang B, Hu J, Sun C, Chen G. BRD4 Inhibition by AZD5153 Promotes Antitumor Immunity via Depolarizing M2 Macrophages. Front Immunol 2020; 11:89. [PMID: 32184777 PMCID: PMC7058627 DOI: 10.3389/fimmu.2020.00089] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/14/2020] [Indexed: 01/13/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC), with its high recurrence rates, urges for reasonable therapeutic strategies that can prolong overall survival. A tumor microenvironment (TME) discloses prognostic and prospective information on cancer, such as the expression level of PD-1 or PD-L1. However, in HGSOC, the impact of the therapies aiming at these targets remains unsatisfying. Tumor-associated macrophages (TAMs) in HGSOC make up a large part of the TMEs and transform between diverse phenotypes under different treatments. AZD5153 inhibiting BRD4, as a potential therapeutic strategy for HGSOC, was demonstrated to confer controversial plasticity on TAMs, which shows the need to uncover its impact on TAMs in HGSOC. Therefore, we established models for TAMs and TAMs co-culturing with T lymphocytes in vitro. Via RT-PCR and flow cytometry, we find that AZD5153 resets TAMs from M2-type macrophages to M1-like macrophages, consequently promoting pro-inflammatory cytokine secretion and thus activating CD8+ cytotoxic T lymphocytes (CTLs) in vitro. This modification occurs on MAF transcripts in TAMs and modified by BRD4, which is the target of AZD5153. Importantly, the 3-D microfluid model demonstrates that AZD5153 sensitizes ovarian cancer to anti-PD-L1 therapy. Our results clarify that besides eliminating tumor cells directly, AZD5153 works as a regulator of the TAM phenotype, providing a novel strategy combining AZD5153 and PD-1/PD-L1 antibody that could benefit HGSOC patients.
Collapse
Affiliation(s)
- Xi Li
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Fu
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Yang
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ensong Guo
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Wu
- Department of Gynecology and Obstetrics, The Central Hospital of Wuhan, Wuhan, China
| | - Jia Huang
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Zhang
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Rourou Xiao
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Kezhen Li
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Beibei Wang
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Junbo Hu
- Department of Gastrointestinal Surgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyang Sun
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Chen
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Law AMK, Valdes-Mora F, Gallego-Ortega D. Myeloid-Derived Suppressor Cells as a Therapeutic Target for Cancer. Cells 2020; 9:cells9030561. [PMID: 32121014 PMCID: PMC7140518 DOI: 10.3390/cells9030561] [Citation(s) in RCA: 294] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/22/2020] [Accepted: 02/24/2020] [Indexed: 12/15/2022] Open
Abstract
The emergence of immunotherapy has been an astounding breakthrough in cancer treatments. In particular, immune checkpoint inhibitors, targeting PD-1 and CTLA-4, have shown remarkable therapeutic outcomes. However, response rates from immunotherapy have been reported to be varied, with some having pronounced success and others with minimal to no clinical benefit. An important aspect associated with this discrepancy in patient response is the immune-suppressive effects elicited by the tumour microenvironment (TME). Immune suppression plays a pivotal role in regulating cancer progression, metastasis, and reducing immunotherapy success. Most notably, myeloid-derived suppressor cells (MDSC), a heterogeneous population of immature myeloid cells, have potent mechanisms to inhibit T-cell and NK-cell activity to promote tumour growth, development of the pre-metastatic niche, and contribute to resistance to immunotherapy. Accumulating research indicates that MDSC can be a therapeutic target to alleviate their pro-tumourigenic functions and immunosuppressive activities to bolster the efficacy of checkpoint inhibitors. In this review, we provide an overview of the general immunotherapeutic approaches and discuss the characterisation, expansion, and activities of MDSCs with the current treatments used to target them either as a single therapeutic target or synergistically in combination with immunotherapy.
Collapse
Affiliation(s)
- Andrew M. K. Law
- Tumour Development Group, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- Correspondence: (A.M.K.L.); (F.V.-M.); (D.G.-O.); Tel.: +61-(0)2-9355-5894 (A.M.K.L); +61-(0)2-9385-0143 (F.V.-M); +61-(0)2-9355-5776 (D.G.-O)
| | - Fatima Valdes-Mora
- Histone Variants Group, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
- Correspondence: (A.M.K.L.); (F.V.-M.); (D.G.-O.); Tel.: +61-(0)2-9355-5894 (A.M.K.L); +61-(0)2-9385-0143 (F.V.-M); +61-(0)2-9355-5776 (D.G.-O)
| | - David Gallego-Ortega
- Tumour Development Group, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
- Correspondence: (A.M.K.L.); (F.V.-M.); (D.G.-O.); Tel.: +61-(0)2-9355-5894 (A.M.K.L); +61-(0)2-9385-0143 (F.V.-M); +61-(0)2-9355-5776 (D.G.-O)
| |
Collapse
|
23
|
Erkes DA, Rosenbaum SR, Field CO, Chervoneva I, Villanueva J, Aplin AE. PLX3397 inhibits the accumulation of intra-tumoral macrophages and improves bromodomain and extra-terminal inhibitor efficacy in melanoma. Pigment Cell Melanoma Res 2019; 33:372-377. [PMID: 31696640 DOI: 10.1111/pcmr.12845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/25/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023]
Abstract
Bromodomain and extra-terminal inhibitors (BETi) delay tumor growth, in part, through tumor cell intrinsic alterations and initiation of anti-tumor CD8+ T-cell responses. By contrast, BETi effects on pro-tumoral immune responses remain unclear. Here, we show that the next-generation BETi, PLX51107, delayed tumor growth to differing degrees in Braf V600E melanoma syngeneic mouse models. These differential responses were associated with the influx of tumor-associated macrophages during BETi treatment. Tumors that were poorly responsive to PLX51107 showed increased influx of colony-stimulating factor-1 receptor (CSF-1R)-positive tumor-associated macrophages. We depleted CSF-1R+ tumor-associated macrophages with the CSF-1R inhibitor, PLX3397, in combination with PLX51107. Treatment with PLX3397 enhanced the efficacy of PLX51107 in poorly responsive Braf V600E syngeneic melanomas in vivo. These findings suggest that tumor-associated macrophage accumulation limits BETi efficacy and that co-treatment with PLX3397 can improve response to PLX51107, offering a potential novel combination therapy for metastatic melanoma patients.
Collapse
Affiliation(s)
- Dan A Erkes
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sheera R Rosenbaum
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Conroy O Field
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Inna Chervoneva
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jessie Villanueva
- Molecular and Cellular Oncogenesis Program, Melanoma Research Center, The Wistar Institute, PA, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
24
|
Shi Y, Manis M, Long J, Wang K, Sullivan PM, Remolina Serrano J, Hoyle R, Holtzman DM. Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. J Exp Med 2019; 216:2546-2561. [PMID: 31601677 PMCID: PMC6829593 DOI: 10.1084/jem.20190980] [Citation(s) in RCA: 285] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 07/21/2019] [Accepted: 08/08/2019] [Indexed: 11/16/2022] Open
Abstract
Shi et al. find that microglia, instead of tau-induced direct neurotoxicity, are the driving force of neurodegeneration in a tauopathy mouse model. Microglia are also required for tau pathogenesis. In addition, apoE strongly regulates neurodegeneration in the setting of tauopathy predominantly by modulating microglial function. Chronic activation of brain innate immunity is a prominent feature of Alzheimer’s disease (AD) and primary tauopathies. However, to what degree innate immunity contributes to neurodegeneration as compared with pathological protein-induced neurotoxicity, and the requirement of a particular glial cell type in neurodegeneration, are still unclear. Here we demonstrate that microglia-mediated damage, rather than pathological tau-induced direct neurotoxicity, is the leading force driving neurodegeneration in a tauopathy mouse model. Importantly, the progression of ptau pathology is also driven by microglia. In addition, we found that APOE, the strongest genetic risk factor for AD, regulates neurodegeneration predominantly by modulating microglial activation, although a minor role of apoE in regulating ptau and insoluble tau formation independent of its immunomodulatory function was also identified. Our results suggest that therapeutic strategies targeting microglia may represent an effective approach to prevent disease progression in the setting of tauopathy.
