1
|
Zhang X, Sun P, Wang Y, Xu L, Li HY, Tong LJ, Tan C, Ding J, Yang CH, Meng LH. JMC14: a novel dual PI3Kδ/CSF1R inhibitor with potent antitumor activity in hematological and solid tumors. Acta Pharmacol Sin 2025:10.1038/s41401-025-01575-x. [PMID: 40389566 DOI: 10.1038/s41401-025-01575-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/22/2025] [Indexed: 05/21/2025]
Abstract
PI3Kδ, predominantly expressed in immune cells and markedly dysregulated in B-cell malignancies, emerges as a promising and well-validated therapeutic target in hematologic cancers. Meanwhile, CSF1R regulates the formation and polarization of tumor-associated macrophages (TAMs), facilitating immune suppression and tumor progression in various solid tumors. Although targeting PI3Kδ or CSF1R has shown promise, the clinical application is often constrained by off-target effects, toxicity, and limited efficacy, particularly in solid malignancies. In this study, we identified JMC14, a novel dual inhibitor targeting PI3Kδ and CSF1R with a distinct structure and favorable selectivity among human kinome, yielding IC50 values of 12 nM against PI3Kδ and 143 nM against CSF1R, respectively. JMC14 preferentially inhibited PI3Kδ-mediated signaling at the cellular level and exhibited robust antiproliferative activity across 10 lines of diffuse large B-cell lymphoma (DLBCL) cells, outperforming the approved PI3Kδ inhibitor idelalisib. Notably, its efficacy negatively correlated with the PI3Kα expression among the cell lines tested, suggesting a compensatory pathway mediated by PI3Kα. Daily oral administration of JMC14 (10, 30, or 100 mg/kg, for 21 days) dose-dependently suppressed tumor progression in xenografts derived from TMD8 cells and DLBCL patients, accompanied by good tolerance. Additionally, M-NFS-60 myeloid leukemia cells, which are dependent on the CSF-1-CSF1R axis for survival and proliferation, were effectively inhibited by JMC14 both in vitro and in vivo, further validating its inhibitory activity targeting CSF1R. Furthermore, JMC14 demonstrated potent antitumor activity in murine triple-negative breast cancer (TNBC), which was associated with its activity to reshape the immune microenvironment by reducing M2-like TAMs, enhancing CD8+ T cell infiltration. Collectively, these findings establish JMC14 as a potent dual PI3Kδ/CSF1R inhibitor with remarkable efficacy against both hematologic and solid malignancies with hyperactivation of PI3Kδ and/or CSF1R, highlighting the potential of JMC14 as a useful probe to dissect the interaction of PI3Kδ and CSF1R in tumor progression and immune reprogramming.
Collapse
Affiliation(s)
- Xi Zhang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Pu Sun
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi Wang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lan Xu
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hui-Yu Li
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lin-Jiang Tong
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Cun Tan
- Division of Small-molecule Drug Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jian Ding
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Chun-Hao Yang
- Division of Small-molecule Drug Research Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Ling-Hua Meng
- Division of Anti-tumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
2
|
Salarian M, Liu S, Tsai HM, Leslie SN, Hayes T, Lo ST, Szardenings AK, Zhang W, Chen G, Sandiego C, Wells L, Nair DG, Kolb HC, Xia CA. Evaluation of [ 18F]JNJ-CSF1R-1 as a Positron Emission Tomography Ligand Targeting Colony-Stimulating Factor 1 Receptor. Mol Imaging Biol 2025; 27:163-172. [PMID: 40009327 DOI: 10.1007/s11307-025-01991-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/09/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE Colony-stimulating factor 1 receptor (CSF1R) signaling plays a pivotal role in neuroinflammation, driving microglia proliferation and activation. CSF1R is considered a hallmark of inflammation in many neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD). Our study aims to evaluate the potential value of 5-cyano-N-(4-(4-(2-([18F]fluoro)ethyl)piperazin-1-yl)-2-(piperidin-1-yl)phenyl)furan-2-carboxamide ([18F]JNJ-CSF1R-1) as a positron emission tomography (PET) ligand targeting CSF1R in preclinical