101
|
Activity of tumor-associated macrophage depletion by CSF1R blockade is highly dependent on the tumor model and timing of treatment. Cancer Immunol Immunother 2021; 70:2401-2410. [PMID: 33511454 PMCID: PMC8289806 DOI: 10.1007/s00262-021-02861-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/11/2021] [Indexed: 12/29/2022]
Abstract
Tumor-associated macrophages (TAMs) are abundant in solid tumors where they exhibit immunosuppressive and pro-tumorigenic functions. Inhibition of TAM proliferation and survival through CSF1R blockade has been widely explored as a cancer immunotherapy. To further define mechanisms regulating CSF1R-targeted therapies, we systematically evaluated the effect of anti-CSF1R treatment on tumor growth and tumor microenvironment (TME) inflammation across multiple murine models. Despite substantial macrophage depletion, anti-CSF1R had minimal effects on the anti-tumor immune response in mice bearing established tumors. In contrast, anti-CSF1R treatment concurrent with tumor implantation resulted in more robust tumor growth inhibition and evidence of enhanced anti-tumor immunity. Our findings suggest only minor contributions of CSF1R-dependent TAMs to the inflammatory state of the TME in established tumors, that immune landscape heterogeneity across different tumor models can influence anti-CSF1R activity, and that alternative treatment schedules and/or TAM depletion strategies may be needed to maximize the clinical benefit of this approach.
Collapse
|
102
|
Cao Q, Huang Q, Wang YA, Li C. Abraxane-induced bone marrow CD11b + myeloid cell depletion in tumor-bearing mice is visualized by μPET-CT with 64Cu-labeled anti-CD11b and prevented by anti-CSF-1. Theranostics 2021; 11:3527-3539. [PMID: 33537102 PMCID: PMC7847669 DOI: 10.7150/thno.49421] [Citation(s) in RCA: 4] [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: 06/12/2020] [Accepted: 12/09/2020] [Indexed: 11/23/2022] Open
Abstract
To investigate the utility of noninvasive µPET-CT with 64Cu-DOTA-anti-CD11b (64Cu-αCD11b) in assessing bone marrow status after anticancer therapies, and the protective role of anti-CSF-1 (αCSF-1) against bone marrow suppression induced by Abraxane. Methods: MDA-MB-435 tumor-bearing mice were treated with Abraxane, αCSF-1, or αCSF-1 plus Abraxane. µPET-CT and biodistribution of 64Cu-αCD11b were performed after intravenous injection of the radiotracer. Cells from mouse bone marrow and MDA-MB-435 tumor were analyzed by flow cytometry. A humanized αCSF-1 was investigated for its role in protecting bone marrow cells, using a transgenic mouse model that expresses functional human CSF-1. Results: μPET-CT showed that 64Cu-αCD11b had high uptake in the bone marrow and spleen of both normal and tumor-bearing mice. Abraxane significantly reduced 64Cu-αCD11b uptake in the bone marrow and spleen of treated mice compared to untreated mice. Interestingly, 64Cu-αCD11b μPET-CT revealed that αCSF-1 alleviated the depletion of bone marrow cells by Abraxane. These changes in the bone marrow population of CD11b+ myeloid cells were confirmed by flow cytometry. Moreover, αCSF-1 potently enhanced tolerance of bone marrow granulocytic myeloid cells to Abraxane, decreased cell migration, and suppressed recruitment of myeloid cells to the tumor microenvironment. The humanized αCSF-1 also alleviated the effects of Abraxane on bone marrow cells in transgenic mice expressing human CSF-1, suggesting clinical relevance of αCSF-1 in prevention of bone marrow suppression in addition to its role in reducing tumor-infiltrating myeloid cells. Conclusions: Abraxane-induced bone marrow CD11b+ myeloid cell depletion in tumor-bearing mice could be noninvasively assessed by μPET-CT with 64Cu-αCD11b and prevented by αCSF-1.
Collapse
Affiliation(s)
- Qizhen Cao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054
| | - Qian Huang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054
| | - Y. Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054
| | - Chun Li
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054
| |
Collapse
|
103
|
Hou J, Karin M, Sun B. Targeting cancer-promoting inflammation - have anti-inflammatory therapies come of age? Nat Rev Clin Oncol 2021; 18:261-279. [PMID: 33469195 DOI: 10.1038/s41571-020-00459-9] [Citation(s) in RCA: 215] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 02/07/2023]
Abstract
The immune system has crucial roles in cancer development and treatment. Whereas adaptive immunity can prevent or constrain cancer through immunosurveillance, innate immunity and inflammation often promote tumorigenesis and malignant progression of nascent cancer. The past decade has witnessed the translation of knowledge derived from preclinical studies of antitumour immunity into clinically effective, approved immunotherapies for cancer. By contrast, the successful implementation of treatments that target cancer-associated inflammation is still awaited. Anti-inflammatory agents have the potential to not only prevent or delay cancer onset but also to improve the efficacy of conventional therapeutics and next-generation immunotherapies. Herein, we review the current clinical advances and experimental findings supporting the utility of an anti-inflammatory approach to the treatment of solid malignancies. Gaining a better mechanistic understanding of the mode of action of anti-inflammatory agents and designing more effective treatment combinations would advance the clinical application of this therapeutic approach.
Collapse
Affiliation(s)
- Jiajie Hou
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA.
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
104
|
Fulop T, Tripathi S, Rodrigues S, Desroches M, Bunt T, Eiser A, Bernier F, Beauregard PB, Barron AE, Khalil A, Plotka A, Hirokawa K, Larbi A, Bocti C, Laurent B, Frost EH, Witkowski JM. Targeting Impaired Antimicrobial Immunity in the Brain for the Treatment of Alzheimer's Disease. Neuropsychiatr Dis Treat 2021; 17:1311-1339. [PMID: 33976546 PMCID: PMC8106529 DOI: 10.2147/ndt.s264910] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and aging is the most common risk factor for developing the disease. The etiology of AD is not known but AD may be considered as a clinical syndrome with multiple causal pathways contributing to it. The amyloid cascade hypothesis, claiming that excess production or reduced clearance of amyloid-beta (Aβ) and its aggregation into amyloid plaques, was accepted for a long time as the main cause of AD. However, many studies showed that Aβ is a frequent consequence of many challenges/pathologic processes occurring in the brain for decades. A key factor, sustained by experimental data, is that low-grade infection leading to production and deposition of Aβ, which has antimicrobial activity, precedes the development of clinically apparent AD. This infection is chronic, low grade, largely clinically silent for decades because of a nearly efficient antimicrobial immune response in the brain. A chronic inflammatory state is induced that results in neurodegeneration. Interventions that appear to prevent, retard or mitigate the development of AD also appear to modify the disease. In this review, we conceptualize further that the changes in the brain antimicrobial immune response during aging and especially in AD sufferers serve as a foundation that could lead to improved treatment strategies for preventing or decreasing the progression of AD in a disease-modifying treatment.