Collapse
Affiliation(s)
- Yang Shi
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - Melissa Manis
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - Justin Long
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - Kairuo Wang
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | | | - Javier Remolina Serrano
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - Rosa Hoyle
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
25
|
Neubert NJ, Schmittnaegel M, Bordry N, Nassiri S, Wald N, Martignier C, Tillé L, Homicsko K, Damsky W, Maby-El Hajjami H, Klaman I, Danenberg E, Ioannidou K, Kandalaft L, Coukos G, Hoves S, Ries CH, Fuertes Marraco SA, Foukas PG, De Palma M, Speiser DE. T cell-induced CSF1 promotes melanoma resistance to PD1 blockade. Sci Transl Med 2019; 10:10/436/eaan3311. [PMID: 29643229 DOI: 10.1126/scitranslmed.aan3311] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 12/15/2017] [Accepted: 02/21/2018] [Indexed: 12/12/2022]
Abstract
Colony-stimulating factor 1 (CSF1) is a key regulator of monocyte/macrophage differentiation that sustains the protumorigenic functions of tumor-associated macrophages (TAMs). We show that CSF1 is expressed in human melanoma, and patients with metastatic melanoma have increased CSF1 in blood compared to healthy subjects. In tumors, CSF1 expression correlated with the abundance of CD8+ T cells and CD163+ TAMs. Human melanoma cell lines consistently produced CSF1 after exposure to melanoma-specific CD8+ T cells or T cell-derived cytokines in vitro, reflecting a broadly conserved mechanism of CSF1 induction by activated CD8+ T cells. Mining of publicly available transcriptomic data sets suggested co-enrichment of CD8+ T cells with CSF1 or various TAM-specific markers in human melanoma, which was associated with nonresponsiveness to programmed cell death protein 1 (PD1) checkpoint blockade in a smaller patient cohort. Combination of anti-PD1 and anti-CSF1 receptor (CSF1R) antibodies induced the regression of BRAFV600E -driven, transplant mouse melanomas, a result that was dependent on the effective elimination of TAMs. Collectively, these data implicate CSF1 induction as a CD8+ T cell-dependent adaptive resistance mechanism and show that simultaneous CSF1R targeting may be beneficial in melanomas refractory to immune checkpoint blockade and, possibly, other T cell-based therapies.
Collapse
Affiliation(s)
- Natalie J Neubert
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland
| | - Martina Schmittnaegel
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Natacha Bordry
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland
| | - Sina Nassiri
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Noémie Wald
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland
| | - Christophe Martignier
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland
| | - Laure Tillé
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland
| | - Krisztian Homicsko
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland.,Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - William Damsky
- Departments of Dermatology and Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Hélène Maby-El Hajjami
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland
| | - Irina Klaman
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Esther Danenberg
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), CH-1005 Lausanne, Switzerland
| | - Kalliopi Ioannidou
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland
| | - Lana Kandalaft
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), CH-1005 Lausanne, Switzerland
| | - George Coukos
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland.,Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), CH-1005 Lausanne, Switzerland
| | - Sabine Hoves
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Carola H Ries
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Silvia A Fuertes Marraco
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland
| | - Periklis G Foukas
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), CH-1005 Lausanne, Switzerland
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Daniel E Speiser
- Ludwig Cancer Research Center and Department of Oncology, University of Lausanne (UNIL), CH-1066 Epalinges, Switzerland.
| |
Collapse
|
26
|
Peng X, Hou P, Chen Y, Dai Y, Ji Y, Shen Y, Su Y, Liu B, Wang Y, Sun D, Jiang Y, Zha C, Xie Z, Ding J, Geng M, Ai J. Preclinical evaluation of 3D185, a novel potent inhibitor of FGFR1/2/3 and CSF-1R, in FGFR-dependent and macrophage-dominant cancer models. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:372. [PMID: 31438996 PMCID: PMC6704710 DOI: 10.1186/s13046-019-1357-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/06/2019] [Indexed: 12/25/2022]
Abstract
Background The interaction between tumor cells and their immunosuppressive microenvironment promotes tumor progression and drug resistance. Thus, simultaneously targeting tumor cells and stromal cells is expected to have synergistic antitumor effects. Herein, we present for the first time a preclinical antitumor investigation of 3D185, a novel dual inhibitor targeting FGFRs, which are oncogenic drivers, and CSF-1R, which is the major survival factor for protumor macrophages. Methods The antitumor characteristics of 3D185 were assessed by a range of assays, including kinase profiling, cell viability, cell migration, immunoblotting, CD8+ T cell suppression, and in vivo antitumor efficacy, followed by flow cytometric and immunohistochemical analyses of tumor-infiltrating immune cells and endothelial cells in nude mice and immune-competent mice. Results 3D185 significantly inhibited the kinase activity of FGFR1/2/3 and CSF-1R, with equal potency and high selectivity over other kinases. 3D185 suppressed FGFR signaling and tumor cell growth in FGFR-driven models both in vitro and in vivo. In addition, 3D185 could inhibit the survival and M2-like polarization of macrophages, reversing the immunosuppressive effect of macrophages on CD8+ T cells as well as CSF1-differentiated macrophage induced-FGFR3-aberrant cancer cell migration. Furthermore, 3D185 inhibited tumor growth via remodeling the tumor microenvironment in TAM-dominated tumor models. Conclusions 3D185 is a promising antitumor candidate drug that simultaneously targets tumor cells and their immunosuppressive microenvironment and has therapeutic potential due to synergistic effects. Our study provides a solid foundation for the investigation of 3D185 in cancer patients, particularly in patients with aberrant FGFR and abundant macrophages, who respond poorly to classic pan-FGFRi treatment. Electronic supplementary material The online version of this article (10.1186/s13046-019-1357-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xia Peng
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Pengcong Hou
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yi Chen
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yang Dai
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yinchun Ji
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yanyan Shen
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yi Su
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Bo Liu
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yueliang Wang
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Deqiao Sun
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yuchen Jiang
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Chuantao Zha
- Shanghai HaiHe Pharmaceutical Co., Ltd, No. 421 Newton Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Zuoquan Xie
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Jian Ding
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Meiyu Geng
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China. .,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China.
| | - Jing Ai
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China. .,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China.
| |
Collapse
|
27
|
Kim MY, Lee K, Shin HI, Jeong D. Specific targeting of PKCδ suppresses osteoclast differentiation by accelerating proteolysis of membrane-bound macrophage colony-stimulating factor receptor. Sci Rep 2019; 9:7044. [PMID: 31065073 PMCID: PMC6504882 DOI: 10.1038/s41598-019-43501-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/25/2019] [Indexed: 01/12/2023] Open
Abstract
c-Fms is the macrophage colony-stimulating factor (M-CSF) receptor, and intracellular signalling via the M-CSF/c-Fms axis mediates both innate immunity and bone remodelling. M-CSF-induced transient proteolytic degradation of c-Fms modulates various biological functions, and protein kinase C (PKC) signalling is activated during this proteolytic process via an unknown mechanism. Notably, the role of specific PKC isoforms involved in c-Fms degradation during osteoclast differentiation is not known. Here, we observed that inactivation of PKCδ by the biochemical inhibitor rottlerin, a cell permeable peptide inhibitor, and short hairpin (sh) RNA suppresses osteoclast differentiation triggered by treatment with M-CSF and receptor activator of NF-κB ligand. Interestingly, inhibition of PKCδ by either inhibitor or gene silencing of PKCδ accelerated M-CSF-induced proteolytic degradation of membrane-bound c-Fms via both the lysosomal pathway and regulated intramembrane proteolysis (RIPping), but did not affect c-fms expression at the mRNA level. Degradation of c-Fms induced by PKCδ inactivation subsequently inhibited M-CSF-induced osteoclastogenic signals, such as extracellular signal-regulated kinase (ERK), c-JUN N-terminal kinase (JNK), p38, and Akt. Furthermore, mice administered PKCδ inhibitors into the calvaria periosteum exhibited a decrease in both osteoclast formation on the calvarial bone surface and the calvarial bone marrow cavity, which reflects osteoclastic bone resorption activity. These data suggest that M-CSF-induced PKCδ activation maintains membrane-anchored c-Fms and allows the sequential cellular events of osteoclastogenic signalling, osteoclast formation, and osteoclastic bone resorption.