models of neuroinflammation. PROCEDURES A cell-based MSD assay was used to measure the IC50 of 5-cyano-N-(4-(4-(2-(fluoro)ethyl)piperazin-1-yl)-2-(piperidin-1-yl)phenyl)furan-2-carboxamide (JNJ-CSF1R-1). JNJ-CSF1R-1 was radiolabeled with fluorine-18. PET imaging was used to evaluate brain uptake, and target engagement of [18F]JNJ-CSF1R-1 in two neuroinflammation mouse models, including systemic lipopolysaccharide (LPS) and AppSAA knock in (KI). CSF1R protein levels in brain tissue were determined by western blot and ELISA assays. [18F]JNJ-CSF1R-1 brain uptake was also measured in a non-human primate (NHP) PET study. RESULTS JNJ-CSF1R-1 is a 12 nM (IC50) inhibitor of CSF1R. [18F]JNJ-CSF1R-1 demonstrated significantly higher brain uptake in both LPS and AD mouse models as measured by the area under the time activity curves (AUC) compared to control animals. In the AppSAA KI model, CSF1R levels increased near amyloid plaques as detected by IHC. [18F]JNJ-CSF1R-1 PET imaging signal showed a good correlation with CSF1R expression levels measured by western blot and ELISA. In an NHP study, [18F]JNJ-CSF1R-1 readily entered the brain and demonstrated reversible kinetics. CONCLUSION [18F]JNJ-CSF1R-1 is a potent and promising CSF1R PET tracer with translational potential for measuring microglia-based neuroinflammatory processes and for tracking the impact of anti-inflammatory therapies.
Collapse
Affiliation(s)
- Mani Salarian
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Shuanglong Liu
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Hsiu-Ming Tsai
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Shannon N Leslie
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Thomas Hayes
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Su-Tang Lo
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
- RayzeBio a Bristol Myers Squibb's Company, San Diego, CA, USA
| | | | - Wei Zhang
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
- RemeGen Biosciences, Inc, San Francisco, CA, USA
| | - Gang Chen
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
- US Rad Bio LLC, San Diego, CA, USA
| | | | | | - Dileep G Nair
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
- Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Institute of Molecular Pathobiochemistry, Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital Aachen, Aachen, Germany
| | - Hartmuth C Kolb
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
- Enigma Biomedical Group, Knoxville, TN, USA
| | - Chunfang A Xia
- Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA.
| |
Collapse
|
3
|
Izuka S, Komai T, Tsuchida Y, Tsuchiya H, Okamura T, Fujio K. The role of monocytes and macrophages in idiopathic inflammatory myopathies: insights into pathogenesis and potential targets. Front Immunol 2025; 16:1567833. [PMID: 40181992 PMCID: PMC11965591 DOI: 10.3389/fimmu.2025.1567833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are heterogeneous autoimmune disorders characterized by muscle inflammation, weakness, and extramuscular manifestations such as interstitial lung disease, skin rash, arthritis, dysphagia, myocarditis and other systemic organ involvement. Although T and B cells have historically been central to the understanding of IIM immunopathology, monocytes and their differentiated progenitor cells, macrophages, are increasingly being recognized as critical mediators of both tissue damage and repair. In subtypes such as dermatomyositis, immune-mediated necrotizing myopathy and antisynthetase syndrome, macrophages infiltrate skeletal muscle and other affected tissues, contributing to inflammation via production of pro-inflammatory cytokines, chemokines, and reactive oxygen species. Dysregulated interferon signaling, mitochondrial stress, and aberrant metabolic states in these cells further perpetuate tissue injury in IIMs. Conversely, certain macrophage subsets can support muscle fiber regeneration and dampen inflammation, underscoring the dual roles these cells can play. Future research into the heterogeneity of monocytes and macrophages, including single-cell transcriptomic and metabolomic approaches, will help clarify disease mechanisms, identify biomarkers of disease activity and prognosis, and guide novel therapeutic strategies targeting these innate immune cells in IIM.