Collapse
Affiliation(s)
- Tamas Fulop
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Shreyansh Tripathi
- Cluster Innovation Centre, North Campus, University of Delhi, Delhi, 110007, India.,Ikerbasque, The Basque Foundation for Science, Bilbao, Spain
| | - Serafim Rodrigues
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain.,Mathematical Computational and Experimental Neuroscience (MCEN), BCAM - The Basque Center for Applied Mathematics, Bilbao, Spain
| | - Mathieu Desroches
- MathNeuro Team, Inria Sophia Antipolis Méditerranée, Sophia Antipolis, France.,Department of Mathematics, Université Côte d'Azur, Nice, France
| | - Ton Bunt
- Izumi Biosciences, Inc., Lexington, MA, USA
| | - Arnold Eiser
- Leonard Davis Institute, University of Pennsylvania, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Francois Bernier
- Morinaga Milk Industry Co., Ltd, Next Generation Science Institute, Kanagawa, Japan
| | - Pascale B Beauregard
- Department of Biology, Faculty of Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Annelise E Barron
- Department of Bioengineering, Stanford School of Medicine, Stanford, CA, USA
| | - Abdelouahed Khalil
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Adam Plotka
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| | - Katsuiku Hirokawa
- Institute of Health and Life Science, Tokyo Med. Dent. University, Tokyo and Nito-Memory Nakanosogo Hospital, Department of Pathology, Tokyo, Japan
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (ASTAR), Immunos Building, Biopolis, Singapore, Singapore
| | - Christian Bocti
- Research Center on Aging, Department of Medicine, Division of Neurology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Benoit Laurent
- Research Center on Aging, Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Eric H Frost
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
105
|
Edwards SC, Hoevenaar WHM, Coffelt SB. Emerging immunotherapies for metastasis. Br J Cancer 2020; 124:37-48. [PMID: 33262520 PMCID: PMC7782509 DOI: 10.1038/s41416-020-01160-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/07/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Major advances in cancer immunotherapy have dramatically expanded the potential to manipulate immune cells in cancer patients with metastatic disease to counteract cancer spread and extend patient lifespan. One of the most successful types of immunotherapy is the immune checkpoint inhibitors, such as anti-CTLA-4 and anti-PD-1, that keep anti-tumour T cells active. However, not every patient with metastatic disease benefits from this class of drugs and patients often develop resistance to these therapies over time. Tremendous research effort is now underway to uncover new immunotherapeutic targets that can be used in patients who are refractory to anti-CTLA-4 or anti-PD-1 treatment. Here, we discuss results from experimental model systems demonstrating that modulating the immune response can negatively affect metastasis formation. We focus on molecules that boost anti-tumour immune cells and opportunities to block immunosuppression, as well as cell-based therapies with enhanced tumour recognition properties for solid tumours. We also present a list of challenges in treating metastatic disease with immunotherapy that must be considered in order to move laboratory observations into clinical practice and maximise patient benefit. ![]()
Collapse
Affiliation(s)
- Sarah C Edwards
- Cancer Research UK Beatson Institute, Glasgow, UK.,Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Wilma H M Hoevenaar
- Cancer Research UK Beatson Institute, Glasgow, UK.,Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Seth B Coffelt
- Cancer Research UK Beatson Institute, Glasgow, UK. .,Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
106
|
Koliaraki V, Henriques A, Prados A, Kollias G. Unfolding innate mechanisms in the cancer microenvironment: The emerging role of the mesenchyme. J Exp Med 2020; 217:133714. [PMID: 32044979 PMCID: PMC7144533 DOI: 10.1084/jem.20190457] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/09/2019] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
Innate mechanisms in the tumor stroma play a crucial role both in the initial rejection of tumors and in cancer promotion. Here, we provide a concise overview of the innate system in cancer and recent advances in the field, including the activation and functions of innate immune cells and the emerging innate properties and modulatory roles of the fibroblastic mesenchyme. Novel insights into the diverse identities and functions of the innate immune and mesenchymal cells in the microenvironment of tumors should lead to improved anticancer therapies.
Collapse
Affiliation(s)
- Vasiliki Koliaraki
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Ana Henriques
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.,Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Alejandro Prados
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - George Kollias
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.,Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.,Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
107
|
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: 257] [Impact Index Per Article: 51.4] [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
|
108
|
Liang P, Wang G, Liu X, Wang Z, Wang J, Gao W. Spatiotemporal combination of thermosensitive polypeptide fused interferon and temozolomide for post-surgical glioblastoma immunochemotherapy. Biomaterials 2020; 264:120447. [PMID: 33069137 DOI: 10.1016/j.biomaterials.2020.120447] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023]
Abstract
Cancer recurrence post surgical resection is of considerable challenge especially in glioblastoma (GBM) therapy. Herein, we demonstrate that interferon-alpha (IFN) fused to a body temperature-sensitive elastin-like polypeptide (IFN-ELP(V)) formed a depot in situ when injected into GBM resection cavity in a mouse brain orthotopic model of GBM. Notably, IFN-ELP(V) in the depot showed a zero-order release kinetics, resulting in dramatically improved pharmacokinetics and biodistribution, and thus inhibited GBM recurrence by stimulating antitumor immunoresponse as compared to IFN. More importantly, when combined with subsequent intraperitoneal injection of temozolomide (TMZ), IFN-ELP(V) could much more effectively suppress post-surgical GBM recurrence than IFN, leading to a remarkably enhanced GBM-free survival rate (60%) over IFN (12.5%). Our findings implicate that the spatiotemporally-programmed combination of IFN-ELP(V) and TMZ leads to the synergy of post-surgical GBM immunochemotherapy, thereby providing a new and effective strategy for cancer therapy.