Collapse
Affiliation(s)
- Mi Yeong Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Kyunghee Lee
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Hong-In Shin
- IHBR, Department of Oral Pathology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea
| | - Daewon Jeong
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, 42415, Korea.
| |
Collapse
|
28
|
Giricz O, Mo Y, Dahlman KB, Cotto-Rios XM, Vardabasso C, Nguyen H, Matusow B, Bartenstein M, Polishchuk V, Johnson DB, Bhagat TD, Shellooe R, Burton E, Tsai J, Zhang C, Habets G, Greally JM, Yu Y, Kenny PA, Fields GB, Pradhan K, Stanley ER, Bernstein E, Bollag G, Gavathiotis E, West BL, Sosman JA, Verma AK. The RUNX1/IL-34/CSF-1R axis is an autocrinally regulated modulator of resistance to BRAF-V600E inhibition in melanoma. JCI Insight 2018; 3:120422. [PMID: 30046005 PMCID: PMC6124424 DOI: 10.1172/jci.insight.120422] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/12/2018] [Indexed: 01/05/2023] Open
Abstract
Resistance to current therapies still impacts a significant number of melanoma patients and can be regulated by epigenetic alterations. Analysis of global cytosine methylation in a cohort of primary melanomas revealed a pattern of early demethylation associated with overexpression of oncogenic transcripts. Loss of methylation and associated overexpression of the CSF 1 receptor (CSF1R) was seen in a majority of tumors and was driven by an alternative, endogenous viral promoter in a subset of samples. CSF1R was particularly elevated in melanomas with BRAF and other MAPK activating mutations. Furthermore, rebound ERK activation after BRAF inhibition was associated with RUNX1-mediated further upregulation of CSF-1R and its ligand IL-34. Importantly, increased CSF-1R and IL-34 overexpression were detected in an independent cohort of resistant melanomas. Inhibition of CSF-1R kinase or decreased CSF-1R expression by RNAi reduced 3-D growth and invasiveness of melanoma cells. Coinhibition of CSF-1R and BRAF resulted in synergistic efficacy in vivo. To our knowledge, our data unveil a previously unknown role for the autocrine-regulated CSF-1R in BRAF V600E resistance and provide a preclinical rationale for targeting this pathway in melanoma.
Collapse
Affiliation(s)
- Orsi Giricz
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Yongkai Mo
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | | | | | - Chiara Vardabasso
- Departments of Oncological Sciences & Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Matthias Bartenstein
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Veronika Polishchuk
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | | | - Tushar D. Bhagat
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | | | | | | | | | | | | | - Yiting Yu
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Paraic A. Kenny
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Gregg B. Fields
- Department of Chemistry and Biochemistry, Florida Atlantic University, Florida, USA
| | - Kith Pradhan
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Emily Bernstein
- Departments of Oncological Sciences & Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Evripidis Gavathiotis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | | | - Amit K. Verma
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| |
Collapse
|
29
|
Gyori D, Lim EL, Grant FM, Spensberger D, Roychoudhuri R, Shuttleworth SJ, Okkenhaug K, Stephens LR, Hawkins PT. Compensation between CSF1R+ macrophages and Foxp3+ Treg cells drives resistance to tumor immunotherapy. JCI Insight 2018; 3:120631. [PMID: 29875321 PMCID: PMC6124419 DOI: 10.1172/jci.insight.120631] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/01/2018] [Indexed: 12/31/2022] Open
Abstract
Redundancy and compensation provide robustness to biological systems but may contribute to therapy resistance. Both tumor-associated macrophages (TAMs) and Foxp3+ regulatory T (Treg) cells promote tumor progression by limiting antitumor immunity. Here we show that genetic ablation of CSF1 in colorectal cancer cells reduces the influx of immunosuppressive CSF1R+ TAMs within tumors. This reduction in CSF1-dependent TAMs resulted in increased CD8+ T cell attack on tumors, but its effect on tumor growth was limited by a compensatory increase in Foxp3+ Treg cells. Similarly, disruption of Treg cell activity through their experimental ablation produced moderate effects on tumor growth and was associated with elevated numbers of CSF1R+ TAMs. Importantly, codepletion of CSF1R+ TAMs and Foxp3+ Treg cells resulted in an increased influx of CD8+ T cells, augmentation of their function, and a synergistic reduction in tumor growth. Further, inhibition of Treg cell activity either through systemic pharmacological blockade of PI3Kδ, or its genetic inactivation within Foxp3+ Treg cells, sensitized previously unresponsive solid tumors to CSF1R+ TAM depletion and enhanced the effect of CSF1R blockade. These findings identify CSF1R+ TAMs and PI3Kδ-driven Foxp3+ Treg cells as the dominant compensatory cellular components of the immunosuppressive tumor microenvironment, with implications for the design of combinatorial immunotherapies.
Collapse
Affiliation(s)
- David Gyori
- Signalling ISP, Babraham Institute, Babraham Research Campus, Cambridge, Cambridgeshire, United Kingdom
| | - Ee Lyn Lim
- Signalling ISP, Babraham Institute, Babraham Research Campus, Cambridge, Cambridgeshire, United Kingdom
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Francis M. Grant
- Signalling ISP, Babraham Institute, Babraham Research Campus, Cambridge, Cambridgeshire, United Kingdom
| | - Dominik Spensberger
- Signalling ISP, Babraham Institute, Babraham Research Campus, Cambridge, Cambridgeshire, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, United Kingdom
| | - Rahul Roychoudhuri
- Signalling ISP, Babraham Institute, Babraham Research Campus, Cambridge, Cambridgeshire, United Kingdom
| | - Stephen J. Shuttleworth
- Karus Therapeutics Ltd., Genesis Building, Library Avenue, Harwell Campus, Oxfordshire, United Kingdom
| | - Klaus Okkenhaug
- Signalling ISP, Babraham Institute, Babraham Research Campus, Cambridge, Cambridgeshire, United Kingdom
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Len R. Stephens
- Signalling ISP, Babraham Institute, Babraham Research Campus, Cambridge, Cambridgeshire, United Kingdom
| | - Phillip T. Hawkins
- Signalling ISP, Babraham Institute, Babraham Research Campus, Cambridge, Cambridgeshire, United Kingdom
| |
Collapse
|
30
|
Interaction between astrocytic colony stimulating factor and its receptor on microglia mediates central sensitization and behavioral hypersensitivity in chronic post ischemic pain model. Brain Behav Immun 2018; 68:248-260. [PMID: 29080683 DOI: 10.1016/j.bbi.2017.10.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 02/05/2023] Open
Abstract
Accumulation of microglia occurs in the dorsal horn in the rodent model of chronic post ischemic pain (CPIP), while the mechanism how microglia affects the development of persistent pain largely remains unknown. Here, using a rodent model of CPIP induced by ischemia-reperfusion (IR) injury in the hindpaw, we observed that microglial accumulation occurred in the ipsilateral dorsal horn after ischemia 3h, and in ipsilateral and contralateral dorsal horn in the rats with ischemia 6h. The accumulated microglia released BDNF, increased neuronal excitability in dorsal horn, and produced pain behaviors in the modeled rodents. We also found significantly increased signaling mediated by astrocytic colony-stimulating factor-1 (CSF1) and microglial CSF1 receptor (CSF1R) in dorsal horn in the ischemia 6h modeled rats. While exogenous M-CSF induced microglial activation and proliferation, BDNF production, neuronal hyperactivity in dorsal horn and behavioral hypersensitivity in the naïve rats, inhibition of astrocytic CSF1/microglial CSF1R signaling by fluorocitric or PLX3397 significantly suppressed microglial activation and proliferation, BDNF upregulation, and neuronal activity in dorsal horn, as well as the mechanical allodynia and thermal hyperalgesia, in the rats with ischemia 6h. Collectively, these results demonstrated that glial CSF1/CSF1R pathway mediated the microglial activation and proliferation, which facilitated the nociceptive output and contributed to the chronic pain induced by IR injury.