Collapse
Affiliation(s)
- Shinji Izuka
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshihiko Komai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yumi Tsuchida
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruka Tsuchiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Hacıosmanoğlu-Aldoğan E, Lama D, Yetke Hİ, Şenol H, Yöntem FD. Necroptotic Suppression of Lung Cancer Cell Proliferation and Migration: A Comprehensive In Vitro and In Silico Study to Determine New Molecular Targets for Pexidartinib. Cell Biochem Funct 2025; 43:e70068. [PMID: 40079395 DOI: 10.1002/cbf.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/19/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
In this study, the cytotoxic effects of pexidartinib (PLX), a tyrosine kinase inhibitor approved for tenosynovial giant cell tumor through inhibition of colony-stimulating factor 1 receptor (CSF1R), against A549 lung adenocarcinoma cells and Beas-2B healthy bronchial cells were investigated by in detailed in-vitro and in-silico studies. Through MTT assays, PLX demonstrated significant inhibition of A549 cell viability with IC50 values of 2.15 and 1.3 µM at 24 and 48 h, respectively, while having minimal effects on Beas-2B cells, with IC50 values of 36.2 and 9.3 µM. The high selectivity index indicates PLX's preferential action against cancerous cells. The mechanism of cell death induced by PLX was further explored using Annexin V/PI staining and flow cytometry, revealing that PLX primarily induces necrosis in A549 cells, with an increase in necrotic cell populations and reduced efficacy at higher concentrations. Western blot analysis showed an upregulation of necroptosis markers (RIP3 and pMLKL) in A549 cells, while apoptotic markers like Caspase-3 remained unchanged. In addition, wound healing assays demonstrated that PLX significantly inhibits A549 cell migration in a dose-dependent manner. Molecular docking studies identified key amino acids involved in PLX binding interactions with target proteins. RIPK1 showed the strongest binding affinity. MD simulations revealed that the PLX-VEGFR2 complex was the most stable. As conclusion, PLX, although approved for tenosynovial giant cell tumors, shows promising potential for lung adenocarcinoma treatment. It selectively inhibits cancer cell viability, induces necroptosis, and reduces cell migration. Its stronger binding to RIPK1 and VEGFR2 more than CSF1R.
Collapse
Affiliation(s)
- Ebru Hacıosmanoğlu-Aldoğan
- Department of Biophysics, Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Fatih, Istanbul, Türkiye
| | - Dilhan Lama
- Faculty of Medicine, Bezmialem Vakif University, Fatih, Istanbul, Türkiye
| | - Hande İpek Yetke
- Department of Biophysics, Faculty of Medicine, Bezmialem Vakif University, Fatih, Istanbul, Türkiye
| | - Halil Şenol
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bezmialem Vakif University, Fatih, Istanbul, Türkiye
| | - Fulya Dal Yöntem
- Department of Biophysics, School of Medicine, Koç University, Sarıyer, Istanbul, Türkiye
- Koç University Research Center for Translational Medicine (KUTTAM), Sarıyer, Istanbul, Türkiye
| |
Collapse
|
5
|
Jumaniyazova E, Lokhonina A, Dzhalilova D, Miroshnichenko E, Kosyreva A, Fatkhudinov T. The Role of Macrophages in Various Types of Tumors and the Possibility of Their Use as Targets for Antitumor Therapy. Cancers (Basel) 2025; 17:342. [PMID: 39941714 PMCID: PMC11815841 DOI: 10.3390/cancers17030342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/07/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
In solid tumors, tumor-associated macrophages (TAMs) are one of the most numerous populations and play an important role in the processes of tumor cell invasion, metastasis, and angiogenesis. Therefore, TAMs are considered promising diagnostic and prognostic biomarkers of tumors, and many attempts have been made to influence these cells as part of antitumor therapy. There are several key principles of action on ТАМs: the inhibition of monocyte/macrophage transition; the destruction of macrophages; the reprogramming of macrophage phenotypes (polarization of M2 macrophages to M1); the stimulation of phagocytic activity of macrophages and CAR-M therapy. Despite the large number of studies in this area, to date, there are no adequate approaches using antitumor therapy based on alterations in TAM functioning that would show high efficacy when administered in a mono-regimen for the treatment of malignant neoplasms. Studies devoted to the evaluation of the efficacy of drugs acting on TAMs are characterized by a small sample and the large heterogeneity of patient groups; in addition, in such studies, chemotherapy or immunotherapy is used, which significantly complicates the evaluation of the effectiveness of the agent acting on TAMs. In this review, we attempted to systematize the evidence on attempts to influence TAMs in malignancies such as lung cancer, breast cancer, colorectal cancer, cervical cancer, prostate cancer, gastric cancer, head and neck squamous cell cancer, and soft tissue sarcomas.