Collapse
Affiliation(s)
- Ping Liang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Guihuai Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Xinyu Liu
- Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Biomedical Engineering Department, Peking University, Beijing, 100191, PR China
| | - Zhuoran Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Jing Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Weiping Gao
- Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Biomedical Engineering Department, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
109
|
Frisch SM, MacFawn IP. Type I interferons and related pathways in cell senescence. Aging Cell 2020; 19:e13234. [PMID: 32918364 PMCID: PMC7576263 DOI: 10.1111/acel.13234] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/18/2020] [Accepted: 08/07/2020] [Indexed: 01/10/2023] Open
Abstract
This review article addresses the largely unanticipated convergence of two landmark discoveries. The first is the discovery of interferons, critical signaling molecules for all aspects of both innate and adaptive immunity, discovered originally by Isaacs and Lindenmann at the National Institute for Medical Research, London, in 1957 (Proceedings of the Royal Society of London. Series B: Biological Sciences, 1957, 147, 258). The second, formerly unrelated discovery, by Leonard Hayflick and Paul Moorhead (Wistar Institute, Philadelphia) is that cultured cells undergo an irreversible but viable growth arrest, termed senescence, after a finite and predictable number of cell divisions (Experimental Cell Research, 1961, 25, 585). This phenomenon was suspected to relate to organismal aging, which was confirmed subsequently (Nature, 2011, 479, 232). Cell senescence has broad‐ranging implications for normal homeostasis, including immunity, and for diverse disease states, including cancer progression and response to therapy (Nature Medicine, 2015, 21, 1424; Cell, 2019, 179, 813; Cell, 2017, 169, 1000; Trends in Cell Biology, 2018, 28, 436; Journal of Cell Biology, 2018, 217, 65). Here, we critically address the bidirectional interplay between interferons (focusing on type I) and cell senescence, with important implications for health and healthspan.
Collapse
Affiliation(s)
- Steven M. Frisch
- Department of Biochemistry and WVU Cancer Institute West Virginia University Morgantown West Virginia USA
| | - Ian P. MacFawn
- Department of Biochemistry and WVU Cancer Institute West Virginia University Morgantown West Virginia USA
| |
Collapse
|
110
|
Shu Y, Cheng P. Targeting tumor-associated macrophages for cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2020; 1874:188434. [PMID: 32956767 DOI: 10.1016/j.bbcan.2020.188434] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023]
Abstract
Macrophages are important effector cells of the innate immune system and are also major components of the tumor microenvironment (TME). Macrophages that are abundant in the TME are called tumor-associated macrophages (TAMs). As TAMs promote strong tumor angiogenesis and support tumor cell survival, they are closely related to tumor growth. Several studies have demonstrated that reducing the density or effects of TAMs can inhibit the growth of tumors, making them targets for cancer immunotherapy, which has become a research hot spot. Several clinical and preclinical trials have studied drugs that inhibit the effects of and reduce the population of phagocytes that target TAMs achieve cancer immunotherapy. In this paper, we summarize the various methods of targeting TAMs for tumor immunotherapy, focusing on TAM mechanisms, sources, and polarization.
Collapse
Affiliation(s)
- Yongheng Shu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ping Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
111
|
Xu F, Wei Y, Tang Z, Liu B, Dong J. Tumor‑associated macrophages in lung cancer: Friend or foe? (Review). Mol Med Rep 2020; 22:4107-4115. [PMID: 33000214 PMCID: PMC7533506 DOI: 10.3892/mmr.2020.11518] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Typically, tumor-associated macrophages (TAMs), an abundant population of leukocytes in lung cancer, are affected by tumor microenvironment (TME) and shift towards either a pro-tumor (M2-like) or an anti-tumor phenotype (M1-like). M2-polarized macrophages, are one of the primary tumor-infiltrating immune cells and were reported to be associated with the promotion of cancer cell growth, invasion, metastasis, and angiogenesis. TAMs are considered a potential target for adjuvant anticancer therapies, and recent therapeutic approaches targeting the M2 polarization of TAMs have shown encouraging results. The present review discusses recent developments in the role of TAMs in cancer, in particular TAMs functions, clinical implication and prospective therapeutic strategies in lung cancer.
Collapse
Affiliation(s)
- Fei Xu
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Zhao Tang
- Department of Integrative Medicine, Huashan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Baojun Liu
- Department of Integrative Medicine, Huashan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital of Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
112
|
Tian X, Zhu X, Meng W, Bai S, Shi M, Xiang S, Zhao C, Wang Y. A 12-immune cell signature to predict relapse and guide chemotherapy for stage II colorectal cancer. Aging (Albany NY) 2020; 12:18363-18383. [PMID: 32855365 PMCID: PMC7585080 DOI: 10.18632/aging.103707] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/06/2020] [Indexed: 01/24/2023]
Abstract
The management of stage II colorectal cancer is still difficult. We aimed to construct a new immune cell-associated signature for prognostic evaluation and guiding chemotherapy in stage II colorectal cancer. We used the "Cell Type Identification by Estimating Relative Subsets of RNA Transcripts" (CIBERSORT) method to estimate the fraction of 22 immune cells by analyzing bulk tumor transcriptomes and a LASSO Cox regression model to select the prognostic immune cells. A 12-immune cell prognostic classifier, ISCRC, was built, which could successfully discriminate the high-risk patients in the training cohort (GSE39582: HR = 3.16, 95% CI: 1.85-5.40, P < 0.0001) and another independent cohorts (GSE14333: HR = 3.47, 95% CI: 1.18-10.15, P =0.0167). The receiver operating characteristic analysis revealed that the AUC of the ISCRC model was significantly greater than that of oncotypeDX model (0.7111 versus 0.5647, p=0.0152). We introduced the propensity score matching analysis to eliminate the selection bias; survival analysis showed relatively poor prognosis after chemotherapy in stage II CRC patients. Furthermore, a nomogram was built for clinicians and did well in the calibration plots. In conclusion, this immune cell-based signature could improve prognostic prediction and may help guide chemotherapy in stage II colorectal cancer patients.
Collapse
Affiliation(s)
- Xianglong Tian
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqiang Zhu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Wenying Meng
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiguang Bai
- Department of Internal Medicine, People's Hospital of Jinping County, Yunnan Province, China
| | - Min Shi
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shihao Xiang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Zhao
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yugang Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
113
|
HDAC7 is an actionable driver of therapeutic antibody resistance by macrophages from CLL patients. Oncogene 2020; 39:5756-5767. [PMID: 32709923 DOI: 10.1038/s41388-020-01394-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/15/2020] [Indexed: 11/09/2022]
Abstract
Resistance, to therapeutic antibodies used to treat chronic lymphocytic leukemia (CLL) patients is common. Monocyte-derived macrophages (MDMs) are a major effector of antitumour responses to therapeutic antibodies and we have previously reported that resistance to therapeutic antibodies, by MDMs, increases as CLL disease progresses. In this study, we examine the effect of a Class IIa-selective HDAC inhibitor (TMP195) on the phagocytic response to opsonised tumor cells or non-opsonised targets by MDMs derived from CLL patients. We report that TMP195 enhances phagocytic responses to antibody-opsonised CLL cells and E. coli within 30 min of treatment. The enhanced response is phenocopied by knockdown of the Class IIa HDAC, HDAC7, or by low concentrations of the pan-HDAC inhibitor, vorinostat. HDAC7 knockdown and inhibition induces hyperacetylation and hyperphosphorylation of Bruton's tyrosine kinase (BTK). Moreover, BTK inhibitors abrogated the enhanced response to HDAC7 inhibition. Our data show that HDAC7 is an actionable driver of resistance to therapeutic antibodies by MDMs derived from CLL patients.