Collapse
|
31
|
Kumar V, Donthireddy L, Marvel D, Condamine T, Wang F, Lavilla-Alonso S, Hashimoto A, Vonteddu P, Behera R, Goins MA, Mulligan C, Nam B, Hockstein N, Denstman F, Shakamuri S, Speicher DW, Weeraratna AT, Chao T, Vonderheide RH, Languino LR, Ordentlich P, Liu Q, Xu X, Lo A, Puré E, Zhang C, Loboda A, Sepulveda MA, Snyder LA, Gabrilovich DI. Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors. Cancer Cell 2017; 32:654-668.e5. [PMID: 29136508 PMCID: PMC5827952 DOI: 10.1016/j.ccell.2017.10.005] [Citation(s) in RCA: 476] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/25/2017] [Accepted: 10/09/2017] [Indexed: 02/07/2023]
Abstract
Tumor-associated macrophages (TAM) contribute to all aspects of tumor progression. Use of CSF1R inhibitors to target TAM is therapeutically appealing, but has had very limited anti-tumor effects. Here, we have identified the mechanism that limited the effect of CSF1R targeted therapy. We demonstrated that carcinoma-associated fibroblasts (CAF) are major sources of chemokines that recruit granulocytes to tumors. CSF1 produced by tumor cells caused HDAC2-mediated downregulation of granulocyte-specific chemokine expression in CAF, which limited migration of these cells to tumors. Treatment with CSF1R inhibitors disrupted this crosstalk and triggered a profound increase in granulocyte recruitment to tumors. Combining CSF1R inhibitor with a CXCR2 antagonist blocked granulocyte infiltration of tumors and showed strong anti-tumor effects.
Collapse
Affiliation(s)
- Vinit Kumar
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | - Douglas Marvel
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Thomas Condamine
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Fang Wang
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Sergio Lavilla-Alonso
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Ayumi Hashimoto
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Prashanthi Vonteddu
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Reeti Behera
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Marlee A Goins
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Charles Mulligan
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Brian Nam
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Neil Hockstein
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Fred Denstman
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Shanti Shakamuri
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - David W Speicher
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Ashani T Weeraratna
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Timothy Chao
- University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | - Lucia R Languino
- Sidney Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | - Qin Liu
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Albert Lo
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Ellen Puré
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Chunsheng Zhang
- Department of Genetics and Pharmacogenomics, MRL, Merck & Co., Inc., Boston, MA 02115, USA
| | - Andrey Loboda
- Department of Genetics and Pharmacogenomics, MRL, Merck & Co., Inc., Boston, MA 02115, USA
| | | | | | - Dmitry I Gabrilovich
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Wiehagen KR, Girgis NM, Yamada DH, Smith AA, Chan SR, Grewal IS, Quigley M, Verona RI. Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor Immunity. Cancer Immunol Res 2017; 5:1109-1121. [PMID: 29097420 DOI: 10.1158/2326-6066.cir-17-0258] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/01/2017] [Accepted: 10/24/2017] [Indexed: 11/16/2022]
Abstract
Efficacious antitumor immune responses must overcome multiple suppressive mechanisms in the tumor microenvironment to control cancer progression. In this study, we demonstrate that dual targeting of suppressive myeloid populations by inhibiting CSF-1/CSF-1R signaling and activation of antigen-presenting cells with agonist anti-CD40 treatment confers superior antitumor efficacy and increased survival compared with monotherapy treatment in preclinical tumor models. Concurrent CSF-1R blockade and CD40 agonism lead to profound changes in the composition of immune infiltrates, causing an overall decrease in immunosuppressive cells and a shift toward a more inflammatory milieu. Anti-CD40/anti-CSF-1R-treated tumors contain decreased tumor-associated macrophages and Foxp3+ regulatory T cells. This combination approach increases maturation and differentiation of proinflammatory macrophages and dendritic cells and also drives potent priming of effector T cells in draining lymph nodes. As a result, tumor-infiltrating effector T cells exhibit improved responses to tumor antigen rechallenge. These studies show that combining therapeutic approaches may simultaneously remove inhibitory immune populations and sustain endogenous antitumor immune responses to successfully impair cancer progression. Cancer Immunol Res; 5(12); 1109-21. ©2017 AACR.
Collapse
Affiliation(s)
| | - Natasha M Girgis
- Janssen Research and Development, Spring House, Pennsylvania.,Constellation Pharmaceuticals, Cambridge, Massachusetts
| | | | | | | | - Iqbal S Grewal
- Janssen Research and Development, Spring House, Pennsylvania
| | - Michael Quigley
- Janssen Research and Development, Spring House, Pennsylvania.,Bristol-Myers Squibb, Princeton, New Jersey
| | - Raluca I Verona
- Janssen Research and Development, Spring House, Pennsylvania.
| |
Collapse
|
33
|
Ao JY, Zhu XD, Chai ZT, Cai H, Zhang YY, Zhang KZ, Kong LQ, Zhang N, Ye BG, Ma DN, Sun HC. Colony-Stimulating Factor 1 Receptor Blockade Inhibits Tumor Growth by Altering the Polarization of Tumor-Associated Macrophages in Hepatocellular Carcinoma. Mol Cancer Ther 2017; 16:1544-1554. [PMID: 28572167 DOI: 10.1158/1535-7163.mct-16-0866] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/23/2017] [Accepted: 05/19/2017] [Indexed: 11/16/2022]
Abstract
Colony-stimulating factor-1 (CSF-1) and its receptor, CSF-1R, regulate the differentiation and function of macrophages and play an important role in macrophage infiltration in the context of hepatocellular carcinoma. The therapeutic effects of CSF-1R blockade in hepatocellular carcinoma remain unclear. In this study, we found that CSF-1R blockade by PLX3397, a competitive inhibitor with high specificity for CSF-1R tyrosine kinase, significantly delayed tumor growth in mouse models. PLX3397 inhibited the proliferation of macrophages in vitro, but intratumoral macrophage infiltration was not decreased by PLX3397 in vivo Gene expression profiling of tumor-associated macrophages (TAM) showed that TAMs from the PLX3397-treated tumors were polarized toward an M1-like phenotype compared with those from vehicle-treated tumors. In addition, PLX3397 treatment increased CD8+ T-cell infiltration, whereas CD4+ T-cell infiltration was decreased. Further study revealed that tumor cell-derived CSF-2 protected TAMs from being depleted by PLX3397. In conclusion, CSF-1R blockade delayed tumor growth by shifting the polarization rather than the depletion of TAMs. CSF-1R blockade warrants further investigation in the treatment of hepatocellular carcinoma. Mol Cancer Ther; 16(8); 1544-54. ©2017 AACR.