Collapse
Affiliation(s)
- Enar Jumaniyazova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
| | - Anastasiya Lokhonina
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Avtsyn Research Institute of Human Morphology, FSBSI Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov, Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997 Moscow, Russia
| | - Dzhuliia Dzhalilova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Avtsyn Research Institute of Human Morphology, FSBSI Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Ekaterina Miroshnichenko
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Avtsyn Research Institute of Human Morphology, FSBSI Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Anna Kosyreva
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Avtsyn Research Institute of Human Morphology, FSBSI Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
- Avtsyn Research Institute of Human Morphology, FSBSI Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov, Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997 Moscow, Russia
| |
Collapse
|
6
|
Li Z, Liu H, Xie Q, Yin G. Macrophage involvement in idiopathic inflammatory myopathy: pathogenic mechanisms and therapeutic prospects. J Inflamm (Lond) 2024; 21:48. [PMID: 39593038 PMCID: PMC11590228 DOI: 10.1186/s12950-024-00422-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Idiopathic inflammatory myopathies are a group of systemic autoimmune diseases characterized by chronic muscle inflammation and diverse clinical manifestations. Macrophages, pivotal components of innate immunity, are implicated in immune responses, inflammation resolution, and tissue repair. Distinct macrophage polarization states play vital roles in disease progression and resolution. Mechanistically, activated macrophages release proinflammatory cytokines, chemokines, and reactive oxygen species, perpetuating immune responses and tissue damage. Dysregulated macrophage polarization contributes to sustained inflammation. Here, we reviewed the intricate contributions of macrophages to IIM pathogenesis and explored novel therapeutic avenues. We discussed emerging strategies targeting macrophages, including receptor-based interventions and macrophage polarization modulation, for IIM treatment. This review underscores the multifaceted involvement of macrophages in IIM pathogenesis and offers insights into potential therapeutic approaches targeting these immune cells for disease management.
Collapse
Affiliation(s)
- Ziqi Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, China
| | - Huan Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, China
| | - Geng Yin
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, China.
- Department of General Practice, West China Hospital, General Practice Medical Center, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Moua T, Baqir M, Ryu JH. What Is on the Horizon for Treatments in Idiopathic Pulmonary Fibrosis? J Clin Med 2024; 13:6304. [PMID: 39518443 PMCID: PMC11546700 DOI: 10.3390/jcm13216304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and often fatal lung disease most commonly encountered in older individuals. Several decades of research have contributed to a better understanding of its pathogenesis, though only two drugs thus far have shown treatment efficacy, i.e., by slowing the decline of lung function. The pathogenesis of IPF remains incompletely understood and involves multiple complex interactions and mechanisms working in tandem or separately to result in unchecked deposition of extracellular matrix components and collagen characteristic of the disease. These mechanisms include aberrant response to injury in the alveolar epithelium, inappropriate communication between epithelial cells and mesenchymal cells, imbalances between oxidative injury and tissue repair, recruitment of inflammatory pathways that induce fibrosis, and cell senescence leading to sustained activation and proliferation of fibroblasts and myofibroblasts. Targeted approaches to each of these mechanistic pathways have led to recent clinical studies evaluating the safety and efficacy of several agents. This review highlights selected concepts in the pathogenesis of IPF as a rationale for understanding current or future therapeutic approaches, followed by a review of several selected agents and their recent or active clinical studies. Current novel therapies include approaches to attenuating or modifying specific cellular or signaling processes in the fibrotic pathway, modifying inflammatory and metabolic derangements, and minimizing inappropriate cell senescence.