Collapse
|
114
|
Leblond MM, Tillé L, Nassiri S, Gilfillan CB, Imbratta C, Schmittnaegel M, Ries CH, Speiser DE, Verdeil G. CD40 Agonist Restores the Antitumor Efficacy of Anti-PD1 Therapy in Muscle-Invasive Bladder Cancer in an IFN I/II-Mediated Manner. Cancer Immunol Res 2020; 8:1180-1192. [DOI: 10.1158/2326-6066.cir-19-0826] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/24/2020] [Accepted: 07/01/2020] [Indexed: 11/16/2022]
|
115
|
Beltraminelli T, De Palma M. Biology and therapeutic targeting of tumour-associated macrophages. J Pathol 2020; 250:573-592. [PMID: 32086811 DOI: 10.1002/path.5403] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/13/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
Macrophages sustain tumour progression by facilitating angiogenesis, promoting immunosuppression, and enhancing cancer cell invasion and metastasis. They also modulate tumour response to anti-cancer therapy in pre-clinical models. This knowledge has motivated the development of agents that target tumour-associated macrophages (TAMs), some of which have been investigated in early clinical trials. Here, we provide a comprehensive overview of the biology and therapeutic targeting of TAMs, highlighting opportunities, setbacks, and new challenges that have emerged after a decade of intense translational and clinical research into these multifaceted immune cells. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tim Beltraminelli
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
116
|
Kozlova N, Grossman JE, Iwanicki MP, Muranen T. The Interplay of the Extracellular Matrix and Stromal Cells as a Drug Target in Stroma-Rich Cancers. Trends Pharmacol Sci 2020; 41:183-198. [DOI: 10.1016/j.tips.2020.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/21/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022]
|
117
|
Garner H, de Visser KE. Immune crosstalk in cancer progression and metastatic spread: a complex conversation. Nat Rev Immunol 2020; 20:483-497. [PMID: 32024984 DOI: 10.1038/s41577-019-0271-z] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
Metastatic disease is responsible for approximately 90% of cancer deaths. For successful dissemination and metastasis, cancer cells must evade detection and destruction by the immune system. This process is enabled by factors secreted by the primary tumour that shape both the intratumoural microenvironment and the systemic immune landscape. Here, we review the evidence of aberrant immune cell crosstalk in metastasis formation and the role that primary tumours play in hijacking these interactions in order to enhance their metastatic potential. Moreover, we highlight the intriguing parallels between the inflammatory pathways underlying inflammatory disorders and cancer progression.
Collapse
Affiliation(s)
- Hannah Garner
- Division of Tumour Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Karin E de Visser
- Division of Tumour Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands. .,Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, Netherlands.
| |
Collapse
|
118
|
Davis CR, Than PA, Khong SML, Rodrigues M, Findlay MW, Navarrete DJ, Ghali S, Vaidya JS, Gurtner GC. Therapeutic Breast Reconstruction Using Gene Therapy-Delivered IFNγ Immunotherapy. Mol Cancer Ther 2020; 19:697-705. [PMID: 31658961 DOI: 10.1158/1535-7163.mct-19-0315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/26/2019] [Accepted: 10/21/2019] [Indexed: 11/16/2022]
Abstract
After mastectomy, breast reconstruction is increasingly performed using autologous tissue with the aim of improving quality of life. During this procedure, autologous tissue is excised, relocated, and reattached using microvascular anastomoses at the site of the extirpated breast. The period during which the tissue is ex vivo may allow genetic modification without any systemic exposure to the vector. Could such access permit delivery of therapeutic agents using the tissue flap as a vehicle? Such delivery may be more targeted and oncologically efficient than systemic therapy, and avoid systemic complications. The cytokine IFNγ has antitumor effects, and systemic toxicity could be circumvented by localized delivery of the IFNγ gene via gene therapy to autologous tissue used for breast reconstruction, which then releases IFNγ and exerts antitumor effects. In a rat model of loco-regional recurrence (LRR) with MADB-106-Luc and MAD-MB-231-Luc breast cancer cells, autologous tissue was transduced ex vivo with an adeno-associated viral vector encoding IFNγ. The "Therapeutic Reconstruction" released IFNγ at the LRR site and eliminated cancer cells, significantly decreased tumor burden, and increased survival compared with sham reconstruction (P <0.05). Mechanistically, localized IFNγ immunotherapy stimulated M1 macrophages to target cancer cells within the regional confines of the modified tumor environment. This concept of "Therapeutic Breast Reconstruction" using ex vivo gene therapy of autologous tissue offers a new application for immunotherapy in breast cancer with a dual therapeutic effect of both reconstructing the ablative defect and delivering local adjuvant immunotherapy.
Collapse
Affiliation(s)
- Christopher R Davis
- Stanford University School of Medicine, Stanford University, Stanford, California.
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Peter A Than
- Stanford University School of Medicine, Stanford University, Stanford, California
| | - Sacha M L Khong
- Stanford University School of Medicine, Stanford University, Stanford, California
| | - Melanie Rodrigues
- Stanford University School of Medicine, Stanford University, Stanford, California
| | - Michael W Findlay
- Stanford University School of Medicine, Stanford University, Stanford, California
- The Peter MacCallum Cancer Centre, Department of Surgery, The University of Melbourne, Melbourne, Australia
| | - Daniel J Navarrete
- Stanford University School of Medicine, Stanford University, Stanford, California
- Department of Microbiology and Immunology, Stanford University, Stanford, California
| | - Shadi Ghali
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Jayant S Vaidya
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Geoffrey C Gurtner
- Stanford University School of Medicine, Stanford University, Stanford, California.
| |
Collapse
|
119
|
Abstract
The progression of cancer is strongly influenced by the crosstalk between cancer cells and immune cells. Immune cells can have both pro- and anti-tumor functions depending on the signals present in the environment. A significant proportion of the immune compartment of most solid tumors consists of tumor-associated macrophages. Although their abundance has been associated with poor prognosis in many solid tumor types, the molecular mechanisms by which cancer cells influence macrophage phenotype and function are largely unknown. In this chapter, we provide a detailed description of in vitro assays to study the impact of cancer cells on macrophages. We provide protocols to obtain macrophages from murine bone marrow and human peripheral blood, and to expose these macrophages to cancer cell-derived secreted molecules using conditioned medium from cancer cells. We describe several assays to assess cancer cell-induced polarization of macrophages. This experimental set-up can be utilized to gain molecular insights into how cancer cells influence macrophages.