Collapse
Affiliation(s)
- Jian-Yang Ao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Zong-Tao Chai
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Hao Cai
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Yuan-Yuan Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Ke-Zhi Zhang
- Department of Hepatobiliary Surgery, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Ling-Qun Kong
- Department of Hepatobiliary Surgery, Binzhou Medical College Affiliated Hospital, Binzhou, Shandong, China
| | - Ning Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Bo-Gen Ye
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - De-Ning Ma
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| |
Collapse
|
34
|
Sawa-Wejksza K, Kandefer-Szerszeń M. Tumor-Associated Macrophages as Target for Antitumor Therapy. Arch Immunol Ther Exp (Warsz) 2017; 66:97-111. [PMID: 28660349 PMCID: PMC5851686 DOI: 10.1007/s00005-017-0480-8] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
Abstract
It is well known that the microenvironment of solid tumors is rich in inflammatory cells that influence tumor growth and development. Macrophages, called tumor-associated macrophages (TAMs), are the most abundant immune cell population present in tumor tissue. Several studies have demonstrated that the density of TAMs is associated with a poor prognosis and positively correlates with tumor growth. Several studies have proved that TAMs may activate and protect tumor stem cells, stimulate their proliferation as well as promote angiogenesis and metastasis. Furthermore, TAMs-derived cytokines and other proteins, such as CCL-17, CCL-22, TGF-β, IL-10, arginase 1, and galectin-3, make a significant contribution to immunosuppression. Since TAMs influence various aspects of cancer progression, there are many attempts to use them as a target for immunotherapy. The numerous studies have shown that the primary tumor growth and the number of metastatic sites can be significantly decreased by decreasing the population of macrophages in tumor tissue, for example, by blocking recruitment of monocytes or eliminating TAMs already present in the tumor tissue. Moreover, there are attempts at reprogramming TAMs into proinflammatory M1 macrophages or neutralizing the protumoral products of TAMs. Another approach uses TAMs for anticancer drug delivery into the tumor environment. In this review, we would like to summarize the clinical and preclinical trials that were focused on macrophages as a target for anticancer therapies.
Collapse
Affiliation(s)
- Katarzyna Sawa-Wejksza
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Martyna Kandefer-Szerszeń
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| |
Collapse
|
35
|
Dammeijer F, Lievense LA, Kaijen-Lambers ME, van Nimwegen M, Bezemer K, Hegmans JP, van Hall T, Hendriks RW, Aerts JG. Depletion of Tumor-Associated Macrophages with a CSF-1R Kinase Inhibitor Enhances Antitumor Immunity and Survival Induced by DC Immunotherapy. Cancer Immunol Res 2017; 5:535-546. [DOI: 10.1158/2326-6066.cir-16-0309] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/30/2017] [Accepted: 05/16/2017] [Indexed: 11/16/2022]
|
36
|
Overcoming immunosuppression in bone metastases. Crit Rev Oncol Hematol 2017; 117:114-127. [PMID: 28600175 DOI: 10.1016/j.critrevonc.2017.05.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/30/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022] Open
Abstract
Bone metastases are present in up to 70% of advanced prostate and breast cancers and occur at significant rates in a variety of other cancers. Bone metastases can be associated with significant morbidity. The establishment of bone metastasis activates several immunosuppressive mechanisms. Hence, understanding the tumor-bone microenvironment is crucial to inform the development of novel therapies. This review describes the current standard of care for patients with bone metastatic disease and novel treatment options targeting the microenvironment. Treatments reviewed include immunotherapies, cryoablation, and targeted therapies. Combinatorial treatment strategies including targeted therapies and immunotherapies show promise in pre-clinical and clinical studies to overcome the suppressive environment and improve treatment of bone metastases.
Collapse
|
37
|
Binnemars-Postma K, Storm G, Prakash J. Nanomedicine Strategies to Target Tumor-Associated Macrophages. Int J Mol Sci 2017; 18:E979. [PMID: 28471401 PMCID: PMC5454892 DOI: 10.3390/ijms18050979] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022] Open
Abstract
In recent years, the influence of the tumor microenvironment (TME) on cancer progression has been better understood. Macrophages, one of the most important cell types in the TME, exist in different subtypes, each of which has a different function. While classically activated M1 macrophages are involved in inflammatory and malignant processes, activated M2 macrophages are more involved in the wound-healing processes occurring in tumors. Tumor-associated macrophages (TAM) display M2 macrophage characteristics and support tumor growth and metastasis by matrix remodeling, neo-angiogenesis, and suppressing local immunity. Due to their detrimental role in tumor growth and metastasis, selective targeting of TAM for the treatment of cancer may prove to be beneficial in the treatment of cancer. Due to the plastic nature of macrophages, their activities may be altered to inhibit tumor growth. In this review, we will discuss the therapeutic options for the modulation and targeting of TAM. Different therapeutic strategies to deplete, inhibit recruitment of, or re-educate TAM will be discussed. Current strategies for the targeting of TAM using nanomedicine are reviewed. Passive targeting using different nanoparticle systems is described. Since TAM display a number of upregulated surface proteins compared to non-TAM, specific targeting using targeting ligands coupled to nanoparticles is discussed in detail.
Collapse
Affiliation(s)
- Karin Binnemars-Postma
- Targeted Therapeutics, Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522NB Enschede, The Netherlands.
| | - Gert Storm
- Targeted Therapeutics, Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522NB Enschede, The Netherlands.
- Department of Pharmaceutics, Utrecht University, 3584CS Utrecht, The Netherlands.
| | - Jai Prakash
- Targeted Therapeutics, Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7522NB Enschede, The Netherlands.
| |
Collapse
|
38
|
Yan H, Zhang P, Kong X, Hou X, Zhao L, Li T, Yuan X, Fu H. Primary Tr1 cells from metastatic melanoma eliminate tumor-promoting macrophages through granzyme B- and perforin-dependent mechanisms. Tumour Biol 2017; 39:1010428317697554. [PMID: 28378637 DOI: 10.1177/1010428317697554] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
In malignant melanoma, tumor-associated macrophages play multiple roles in promoting tumor growth, such as inducing the transformation of melanocytes under ultraviolet irradiation, increasing angiogenesis in melanomas, and suppressing antitumor immunity. Because granzyme B- and perforin-expressing Tr1 cells could specifically eliminate antigen-presenting cells of myeloid origin, we examined whether Tr1 cells in melanoma could eliminate tumor-promoting macrophages and how the interaction between Tr1 cells and macrophages could affect the growth of melanoma cells. Tr1 cells were characterized by high interleukin 10 secretion and low Foxp3 expression and were enriched in the CD4+CD49b+LAG-3+ T-cell fraction. Macrophages derived from peripheral blood monocytes in the presence of modified melanoma-conditioned media demonstrated tumor-promoting capacity, exemplified by improving the proliferation of cocultured A375 malignant melanoma cells. But when primary Tr1 cells were present in the macrophage-A375 coculture, the growth of A375 cells was abrogated. The conventional CD25+ Treg cells, however, were unable to inhibit macrophage-mediated increase in tumor cell growth. Further analyses showed that Tr1 cells did not directly eliminate A375 cells, but mediated the killing of tumor-promoting macrophages through the secretion of granzyme B and perforin. The tumor-infiltrating interleukin 10+Foxp3-CD4+ T cells expressed very low levels of granzyme B and perforin, possibly suggested the downregulation of Tr1 cytotoxic capacity in melanoma tumors. Together, these data demonstrated an antitumor function of Tr1 cells through the elimination of tumor-promoting macrophages, which was not shared by conventional Tregs.