Collapse
Affiliation(s)
- Teng Moua
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; (M.B.); (J.H.R.)
| | | | | |
Collapse
|
8
|
Tomassetti C, Insinga G, Gimigliano F, Morrione A, Giordano A, Giurisato E. Insights into CSF-1R Expression in the Tumor Microenvironment. Biomedicines 2024; 12:2381. [PMID: 39457693 PMCID: PMC11504891 DOI: 10.3390/biomedicines12102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/30/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The colony-stimulating factor 1 receptor (CSF-1R) plays a pivotal role in orchestrating cellular interactions within the tumor microenvironment (TME). Although the CSF-1R has been extensively studied in myeloid cells, the expression of this receptor and its emerging role in other cell types in the TME need to be further analyzed. This review explores the multifaceted functions of the CSF-1R across various TME cellular populations, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), cancer-associated fibroblasts (CAFs), endothelial cells (ECs), and cancer stem cells (CSCs). The activation of the CSF-1R by its ligands, colony-stimulating factor 1 (CSF-1) and Interleukin-34 (IL-34), regulates TAM polarization towards an immunosuppressive M2 phenotype, promoting tumor progression and immune evasion. Similarly, CSF-1R signaling influences MDSCs to exert immunosuppressive functions, hindering anti-tumor immunity. In DCs, the CSF-1R alters antigen-presenting capabilities, compromising immune surveillance against cancer cells. CSF-1R expression in CAFs and ECs regulates immune modulation, angiogenesis, and immune cell trafficking within the TME, fostering a pro-tumorigenic milieu. Notably, the CSF-1R in CSCs contributes to tumor aggressiveness and therapeutic resistance through interactions with TAMs and the modulation of stemness features. Understanding the diverse roles of the CSF-1R in the TME underscores its potential as a therapeutic target for cancer treatment, aiming at disrupting pro-tumorigenic cellular crosstalk and enhancing anti-tumor immune responses.
Collapse
Affiliation(s)
- Caterina Tomassetti
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Gaia Insinga
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.I.); (F.G.)
| | - Francesca Gimigliano
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.I.); (F.G.)
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| |
Collapse
|
9
|
Li SL, Hou HY, Chu X, Zhu YY, Zhang YJ, Duan MD, Liu J, Liu Y. Nanomaterials-Involved Tumor-Associated Macrophages' Reprogramming for Antitumor Therapy. ACS NANO 2024; 18:7769-7795. [PMID: 38420949 DOI: 10.1021/acsnano.3c12387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Tumor-associated macrophages (TAMs) play pivotal roles in tumor development. As primary contents of tumor environment (TME), TAMs secrete inflammation-related substances to regulate tumoral occurrence and development. There are two kinds of TAMs: the tumoricidal M1-like TAMs and protumoral M2-like TAMs. Reprogramming TAMs from immunosuppressive M2 to immunocompetent M1 phenotype is considered a feasible way to improve immunotherapeutic efficiency. Notably, nanomaterials show great potential for biomedical fields due to their controllable structures and properties. There are many types of nanomaterials that exhibit great regulatory activities for TAMs' reprogramming. In this review, the recent progress of nanomaterials-involved TAMs' reprogramming is comprehensively discussed. The various nanomaterials for TAMs' reprogramming and the reprogramming strategies are summarized and introduced. Additionally, the challenges and perspectives of TAMs' reprogramming for efficient therapy are discussed, aiming to provide inspiration for TAMs' regulator design and promote the development of TAMs-mediated immunotherapy.