Collapse
|
120
|
Khandia R, Munjal A. Interplay between inflammation and cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 119:199-245. [DOI: 10.1016/bs.apcsb.2019.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
121
|
Current Strategies to Target Tumor-Associated-Macrophages to Improve Anti-Tumor Immune Responses. Cells 2019; 9:cells9010046. [PMID: 31878087 PMCID: PMC7017001 DOI: 10.3390/cells9010046] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
: Established evidence demonstrates that tumor-infiltrating myeloid cells promote rather than stop-cancer progression. Tumor-associated macrophages (TAMs) are abundantly present at tumor sites, and here they support cancer proliferation and distant spreading, as well as contribute to an immune-suppressive milieu. Their pro-tumor activities hamper the response of cancer patients to conventional therapies, such as chemotherapy or radiotherapy, and also to immunotherapies based on checkpoint inhibition. Active research frontlines of the last years have investigated novel therapeutic strategies aimed at depleting TAMs and/or at reprogramming their tumor-promoting effects, with the goal of re-establishing a favorable immunological anti-tumor response within the tumor tissue. In recent years, numerous clinical trials have included pharmacological strategies to target TAMs alone or in combination with other therapies. This review summarizes the past and current knowledge available on experimental tumor models and human clinical studies targeting TAMs for cancer treatment.
Collapse
|
122
|
Tian X, Wu Y, Yang Y, Wang J, Niu M, Gao S, Qin T, Bao D. Long noncoding RNA LINC00662 promotes M2 macrophage polarization and hepatocellular carcinoma progression via activating Wnt/β-catenin signaling. Mol Oncol 2019; 14:462-483. [PMID: 31785055 PMCID: PMC6998656 DOI: 10.1002/1878-0261.12606] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/29/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022] Open
Abstract
Tumor-associated macrophages have important roles in hepatocellular carcinoma (HCC) initiation and progression. Long noncoding RNAs (lncRNAs) have also been reported to be involved in HCC. In this study, we explored how lncRNA LINC00662 may influence HCC progression through both tumor cell-dependent and macrophage-dependent mechanisms. LINC00662 was found to be upregulated in HCC, and high LINC00662 levels correlated with poor survival of HCC patients. LINC00662 upregulated WNT3A expression and secretion via competitively binding miR-15a, miR-16, and miR-107. Through inducing WNT3A secretion, LINC00662 activated Wnt/β-catenin signaling in HCC cells in an autocrine manner and further promoted HCC cell proliferation, cell cycle, and tumor cell invasion, while repressing HCC cell apoptosis. In addition, acting through WNT3A secretion, LINC00662 activated Wnt/β-catenin signaling in macrophages in a paracrine manner and further promoted M2 macrophage polarization. Via activating Wnt/β-catenin signaling and M2 macrophages polarization, LINC00662 significantly promoted HCC tumor growth and metastasis in vivo. Hence, targeting LINC00662 may provide novel therapeutic strategy against HCC.
Collapse
Affiliation(s)
- Xiaohui Tian
- Department of Clinical Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, China
| | - Yuanyuan Wu
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, China
| | - Yating Yang
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, China
| | - Jiaxin Wang
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, China
| | - Menglan Niu
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, China
| | - Shanjun Gao
- Microbiome Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, China
| | - Tao Qin
- Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, China
| | - Dengke Bao
- Department of Clinical Laboratory, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, China.,Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, China.,Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, China
| |
Collapse
|
123
|
Abstract
Resistance to cancer therapy remains a major challenge in clinical oncology. Although the initial treatment phase is often successful, eventual resistance, characterized by tumour relapse or spread, is discouraging. The majority of studies devoted to investigating the basis of resistance have focused on tumour-related changes that contribute to therapy resistance and tumour aggressiveness. However, over the last decade, the diverse roles of various host cells in promoting therapy resistance have become more appreciated. A growing body of evidence demonstrates that cancer therapy can induce host-mediated local and systemic responses, many of which shift the delicate balance within the tumour microenvironment, ultimately facilitating or supporting tumour progression. In this Review, recent advances in understanding how the host response to different cancer therapies may promote therapy resistance are discussed, with a focus on therapy-induced immunological, angiogenic and metastatic effects. Also summarized is the potential of evaluating the host response to cancer therapy in an era of precision medicine in oncology.
Collapse
Affiliation(s)
- Yuval Shaked
- Department of Cell Biology and Cancer Science, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
124
|
Tuit S, Salvagno C, Kapellos TS, Hau CS, Seep L, Oestreich M, Klee K, de Visser KE, Ulas T, Schultze JL. Transcriptional Signature Derived from Murine Tumor-Associated Macrophages Correlates with Poor Outcome in Breast Cancer Patients. Cell Rep 2019; 29:1221-1235.e5. [PMID: 31665635 PMCID: PMC7057267 DOI: 10.1016/j.celrep.2019.09.067] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/23/2019] [Accepted: 09/20/2019] [Indexed: 12/20/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are frequently the most abundant immune cells in cancers and are associated with poor survival. Here, we generated TAM molecular signatures from K14cre;Cdh1flox/flox;Trp53flox/flox (KEP) and MMTV-NeuT (NeuT) transgenic mice that resemble human invasive lobular carcinoma (ILC) and HER2+ tumors, respectively. Determination of TAM-specific signatures requires comparison with healthy mammary tissue macrophages to avoid overestimation of gene expression differences. TAMs from the two models feature a distinct transcriptomic profile, suggesting that the cancer subtype dictates their phenotype. The KEP-derived signature reliably correlates with poor overall survival in ILC but not in triple-negative breast cancer patients, indicating that translation of murine TAM signatures to patients is cancer subtype dependent. Collectively, we show that a transgenic mouse tumor model can yield a TAM signature relevant for human breast cancer outcome prognosis and provide a generalizable strategy for determining and applying immune cell signatures provided the murine model reflects the human disease.