Collapse
Affiliation(s)
- Hongxia Yan
- 1 Department of Dermatology, The First People's Hospital of Jining City, Jining, China
| | - Ping Zhang
- 2 Jining Maternity and Child Health Care Hospital, Jining, China
| | - Xue Kong
- 3 Department of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xianglian Hou
- 4 Department of Supply and Services, Jiaxiang County People's Hospital, Jining, China
| | - Li Zhao
- 1 Department of Dermatology, The First People's Hospital of Jining City, Jining, China
| | - Tianhang Li
- 1 Department of Dermatology, The First People's Hospital of Jining City, Jining, China
| | - Xiaozhou Yuan
- 5 DICAT Biomedical Computation Centre, Vancouver, BC, Canada
| | - Hongjun Fu
- 1 Department of Dermatology, The First People's Hospital of Jining City, Jining, China
| |
Collapse
|
39
|
Wen ZQ, Li XG, Zhang YJ, Ling ZH, Lin XJ. Osteosarcoma cell-intrinsic colony stimulating factor-1 receptor functions to promote tumor cell metastasis through JAG1 signaling. Am J Cancer Res 2017; 7:801-815. [PMID: 28469954 PMCID: PMC5411789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 01/02/2017] [Indexed: 06/07/2023] Open
Abstract
Therapeutic antibodies or inhibitors targeting CSF-1R block colony stimulating factor-1/colony stimulating factor-1 receptor (CSF-1/CSF-R) signaling, and have shown remarkable efficacy in the treatment of cancer. However, little is known about tumor cell-intrinsic CSF-1R effects. Here, we show that human osteosarcomas contain CSF-1R-expressing cancer subpopulations, and demonstrate that osteosarcoma cell-intrinsic CSF-1R promotes growth in vitro and in vivo. CSF-1R inhibition in osteosarcoma cells by RNA interference suppresses cell proliferation and tumor growth in mice. Conversely, CSF-1R overexpression enhances cell proliferation and accelerates tumor growth. CSF-1R overexpression can significantly enhance osteosarcoma cell migration, invasion, and epithelial-mesenchymal transition (EMT), whereas silencing CSF-1R inhibits these processes. Microarray analysis suggests that jagged 1 (JAG1) can function as a downstream mediator of CSF-1R. Moreover, we report a signaling pathway involving CSF-1R and JAG1 that sustains osteosarcoma cell migration and invasion. Our results identify osteosarcoma cell intrinsic functions of the CSF-1R/JAG1 axis in dissemination of osteosarcoma cells.
Collapse
Affiliation(s)
- Zhi-Qiang Wen
- Department of Orthopaedics, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityNo. 79 Qingchun Road, Hangzhou, Zhejiang, China
| | - Xi-Gong Li
- Department of Orthopaedics, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityNo. 79 Qingchun Road, Hangzhou, Zhejiang, China
| | - Yi-Jun Zhang
- Department of Orthopaedics, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityNo. 79 Qingchun Road, Hangzhou, Zhejiang, China
| | - Zhi-Heng Ling
- Department of Orthopaedics, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityNo. 79 Qingchun Road, Hangzhou, Zhejiang, China
| | - Xiang-Jin Lin
- Department of Orthopaedics, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityNo. 79 Qingchun Road, Hangzhou, Zhejiang, China
| |
Collapse
|
40
|
Mendel I, Yacov N, Salem Y, Propheta-Meiran O, Ishai E, Breitbart E. Identification of Motile Sperm Domain-Containing Protein 2 as Regulator of Human Monocyte Migration. THE JOURNAL OF IMMUNOLOGY 2017; 198:2125-2132. [PMID: 28137892 DOI: 10.4049/jimmunol.1601662] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/28/2016] [Indexed: 01/12/2023]
Abstract
Binding of chemokines to their cognate receptors on monocytes instigates a cascade of events that directs these cells to migrate to sites of inflammation and cancerous tissues. Although targeting of selected chemokine receptors on monocytes exhibited preclinical efficacy, attempts to translate these studies to the clinic have failed thus far, possibly due to redundancy of the target receptor. We reveal that motile sperm domain-containing protein 2 (MOSPD2), a protein with a previously unknown function, regulates monocyte migration in vitro. This protein was found to be expressed on the cytoplasmic membrane of human monocytes. Silencing or neutralizing MOSPD2 in monocytes restricted their migration when induced by different chemokines. Mechanistically, silencing MOSPD2 inhibited signaling events following chemokine receptor ligation. When tested for expression in other immune cell subsets, MOSPD2 was apparent also, though less abundantly, in neutrophils, but not in lymphocytes. Thus, in the presence of neutralizing Abs, neutrophil migration was inhibited to some extent whereas lymphocyte migration remained intact. In view of these results, we suggest MOSPD2 as a potential target protein for treating diseases in which monocyte and neutrophil accumulation is correlated with pathogenesis.
Collapse
Affiliation(s)
| | - Niva Yacov
- VBL Therapeutics, Or Yehuda 6037604, Israel
| | | | | | - Eti Ishai
- VBL Therapeutics, Or Yehuda 6037604, Israel
| | | |
Collapse
|
41
|
The tumour microenvironment harbours ontogenically distinct dendritic cell populations with opposing effects on tumour immunity. Nat Commun 2016; 7:13720. [PMID: 28008905 PMCID: PMC5196231 DOI: 10.1038/ncomms13720] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 10/25/2016] [Indexed: 12/16/2022] Open
Abstract
Various steady state and inflamed tissues have been shown to contain a heterogeneous DC population consisting of developmentally distinct subsets, including cDC1s, cDC2s and monocyte-derived DCs, displaying differential functional specializations. The identification of functionally distinct tumour-associated DC (TADC) subpopulations could prove essential for the understanding of basic TADC biology and for envisaging targeted immunotherapies. We demonstrate that multiple mouse tumours as well as human tumours harbour ontogenically discrete TADC subsets. Monocyte-derived TADCs are prominent in tumour antigen uptake, but lack strong T-cell stimulatory capacity due to NO-mediated immunosuppression. Pre-cDC-derived TADCs have lymph node migratory potential, whereby cDC1s efficiently activate CD8+ T cells and cDC2s induce Th17 cells. Mice vaccinated with cDC2s displayed a reduced tumour growth accompanied by a reprogramming of pro-tumoural TAMs and a reduction of MDSCs, while cDC1 vaccination strongly induces anti-tumour CTLs. Our data might prove important for therapeutic interventions targeted at specific TADC subsets or their precursors. Dendritic cells are antigen-presenting cells consisting of distinct subsets originating from different lineages. Here, the authors identify the subsets of dendritic cells populating the tumour tissue in both mice and humans and find they have opposing functions in regulating the anti-tumour immune response.
Collapse
|
42
|
Beyranvand Nejad E, Welters MJP, Arens R, van der Burg SH. The importance of correctly timing cancer immunotherapy. Expert Opin Biol Ther 2016; 17:87-103. [PMID: 27802061 DOI: 10.1080/14712598.2017.1256388] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The treatment options for cancer-surgery, radiotherapy and chemotherapy-are now supplemented with immunotherapy. Previously underappreciated but now gaining strong interest are the immune modulatory properties of the three conventional modalities. Moreover, there is a better understanding of the needs and potential of the different immune therapeutic platforms. Key to improved treatment will be the combinations of modalities that complete each other's shortcomings. Area covered: Tumor-specific T-cells are required for optimal immunotherapy. In this review, the authors focus on the correct timing of different types of chemotherapeutic agents or immune modulators and immunotherapeutic drugs, not only for the activation and expansion of tumor-specific T-cells but also to support and enhance their anti-tumor efficacy. Expert opinion: At an early phase of disease, clinical success can be obtained using single treatment modalities but at later disease stages, combinations of several modalities are required. The gain in success is determined by a thorough understanding of the direct and indirect immune effects of the modalities used. Profound knowledge of these effects requires optimal tuning of immunomonitoring. This will guide the appropriate combination of treatments and allow for correct sequencing the order and interval of the different therapeutic modalities.