Collapse
Affiliation(s)
- Shu-Lan Li
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Hua-Ying Hou
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Xu Chu
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Ying Zhu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Juan Zhang
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Meng-Die Duan
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Junyi Liu
- Albany Medical College, New York 12208, United States
| | - Yi Liu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, P. R. China
| |
Collapse
|
10
|
Elfstrum AK, Bapat AS, Schwertfeger KL. Defining and targeting macrophage heterogeneity in the mammary gland and breast cancer. Cancer Med 2024; 13:e7053. [PMID: 38426622 PMCID: PMC10905685 DOI: 10.1002/cam4.7053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION Macrophages are innate immune cells that are associated with extensive phenotypic and functional plasticity and contribute to normal development, tissue homeostasis, and diseases such as cancer. In this review, we discuss the heterogeneity of tissue resident macrophages in the normal mammary gland and tumor-associated macrophages in breast cancer. Tissue resident macrophages are required for mammary gland development, where they have been implicated in promoting extracellular matrix remodeling, apoptotic clearance, and cellular crosstalk. In the context of cancer, tumor-associated macrophages are key drivers of growth and metastasis via their ability to promote matrix remodeling, angiogenesis, lymphangiogenesis, and immunosuppression. METHOD We identified and summarized studies in Pubmed that describe the phenotypic and functional heterogeneity of macrophages and the implications of targeting individual subsets, specifically in the context of mammary gland development and breast cancer. We also identified and summarized recent studies using single-cell RNA sequencing to identify and describe macrophage subsets in human breast cancer samples. RESULTS Advances in single-cell RNA sequencing technologies have yielded nuances in macrophage heterogeneity, with numerous macrophage subsets identified in both the normal mammary gland and breast cancer tissue. Macrophage subsets contribute to mammary gland development and breast cancer progression in differing ways, and emerging studies highlight a role for spatial localization in modulating their phenotype and function. CONCLUSION Understanding macrophage heterogeneity and the unique functions of each subset in both normal mammary gland development and breast cancer progression may lead to more promising targets for the treatment of breast cancer.
Collapse
Affiliation(s)
- Alexis K. Elfstrum
- Microbiology, Immunology, and Cancer Biology Graduate ProgramUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Aditi S. Bapat
- Molecular Pharmacology and Therapeutics Graduate ProgramUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Kathryn L. Schwertfeger
- Department of Laboratory Medicine and PathologyUniversity of MinnesotaMinneapolisMinnesotaUSA
- Masonic Cancer CenterUniversity of MinnesotaMinneapolisMinnesotaUSA
- Center for ImmunologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
11
|
Vieira GDS, Kimura TDC, Scarini JF, de Lima-Souza RA, Lavareze L, Emerick C, Gonçalves MT, Damas II, Figueiredo-Maciel T, Sales de Sá R, Aquino IG, Gonçalves de Paiva JP, Fernandes PM, Gonçalves MWA, Kowalski LP, Altemani A, Fillmore GC, Mariano FV, Egal ESA. Hematopoietic colony-stimulating factors in head and neck cancers: Recent advances and therapeutic challenges. Cytokine 2024; 173:156417. [PMID: 37944421 DOI: 10.1016/j.cyto.2023.156417] [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: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Colony-stimulating factors (CSFs) are key cytokines responsible for the production, maturation, and mobilization of the granulocytic and macrophage lineages from the bone marrow, which have been gaining attention for playing pro- and/or anti-tumorigenic roles in cancer. Head and neck cancers (HNCs) represent a group of heterogeneous neoplasms with high morbidity and mortality worldwide. Treatment for HNCs is still limited even with the advancements in cancer immunotherapy. Novel treatments for patients with recurrent and metastatic HNCs are urgently needed. This article provides an in-depth review of the role of hematopoietic cytokines such as granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF), and interleukin-3 (IL-3; also known as multi-CSF) in the HNCs tumor microenvironment. We have reviewed current results from clinical trials using CSFs as adjuvant therapy to treat HNCs patients, and also clinical findings reported to date on the therapeutic application of CSFs toxicities arising from chemoradiotherapy.