Collapse
Affiliation(s)
- Sander Tuit
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, 53113 Bonn, Germany; Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
| | - Camilla Salvagno
- Division of Tumor Biology & Immunology, Oncode Institute, the Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Theodore S Kapellos
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, 53113 Bonn, Germany
| | - Cheei-Sing Hau
- Division of Tumor Biology & Immunology, Oncode Institute, the Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Lea Seep
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, 53113 Bonn, Germany
| | - Marie Oestreich
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, 53113 Bonn, Germany
| | - Kathrin Klee
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, 53113 Bonn, Germany
| | - Karin E de Visser
- Division of Tumor Biology & Immunology, Oncode Institute, the Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands.
| | - Thomas Ulas
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, 53113 Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, 53113 Bonn, Germany; Platform for Single Cell Genomics and Epigenomics (PRECISE) at the German Center for Neurodegenerative Diseases and the University of Bonn, 53127 Bonn, Germany.
| |
Collapse
|
125
|
Abstract
New therapies that promote antitumour immunity have been recently developed. Most of these immunomodulatory approaches have focused on enhancing T-cell responses, either by targeting inhibitory pathways with immune checkpoint inhibitors, or by targeting activating pathways, as with chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to unprecedented successes, only a minority of patients with cancer benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for long-lasting, multilayered tumour control.
Collapse
|
126
|
Tsuchiya N, Zhang R, Iwama T, Ueda N, Liu T, Tatsumi M, Sasaki Y, Shimoda R, Osako Y, Sawada Y, Kubo Y, Miyashita A, Fukushima S, Cheng Z, Nakaki R, Takubo K, Okada S, Kaneko S, Ihn H, Kaisho T, Nishimura Y, Senju S, Endo I, Nakatsura T, Uemura Y. Type I Interferon Delivery by iPSC-Derived Myeloid Cells Elicits Antitumor Immunity via XCR1 + Dendritic Cells. Cell Rep 2019; 29:162-175.e9. [PMID: 31577946 DOI: 10.1016/j.celrep.2019.08.086] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/29/2019] [Accepted: 08/27/2019] [Indexed: 01/28/2023] Open
Abstract
Type I interferons (IFNs) play important roles in antitumor immunity. We generated IFN-α-producing cells by genetically engineered induced pluripotent stem cell (iPSC)-derived proliferating myeloid cells (iPSC-pMCs). Local administration of IFN-α-producing iPSC-pMCs (IFN-α-iPSC-pMCs) alters the tumor microenvironment and propagates the molecular signature associated with type I IFN. The gene-modified cell actively influences host XCR1+ dendritic cells to enhance CD8+ T cell priming, resulting in CXCR3-dependent and STING-IRF3 pathway-independent systemic tumor control. Administration of IFN-α-iPSC-pMCs in combination with immune checkpoint blockade overcomes resistance to single-treatment modalities and generates long-lasting antitumor immunity. These preclinical data suggest that IFN-α-iPSC-pMCs might constitute effective immune-stimulating agents for cancer that are refractory to checkpoint blockade.
Collapse
Affiliation(s)
- Nobuhiro Tsuchiya
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan; Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Rong Zhang
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan; Division of Immunology, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Tatsuaki Iwama
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Norihiro Ueda
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan; Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Tianyi Liu
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan; Key Laboratory of Cancer Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Minako Tatsumi
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Yutaka Sasaki
- Department of Stem Cell Biology and Regenerative Medicine, Graduate School of Medical Science, Kansai Medical University, Hirakata 573-1010, Japan
| | | | | | - Yu Sawada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Yosuke Kubo
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Azusa Miyashita
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Zhao Cheng
- Department of Hematology, Institute of Molecular Hematology, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan 410011, China
| | | | - Keiyo Takubo
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto 860-8556, Japan
| | - Shin Kaneko
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan.
| | - Itaru Endo
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan; Division of Immunology, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan.
| |
Collapse
|
127
|
Karagiannis GS, Condeelis JS, Oktay MH. Chemotherapy-Induced Metastasis: Molecular Mechanisms, Clinical Manifestations, Therapeutic Interventions. Cancer Res 2019; 79:4567-4576. [PMID: 31431464 PMCID: PMC6744993 DOI: 10.1158/0008-5472.can-19-1147] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/20/2019] [Accepted: 06/13/2019] [Indexed: 12/21/2022]
Abstract
Chemotherapy offers long-term clinical benefits to many patients with advanced cancer. However, recent evidence has linked the cytotoxic effects of chemotherapy with the de novo elicitation of a prometastatic tumor microenvironment. This "modified" tumor microenvironment is triggered by a chemotherapy-driven cytokine storm or through direct effects of certain chemotherapeutics on stromal and/or immune cells, the most critical being tumor-associated macrophages. These chemotherapy-educated cells act as facilitators in tumor-host cell interactions promoting the establishment of distant metastasis. Certain clinical studies now offer substantial evidence that prometastatic changes are indeed identified in the tumor microenvironment of certain patient subpopulations, especially those that do not present with any pathologic response after neoadjuvant chemotherapy. Deciphering the exact contextual prerequisites for chemotherapy-driven metastasis will be paramount for designing novel mechanism-based treatments for circumventing chemotherapy-induced metastasis.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York.
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
- Department of Surgery, Montefiore Medical Center, Bronx, New York
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York.
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
- Department of Pathology, Montefiore Medical Center, Bronx, New York
| |
Collapse
|
128
|
Chen SH, Chang JY. New Insights into Mechanisms of Cisplatin Resistance: From Tumor Cell to Microenvironment. Int J Mol Sci 2019; 20:ijms20174136. [PMID: 31450627 PMCID: PMC6747329 DOI: 10.3390/ijms20174136] [Citation(s) in RCA: 263] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022] Open
Abstract
Although cisplatin has been a pivotal chemotherapy drug in treating patients with various types of cancer for decades, drug resistance has been a major clinical impediment. In general, cisplatin exerts cytotoxic effects in tumor cells mainly through the generation of DNA-platinum adducts and subsequent DNA damage response. Accordingly, considerable effort has been devoted to clarify the resistance mechanisms inside tumor cells, such as decreased drug accumulation, enhanced detoxification activity, promotion of DNA repair capacity, and inactivated cell death signaling. However, recent advances in high-throughput techniques, cell culture platforms, animal models, and analytic methods have also demonstrated that the tumor microenvironment plays a key role in the development of cisplatin resistance. Recent clinical successes in combination treatments with cisplatin and novel agents targeting components in the tumor microenvironment, such as angiogenesis and immune cells, have also supported the therapeutic value of these components in cisplatin resistance. In this review, we summarize resistance mechanisms with respect to a single tumor cell and crucial components in the tumor microenvironment, particularly focusing on favorable results from clinical studies. By compiling emerging evidence from preclinical and clinical studies, this review may provide insights into the development of a novel approach to overcome cisplatin resistance.
Collapse
Affiliation(s)
- Shang-Hung Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan.