Collapse
Affiliation(s)
- Elham Beyranvand Nejad
- a Department of Medical Oncology , Leiden University Medical Center , Leiden , The Netherlands.,b Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , Leiden , The Netherlands
| | - Marij J P Welters
- a Department of Medical Oncology , Leiden University Medical Center , Leiden , The Netherlands
| | - Ramon Arens
- b Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , Leiden , The Netherlands
| | - Sjoerd H van der Burg
- a Department of Medical Oncology , Leiden University Medical Center , Leiden , The Netherlands
| |
Collapse
|
43
|
Vaccine-induced tumor regression requires a dynamic cooperation between T cells and myeloid cells at the tumor site. Oncotarget 2016; 6:27832-46. [PMID: 26337837 PMCID: PMC4695029 DOI: 10.18632/oncotarget.4940] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/29/2015] [Indexed: 12/31/2022] Open
Abstract
Most cancer immunotherapies under present investigation are based on the belief that cytotoxic T cells are the most important anti-tumoral immune cells, whereas intra-tumoral macrophages would rather play a pro-tumoral role. We have challenged this antagonistic point of view and searched for collaborative contributions by tumor-infiltrating T cells and macrophages, reminiscent of those observed in anti-infectious responses. We demonstrate that, in a model of therapeutic vaccination, cooperation between myeloid cells and T cells is indeed required for tumor rejection. Vaccination elicited an early rise of CD11b+ myeloid cells that preceded and conditioned the intra-tumoral accumulation of CD8+ T cells. Conversely, CD8+ T cells and IFNγ production activated myeloid cells were required for tumor regression. A 4-fold reduction of CD8+ T cell infiltrate in CXCR3KO mice did not prevent tumor regression, whereas a reduction of tumor-infiltrating myeloid cells significantly interfered with vaccine efficiency. We show that macrophages from regressing tumors can kill tumor cells in two ways: phagocytosis and TNFα release. Altogether, our data suggest new strategies to improve the efficiency of cancer immunotherapies, by promoting intra-tumoral cooperation between macrophages and T cells.
Collapse
|
44
|
Torres L, Danver J, Ji K, Miyauchi JT, Chen D, Anderson ME, West BL, Robinson JK, Tsirka SE. Dynamic microglial modulation of spatial learning and social behavior. Brain Behav Immun 2016; 55:6-16. [PMID: 26348580 PMCID: PMC4779430 DOI: 10.1016/j.bbi.2015.09.001] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 08/29/2015] [Accepted: 09/01/2015] [Indexed: 11/29/2022] Open
Abstract
Microglia are active players in inflammation, but also have important supporting roles in CNS maintenance and function, including modulation of neuronal activity. We previously observed an increase in the frequency of excitatory postsynaptic current in organotypic brain slices after depletion of microglia using clodronate. Here, we describe that local hippocampal depletion of microglia by clodronate alters performance in tests of spatial memory and sociability. Global depletion of microglia by high-dose oral administration of a Csf1R inhibitor transiently altered spatial memory but produced no change in sociability behavior. Microglia depletion and behavior effects were both reversible, consistent with a dynamic role for microglia in the regulation of such behaviors.
Collapse
Affiliation(s)
- Luisa Torres
- Program in Molecular and Cellular Pharmacology Stony Brook University, New York 11794-8651,Department of Pharmacological Sciences, Stony Brook University, New York 11794-8651
| | - Joan Danver
- Program in Biochemistry and Cell Biology, Stony Brook University, New York 11794-8651
| | - Kyungmin Ji
- Department of Pharmacological Sciences, Stony Brook University, New York 11794-8651
| | | | | | - Maria E. Anderson
- Integrated Neuroscience Area, Department of Psychology, Stony Brook University, Stony Brook, New York 11794-2500
| | | | - John K. Robinson
- Integrated Neuroscience Area, Department of Psychology, Stony Brook University, Stony Brook, New York 11794-2500
| | - Stella E. Tsirka
- Program in Molecular and Cellular Pharmacology Stony Brook University, New York 11794-8651,Department of Pharmacological Sciences, Stony Brook University, New York 11794-8651,Corresponding Author: Stella E. Tsirka, Ph.D., Department of Pharmacological Sciences, BST8-192, Stony Brook, NY 11794-8651; Tel: 631-444-3859;
| |
Collapse
|
45
|
Abstract
Recent clinical trials have demonstrated the ability to durably control cancer in some patients by manipulating T lymphocytes. These immunotherapies are revolutionizing cancer treatment but benefit only a minority of patients. It is thus a crucial time for clinicians, cancer scientists and immunologists to determine the next steps in shifting cancer treatment towards better cancer control. This Review describes recent advances in our understanding of tumour-associated myeloid cells. These cells remain less studied than T lymphocytes but have attracted particular attention because their presence in tumours is often linked to altered patient survival. Also, experimental studies indicate that myeloid cells modulate key cancer-associated activities, including immune evasion, and affect virtually all types of cancer therapy. Consequently, targeting myeloid cells could overcome limitations of current treatment options.
Collapse
Affiliation(s)
- Camilla Engblom
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts 02114, USA
- Graduate Program in Immunology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Christina Pfirschke
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
46
|
Buqué A, Bloy N, Aranda F, Cremer I, Eggermont A, Fridman WH, Fucikova J, Galon J, Spisek R, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch-Small molecules targeting the immunological tumor microenvironment for cancer therapy. Oncoimmunology 2016; 5:e1149674. [PMID: 27471617 PMCID: PMC4938376 DOI: 10.1080/2162402x.2016.1149674] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 12/21/2022] Open
Abstract
Progressing malignancies establish robust immunosuppressive networks that operate both systemically and locally. In particular, as tumors escape immunosurveillance, they recruit increasing amounts of myeloid and lymphoid cells that exert pronounced immunosuppressive effects. These cells not only prevent the natural recognition of growing neoplasms by the immune system, but also inhibit anticancer immune responses elicited by chemo-, radio- and immuno therapeutic interventions. Throughout the past decade, multiple strategies have been devised to counteract the accumulation or activation of tumor-infiltrating immunosuppressive cells for therapeutic purposes. Here, we review recent preclinical and clinical advances on the use of small molecules that target the immunological tumor microenvironment for cancer therapy. These agents include inhibitors of indoleamine 2,3-dioxigenase 1 (IDO1), prostaglandin E2, and specific cytokine receptors, as well as modulators of intratumoral purinergic signaling and arginine metabolism.
Collapse
Affiliation(s)
- Aitziber Buqué
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Norma Bloy
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Isabelle Cremer
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | | | - Wolf Hervé Fridman
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers, Paris, France
| | - Radek Spisek
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Eric Tartour
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- INSERM, U970, Paris, France
- Paris-Cardiovascular Research Center (PARCC), Paris, France
- Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou (HEGP), AP-HP, Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, U1015, CICBT507, Villejuif, France
| | - Guido Kroemer
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
47
|
Holmgaard RB, Brachfeld A, Gasmi B, Jones DR, Mattar M, Doman T, Murphy M, Schaer D, Wolchok JD, Merghoub T. Timing of CSF-1/CSF-1R signaling blockade is critical to improving responses to CTLA-4 based immunotherapy. Oncoimmunology 2016; 5:e1151595. [PMID: 27622016 DOI: 10.1080/2162402x.2016.1151595] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/03/2015] [Accepted: 02/03/2016] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED Colony stimulating factor-1 (CSF-1) is produced by a variety of cancers and recruits myeloid cells that suppress antitumor immunity, including myeloid-derived suppressor cells (MDSCs.) Here, we show that both CSF-1 and its receptor (CSF-1R) are frequently expressed in tumors from cancer patients, and that this expression correlates with tumor-infiltration of MDSCs. Furthermore, we demonstrate that these tumor-infiltrating MDSCs are highly immunosuppressive but can be reprogrammed toward an antitumor phenotype in vitro upon CSF-1/CSF-1R signaling blockade. Supporting these findings, we show that inhibition of CSF-1/CSF-1R signaling using an anti-CSF-1R antibody can regulate both the number and the function of MDSCs in murine tumors in vivo. We further find that treatment with anti-CSF-1R antibody induces antitumor T-cell responses and tumor regression in multiple tumor models when combined with CTLA-4 blockade therapy. However, this occurs only when administered after or concurrent with CTLA-4 blockade, indicating that timing of each therapeutic intervention is critical for optimal antitumor responses. Importantly, MDSCs present within murine tumors after CTLA-4 blockade showed increased expression of CSF-1R and were capable of suppressing T cell proliferation, and CSF-1/CSF-1R expression in the human tumors was not reduced after treatment with CTLA-4 blockade immunotherapy. Taken together, our findings suggest that CSF-1R-expressing MDSCs can be targeted to modulate the tumor microenvironment and that timing of CSF-1/CSF-1R signaling blockade is critical to improving responses to checkpoint based immunotherapy. SIGNIFICANCE Infiltration by immunosuppressive myeloid cells contributes to tumor immune escape and can render patients resistant or less responsive to therapeutic intervention with checkpoint blocking antibodies. Our data demonstrate that blocking CSF-1/CSF-1R signaling using a monoclonal antibody directed to CSF-1R can regulate both the number and function of tumor-infiltrating immunosuppressive myeloid cells. In addition, our findings suggest that reprogramming myeloid responses may be a key in effectively enhancing cancer immunotherapy, offering several new potential combination therapies for future clinical testing. More importantly for clinical trial design, the timing of these interventions is critical to achieving improved tumor protection.