Collapse
Affiliation(s)
- Gustavo de Souza Vieira
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Talita de Carvalho Kimura
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - João Figueira Scarini
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Reydson Alcides de Lima-Souza
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Luccas Lavareze
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Carolina Emerick
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Mayara Trevizol Gonçalves
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Ingrid Iara Damas
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Tayná Figueiredo-Maciel
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Raisa Sales de Sá
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Iara Gonçalves Aquino
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - João Paulo Gonçalves de Paiva
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Patrícia Maria Fernandes
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Moisés Willian Aparecido Gonçalves
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery, School of Medicine, University of São Paulo (USP), São Paulo, Brazil; Department of Head and Neck Surgery and Otolaryngology, AC Camargo Cancer Center, São Paulo, Brazil
| | - Albina Altemani
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Gary Chris Fillmore
- Biorepository and Molecular Pathology, Huntsman Cancer Institute, University of Utah (UU), Salt Lake City, UT, United States
| | - Fernanda Viviane Mariano
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Erika Said Abu Egal
- Department of Pathology, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Biorepository and Molecular Pathology, Huntsman Cancer Institute, University of Utah (UU), Salt Lake City, UT, United States.
| |
Collapse
|
12
|
McWhorter R, Bonavida B. The Role of TAMs in the Regulation of Tumor Cell Resistance to Chemotherapy. Crit Rev Oncog 2024; 29:97-125. [PMID: 38989740 DOI: 10.1615/critrevoncog.2024053667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Tumor-associated macrophages (TAMs) are the predominant cell infiltrate in the immunosuppressive tumor microenvironment (TME). TAMs are central to fostering pro-inflammatory conditions, tumor growth, metastasis, and inhibiting therapy responses. Many cancer patients are innately refractory to chemotherapy and or develop resistance following initial treatments. There is a clinical correlation between the level of TAMs in the TME and chemoresistance. Hence, the pivotal role of TAMs in contributing to chemoresistance has garnered significant attention toward targeting TAMs to reverse this resistance. A prerequisite for such an approach requires a thorough understanding of the various underlying mechanisms by which TAMs inhibit response to chemotherapeutic drugs. Such mechanisms include enhancing drug efflux, regulating drug metabolism and detoxification, supporting cancer stem cell (CSCs) resistance, promoting epithelial-mesenchymal transition (EMT), inhibiting drug penetration and its metabolism, stimulating angiogenesis, impacting inhibitory STAT3/NF-κB survival pathways, and releasing specific inhibitory cytokines including TGF-β and IL-10. Accordingly, several strategies have been developed to overcome TAM-modulated chemoresistance. These include novel therapies that aim to deplete TAMs, repolarize them toward the anti-tumor M1-like phenotype, or block recruitment of monocytes into the TME. Current results from TAM-targeted treatments have been unimpressive; however, the use of TAM-targeted therapies in combination appears promising These include targeting TAMs with radiotherapy, chemotherapy, chemokine receptor inhibitors, immunotherapy, and loaded nanoparticles. The clinical limitations of these strategies are discussed.
Collapse
Affiliation(s)
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
13
|
Altomonte S, Pike VW. Candidate Tracers for Imaging Colony-Stimulating Factor 1 Receptor in Neuroinflammation with Positron Emission Tomography: Issues and Progress. ACS Pharmacol Transl Sci 2023; 6:1632-1650. [PMID: 37974622 PMCID: PMC10644394 DOI: 10.1021/acsptsci.3c00213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Indexed: 11/19/2023]
Abstract
The tyrosine kinase, colony-stimulating factor 1 receptor (CSF1R), has attracted attention as a potential biomarker of neuroinflammation for imaging studies with positron emission tomography (PET), especially because of its location on microglia and its role in microglia proliferation. The development of an effective radiotracer for specifically imaging and quantifying brain CSF1R is highly challenging. Here we review the progress that has been made on PET tracer development and discuss issues that have arisen and which remain to be addressed and resolved.