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
129
|
Di Mitri D, Mirenda M, Vasilevska J, Calcinotto A, Delaleu N, Revandkar A, Gil V, Boysen G, Losa M, Mosole S, Pasquini E, D'Antuono R, Masetti M, Zagato E, Chiorino G, Ostano P, Rinaldi A, Gnetti L, Graupera M, Martins Figueiredo Fonseca AR, Pereira Mestre R, Waugh D, Barry S, De Bono J, Alimonti A. Re-education of Tumor-Associated Macrophages by CXCR2 Blockade Drives Senescence and Tumor Inhibition in Advanced Prostate Cancer. Cell Rep 2019; 28:2156-2168.e5. [PMID: 31433989 PMCID: PMC6715643 DOI: 10.1016/j.celrep.2019.07.068] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/05/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor-associated macrophages (TAMs) represent a major component of the tumor microenvironment supporting tumorigenesis. TAMs re-education has been proposed as a strategy to promote tumor inhibition. However, whether this approach may work in prostate cancer is unknown. Here we find that Pten-null prostate tumors are strongly infiltrated by TAMs expressing C-X-C chemokine receptor type 2 (CXCR2), and activation of this receptor through CXCL2 polarizes macrophages toward an anti-inflammatory phenotype. Notably, pharmacological blockade of CXCR2 receptor by a selective antagonist promoted the re-education of TAMs toward a pro-inflammatory phenotype. Strikingly, CXCR2 knockout monocytes infused in Ptenpc-/-; Trp53pc-/- mice differentiated in tumor necrosis factor alpha (TNF-α)-releasing pro-inflammatory macrophages, leading to senescence and tumor inhibition. Mechanistically, PTEN-deficient tumor cells are vulnerable to TNF-α-induced senescence, because of an increase of TNFR1. Our results identify TAMs as targets in prostate cancer and describe a therapeutic strategy based on CXCR2 blockade to harness anti-tumorigenic potential of macrophages against this disease.
Collapse
Affiliation(s)
- Diletta Di Mitri
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Michela Mirenda
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland
| | | | | | - Nicolas Delaleu
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; Swiss Institute of Bioinformatics, Lausanne, Switzerland; 2C SysBioMed, 6646 Contra, Switzerland
| | | | - Veronica Gil
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Gunther Boysen
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Marco Losa
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland
| | - Simone Mosole
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland
| | | | - Rocco D'Antuono
- Institute for Research in Biomedicine (IRB), 6500 Bellinzona, Switzerland
| | - Michela Masetti
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Elena Zagato
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland
| | - Giovanna Chiorino
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia, Via Malta, 3, 13900 Biella, Italy
| | - Paola Ostano
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia, Via Malta, 3, 13900 Biella, Italy
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland
| | - Letizia Gnetti
- Pathology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Mariona Graupera
- Vascular Signalling Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Program Against Cancer Therapeutic Resistance (ProCURE), Barcelona, Spain; CIBERONC, Madrid, Spain
| | - Ana Raquel Martins Figueiredo Fonseca
- Vascular Signalling Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Program Against Cancer Therapeutic Resistance (ProCURE), Barcelona, Spain; CIBERONC, Madrid, Spain
| | - Ricardo Pereira Mestre
- Medical Oncology, Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland
| | - David Waugh
- Movember Centre of Excellence, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Simon Barry
- IMED Oncology AstraZeneca, Li KaShing Centre, Cambridge, UK
| | - Johann De Bono
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Faculty of Medicine, Università della Svizzera Italiana, 1011 Lugano, Switzerland; Department of Medicine, University of Padua, 35131 Padua, Italy; Medical Oncology, Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland.
| |
Collapse
|
130
|
Gomez-Roca CA, Italiano A, Le Tourneau C, Cassier PA, Toulmonde M, D'Angelo SP, Campone M, Weber KL, Loirat D, Cannarile MA, Jegg AM, Ries C, Christen R, Meneses-Lorente G, Jacob W, Klaman I, Ooi CH, Watson C, Wonde K, Reis B, Michielin F, Rüttinger D, Delord JP, Blay JY. Phase I study of emactuzumab single agent or in combination with paclitaxel in patients with advanced/metastatic solid tumors reveals depletion of immunosuppressive M2-like macrophages. Ann Oncol 2019; 30:1381-1392. [PMID: 31114846 PMCID: PMC8887589 DOI: 10.1093/annonc/mdz163] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Emactuzumab is a monoclonal antibody against the colony-stimulating factor-1 receptor and targets tumor-associated macrophages (TAMs). This study assessed the safety, clinical activity, pharmacokinetics (PK) and pharmacodynamics (PD) of emactuzumab, as monotherapy and in combination with paclitaxel, in patients with advanced solid tumors. PATIENTS AND METHODS This open-label, phase Ia/b study comprised two parts (dose escalation and dose expansion), each containing two arms (emactuzumab, every 2 or 3 weeks, as monotherapy or in combination with paclitaxel 80 mg/m2 weekly). The dose-escalation part explored the maximum tolerated dose and optimal biological dose (OBD). The dose-expansion part extended the safety assessment and investigated the objective response rate. A PK/PD analysis of serial blood, skin and tumor biopsies was used to explore proof of mechanism and confirm the OBD. RESULTS No maximum tolerated dose was reached in either study arm, and the safety profile of emactuzumab alone and in combination does not appear to preclude its use. No patients receiving emactuzumab monotherapy showed an objective response; the objective response rate for emactuzumab in combination with paclitaxel was 7% across all doses. Skin macrophages rather than peripheral blood monocytes or circulating colony-stimulating factor-1 were identified as an optimal surrogate PD marker to select the OBD. Emactuzumab treatment alone and in combination with paclitaxel resulted in a plateau of immunosuppressive TAM reduction at the OBD of 1000 mg administered every 2 weeks. CONCLUSIONS Emactuzumab showed specific reduction of immunosuppressive TAMs at the OBD in both treatment arms but did not result in clinically relevant antitumor activity alone or in combination with paclitaxel. (ClinicalTrials.gov Identifier: NCT01494688).