Collapse
Affiliation(s)
- Rikke B Holmgaard
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Alexandra Brachfeld
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Billel Gasmi
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - David R Jones
- Department of Surgery, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Marissa Mattar
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | | | | | | | - Jedd D Wolchok
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College and Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Taha Merghoub
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| |
Collapse
|
48
|
Holmgaard RB, Zamarin D, Lesokhin A, Merghoub T, Wolchok JD. Targeting myeloid-derived suppressor cells with colony stimulating factor-1 receptor blockade can reverse immune resistance to immunotherapy in indoleamine 2,3-dioxygenase-expressing tumors. EBioMedicine 2016; 6:50-58. [PMID: 27211548 PMCID: PMC4856741 DOI: 10.1016/j.ebiom.2016.02.024] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 12/21/2022] Open
Abstract
Tumor indoleamine 2,3-dioxygenase (IDO) promotes immunosuppression by direct action on effector T cells and Tregs and through recruitment, expansion and activation of myeloid-derived suppressor cells (MDSCs). Targeting of MDSCs is clinically being explored as a therapeutic strategy, though optimal targeting strategies and biomarkers predictive of response are presently unknown. Maturation and tumor recruitment of MDSCs are dependent on signaling through the receptor tyrosine kinase CSF-1R on myeloid cells. Here, we show that MDSCs are the critical cell population in IDO-expressing B16 tumors in mediating accelerated tumor outgrowth and resistance to immunotherapy. Using a clinically relevant drug, we show that inhibition of CSF-1R signaling can functionally block tumor-infiltrating MDSCs and enhance anti-tumor T cell responses. Furthermore, inhibition of CSF-1R sensitizes IDO-expressing tumors to immunotherapy with T cell checkpoint blockade, and combination of CSF-1R blockade with IDO inhibitors potently elicits tumor regression. These findings provide evidence for a critical and functional role for MDSCs on the in vivo outcome of IDO-expressing tumors. Tumor-infiltrating MDSCs promote accelerated outgrowth of IDO-expressing tumors. MDSCs infiltrating IDO-expressing tumors mediate resistance to immunotherapy. Combination of CSF-1R blockade with IDO inhibitors potently elicits tumor regression. CSF-1R blockade sensitizes tumors to the effects of immune checkpoint blockade.
Our data demonstrate that therapy with CSF-1R-blocking agents offers therapeutic benefit as a single agent and potentiates the effect of immunotherapeutic agents in IDO-expressing tumors infiltrated with CSF-1R-expressing MDSCs. These findings provide important insights into basic mechanisms underlying IDO mediated immune suppression and resistance to immunotherapies. In addition, it provides a strong rationale for therapeutic combinations with CSF-1R inhibitors in tumors expressing elevated IDO and highly infiltrated with MDSCs as predictive biomarkers.
Collapse
Affiliation(s)
- Rikke B Holmgaard
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Dmitriy Zamarin
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States; Weill Cornell Medical College, New York, NY 10065, United States; Graduate School of Medical Sciences of Cornell University, New York, NY 10065, United States
| | - Alexander Lesokhin
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States; Weill Cornell Medical College, New York, NY 10065, United States; Graduate School of Medical Sciences of Cornell University, New York, NY 10065, United States
| | - Taha Merghoub
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jedd D Wolchok
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States; Weill Cornell Medical College, New York, NY 10065, United States; Graduate School of Medical Sciences of Cornell University, New York, NY 10065, United States.
| |
Collapse
|
49
|
Rosenfeld L, Shirian J, Zur Y, Levaot N, Shifman JM, Papo N. Combinatorial and Computational Approaches to Identify Interactions of Macrophage Colony-stimulating Factor (M-CSF) and Its Receptor c-FMS. J Biol Chem 2015; 290:26180-93. [PMID: 26359491 PMCID: PMC4646268 DOI: 10.1074/jbc.m115.671271] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/06/2015] [Indexed: 01/06/2023] Open
Abstract
The molecular interactions between macrophage colony-stimulating factor (M-CSF) and the tyrosine kinase receptor c-FMS play a key role in the immune response, bone metabolism, and the development of some cancers. Because no x-ray structure is available for the human M-CSF · c-FMS complex, the binding epitope for this complex is largely unknown. Our goal was to identify the residues that are essential for binding of the human M-CSF to c-FMS. For this purpose, we used a yeast surface display (YSD) approach. We expressed a combinatorial library of monomeric M-CSF (M-CSFM) single mutants and screened this library to isolate variants with reduced affinity for c-FMS using FACS. Sequencing yielded a number of single M-CSFM variants with mutations both in the direct binding interface and distant from the binding site. In addition, we used computational modeling to map the identified mutations onto the M-CSFM structure and to classify the mutations into three groups as follows: those that significantly decrease protein stability; those that destroy favorable intermolecular interactions; and those that decrease affinity through allosteric effects. To validate the YSD and computational data, M-CSFM and three variants were produced as soluble proteins; their affinity and structure were analyzed; and very good correlations with both YSD data and computational predictions were obtained. By identifying the M-CSFM residues critical for M-CSF · c-FMS interactions, we have laid down the basis for a deeper understanding of the M-CSF · c-FMS signaling mechanism and for the development of target-specific therapeutic agents with the ability to sterically occlude the M-CSF·c-FMS binding interface.
Collapse
Affiliation(s)
- Lior Rosenfeld
- From the Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, and
| | - Jason Shirian
- the Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Yuval Zur
- From the Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, and the Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva 8410501 and
| | - Noam Levaot
- the Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva 8410501 and
| | - Julia M Shifman
- the Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Niv Papo
- From the Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, and
| |
Collapse
|
50
|
CSF-1/CSF-1R targeting agents in clinical development for cancer therapy. Curr Opin Pharmacol 2015; 23:45-51. [PMID: 26051995 DOI: 10.1016/j.coph.2015.05.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/07/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023]
Abstract
Macrophage infiltration has been identified as an independent poor prognostic factor for several cancer entities. In mouse tumor models macrophages orchestrate various tumor-promoting processes. This observation sparked an interest to therapeutically target these plastic innate immune cells. To date, blockade of colony stimulating factor-1 or its receptor represents the only truly selective approach to manipulate macrophages in cancer patients. Here, we discuss the currently available information on efficacy and safety of various CSF-1/CSF-1R inhibitors in cancer patients and highlight potential combination partners emerging from preclinical studies while considering the differences between mouse and human macrophage biology.
Collapse
|