Collapse
Affiliation(s)
- C A Gomez-Roca
- Department of Medicine & Clinical Research Unit, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse (IUCT-Oncopole), Toulouse.
| | - A Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux.
| | - C Le Tourneau
- Department of Drug Development and Innovation, Institut Curie, Paris & Saint-Cloud; INSERM U900 Research Unit, Saint-Cloud; Paris-Saclay University, Paris
| | - P A Cassier
- Department of Medicine, Centre Léon Bérard, Lyon, France
| | - M Toulmonde
- Department of Medical Oncology, Institut Bergonié, Bordeaux
| | - S P D'Angelo
- Memorial Sloan Kettering Cancer Center, New York; Weill Cornell Medical College, New York, USA
| | - M Campone
- ICO René Gauducheau, Saint-Herblain, France
| | - K L Weber
- Department of Orthopedic Oncology, Penn Medicine, Pennsylvania, USA
| | - D Loirat
- Department of Drug Development and Innovation, Institut Curie, Paris & Saint-Cloud
| | - M A Cannarile
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - A-M Jegg
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - C Ries
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - R Christen
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - G Meneses-Lorente
- Roche Innovation Center Welwyn, Roche Pharmaceutical Research and Early Development, Welwyn Garden City
| | - W Jacob
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - I Klaman
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - C-H Ooi
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - C Watson
- A4P Consulting Ltd, Sandwich, UK
| | - K Wonde
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - B Reis
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - F Michielin
- Licensing and Early Development (LEAD) Safety Science, Roche Innovation Center Basel, Basel, Switzerland
| | - D Rüttinger
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - J-P Delord
- Department of Drug Development and Innovation, Institut Curie, Paris & Saint-Cloud
| | - J-Y Blay
- Department of Medicine & Clinical Research Unit, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse (IUCT-Oncopole), Toulouse
| |
Collapse
|
131
|
Burgos-Panadero R, Lucantoni F, Gamero-Sandemetrio E, Cruz-Merino LDL, Álvaro T, Noguera R. The tumour microenvironment as an integrated framework to understand cancer biology. Cancer Lett 2019; 461:112-122. [PMID: 31325528 DOI: 10.1016/j.canlet.2019.07.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/11/2019] [Accepted: 07/14/2019] [Indexed: 01/18/2023]
Abstract
Cancer cells all share the feature of being immersed in a complex environment with altered cell-cell/cell-extracellular element communication, physicochemical information, and tissue functions. The so-called tumour microenvironment (TME) is becoming recognised as a key factor in the genesis, progression and treatment of cancer lesions. Beyond genetic mutations, the existence of a malignant microenvironment forms the basis for a new perspective in cancer biology where connections at the system level are fundamental. From this standpoint, different aspects of tumour lesions such as morphology, aggressiveness, prognosis and treatment response can be considered under an integrated vision, giving rise to a new field of study and clinical management. Nowadays, somatic mutation theory is complemented with study of TME components such as the extracellular matrix, immune compartment, stromal cells, metabolism and biophysical forces. In this review we examine recent studies in this area and complement them with our own research data to propose a classification of stromal changes. Exploring these avenues and gaining insight into malignant phenotype remodelling, could reveal better ways to characterize this disease and its potential treatment.
Collapse
Affiliation(s)
- Rebeca Burgos-Panadero
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain; CIBERONC, Madrid, Spain
| | - Federico Lucantoni
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain
| | - Esther Gamero-Sandemetrio
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain; CIBERONC, Madrid, Spain
| | | | - Tomás Álvaro
- CIBERONC, Madrid, Spain; Hospital Verge de la Cinta, Tortosa, Tarragona, Spain.
| | - Rosa Noguera
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain; CIBERONC, Madrid, Spain.
| |
Collapse
|
132
|
Abstract
Recent studies have shown that genomic instability in tumor cells leads to activation of inflammatory signaling through the cGAS/STING pathway. In this review, we describe multiple ways by which genomic instability leads to cGAS/STING-mediated inflammatory signaling, as well as the consequences for tumor development and the tumor microenvironment. Also, we elaborate on how tumor cells have apparently evolved to escape the immune surveillance mechanisms that are triggered by cGAS/STING signaling. Finally, we describe how cGAS/STING-mediated inflammatory signaling can be therapeutically targeted to improve therapy responses.
Collapse
Affiliation(s)
- Francien Talens
- a Department of Medical Oncology, University Medical Center Groningen, University of Groningen , Groningen , the Netherlands
| | - Marcel A T M Van Vugt
- a Department of Medical Oncology, University Medical Center Groningen, University of Groningen , Groningen , the Netherlands
| |
Collapse
|
133
|
Sprooten J, Agostinis P, Garg AD. Type I interferons and dendritic cells in cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:217-262. [PMID: 31810554 DOI: 10.1016/bs.ircmb.2019.06.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type I interferons (IFNs) facilitate cancer immunosurveillance, antitumor immunity and antitumor efficacy of conventional cell death-inducing therapies (chemotherapy/radiotherapy) as well as immunotherapy. Moreover, it is clear that dendritic cells (DCs) play a significant role in aiding type I IFN-driven immunity. Owing to these antitumor properties several immunotherapies involving, or inducing, type I IFNs have received considerable clinical attention, e.g., recombinant IFNα2 or agonists targeting pattern recognition receptor (PRR) pathways like Toll-like receptors (TLRs), cGAS-STING or RIG-I/MDA5/MAVS. A series of preclinical and clinical evidence concurs that the success of anticancer therapy hinges on responsiveness of both cancer cells and DCs to type I IFNs. In this article, we discuss this link between type I IFNs and DCs in the context of cancer biology, with particular attention to mechanisms behind type I IFN production, their impact on DC driven anticancer immunity, and the implications of this for cancer immunotherapy, including DC-based vaccines.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Death Research & Therapy (CDRT) Unit, Department for Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Unit, Department for Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Unit, Department for Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
134
|
Singh S, Chakrabarti R. Consequences of EMT-Driven Changes in the Immune Microenvironment of Breast Cancer and Therapeutic Response of Cancer Cells. J Clin Med 2019; 8:jcm8050642. [PMID: 31075939 PMCID: PMC6572359 DOI: 10.3390/jcm8050642] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/30/2019] [Accepted: 05/04/2019] [Indexed: 02/06/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a process through which epithelial cells lose their epithelial characteristics and cell–cell contact, thus increasing their invasive potential. In addition to its well-known roles in embryonic development, wound healing, and regeneration, EMT plays an important role in tumor progression and metastatic invasion. In breast cancer, EMT both increases the migratory capacity and invasive potential of tumor cells, and initiates protumorigenic alterations in the tumor microenvironment (TME). In particular, recent evidence has linked increased expression of EMT markers such as TWIST1 and MMPs in breast tumors with increased immune infiltration in the TME. These immune cells then provide cues that promote immune evasion by tumor cells, which is associated with enhanced tumor progression and metastasis. In the current review, we will summarize the current knowledge of the role of EMT in the biology of different subtypes of breast cancer. We will further explore the correlation between genetic switches leading to EMT and EMT-induced alterations within the TME that drive tumor growth and metastasis, as well as their possible effect on therapeutic response in breast cancer.
Collapse
Affiliation(s)
- Snahlata Singh
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Rumela Chakrabarti
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
135
|
Crunkhorn S. Targeting macrophages enhances chemotherapy. Nat Rev Drug Discov 2019:10.1038/d41573-019-00059-y. [PMID: 31048804 DOI: 10.1038/d41573-019-00059-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
136
|
|