Collapse
Affiliation(s)
- Stefano Altomonte
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes
of Health, Building 10,
B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes
of Health, Building 10,
B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| |
Collapse
|
14
|
Huang G, Cierpicki T, Grembecka J. 2-Aminobenzothiazoles in anticancer drug design and discovery. Bioorg Chem 2023; 135:106477. [PMID: 36989736 PMCID: PMC10718064 DOI: 10.1016/j.bioorg.2023.106477] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/26/2023] [Accepted: 03/10/2023] [Indexed: 03/22/2023]
Abstract
Cancer is one of the major causes of mortality and morbidity worldwide. Substantial research efforts have been made to develop new chemical entities with improved anticancer efficacy. 2-Aminobenzothiazole is an important class of heterocycles containing one sulfur and two nitrogen atoms, which is associated with a broad spectrum of medical and pharmacological activities, including antitumor, antibacterial, antimalarial, anti-inflammatory, and antiviral activities. In recent years, an extraordinary collection of potent and low-toxicity 2-aminobenzothiazole compounds have been discovered as new anticancer agents. Herein, we provide a comprehensive review of this class of compounds based on their activities against tumor-related proteins, including tyrosine kinases (CSF1R, EGFR, VEGFR-2, FAK, and MET), serine/threonine kinases (Aurora, CDK, CK, RAF, and DYRK2), PI3K kinase, BCL-XL, HSP90, mutant p53 protein, DNA topoisomerase, HDAC, NSD1, LSD1, FTO, mPGES-1, SCD, hCA IX/XII, and CXCR. In addition, the anticancer potentials of 2-aminobenzothiazole-derived chelators and metal complexes are also described here. Moreover, the design strategies, mechanism of actions, structure-activity relationships (SAR) and more advanced stages of pre-clinical development of 2-aminobenzothiazoles as new anticancer agents are extensively reviewed in this article. Finally, the examples that 2-aminobenzothiazoles showcase an advantage over other heterocyclic systems are also highlighted.
Collapse
Affiliation(s)
- Guang Huang
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
15
|
Aarhus TI, Bjørnstad F, Wolowczyk C, Larsen KU, Rognstad L, Leithaug T, Unger A, Habenberger P, Wolf A, Bjørkøy G, Pridans C, Eickhoff J, Klebl B, Hoff BH, Sundby E. Synthesis and Development of Highly Selective Pyrrolo[2,3- d]pyrimidine CSF1R Inhibitors Targeting the Autoinhibited Form. J Med Chem 2023; 66:6959-6980. [PMID: 37191268 DOI: 10.1021/acs.jmedchem.3c00428] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Colony-stimulating factor-1 receptor (CSF1R) is a receptor tyrosine kinase that controls the differentiation and maintenance of most tissue-resident macrophages, and the inhibition of CSF1R has been suggested as a possible therapy for a range of human disorders. Herein, we present the synthesis, development, and structure-activity relationship of a series of highly selective pyrrolo[2,3-d]pyrimidines, showing subnanomolar enzymatic inhibition of this receptor and with excellent selectivity toward other kinases in the platelet-derived growth factor receptor (PDGFR) family. The crystal structure of the protein and 23 revealed that the binding conformation of the protein is DFG-out-like. The most promising compounds in this series were profiled for cellular potency and subjected to pharmacokinetic profiling and in vivo stability, indicating that this compound class could be relevant in a potential disease setting. Additionally, these compounds inhibited primarily the autoinhibited form of the receptor, contrasting the behavior of pexidartinib, which could explain the exquisite selectivity of these structures.
Collapse
Affiliation(s)
- Thomas Ihle Aarhus
- Department of Materials Science & Engineering, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Frithjof Bjørnstad
- Department of Materials Science & Engineering, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Camilla Wolowczyk
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | | | - Line Rognstad
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Trygve Leithaug
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Anke Unger
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Peter Habenberger
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Alexander Wolf
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Geir Bjørkøy
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Clare Pridans
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, U.K
| | - Jan Eickhoff
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Bert Klebl
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Bård H Hoff
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Eirik Sundby
- Department of Materials Science & Engineering, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| |
Collapse
|
16
|
Conte M, De Feo MS, Sidrak MMA, Corica F, Gorica J, Granese GM, Filippi L, De Vincentis G, Frantellizzi V. Imaging of Tauopathies with PET Ligands: State of the Art and Future Outlook. Diagnostics (Basel) 2023; 13:diagnostics13101682. [PMID: 37238166 DOI: 10.3390/diagnostics13101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: Tauopathies are a group of diseases characterized by the deposition of abnormal tau protein. They are distinguished into 3R, 4R, and 3R/4R tauopathies and also include Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). Positron emission tomography (PET) imaging represents a pivotal instrument to guide clinicians. This systematic review aims to summarize the current and novel PET tracers. (2) Methods: Literature research was conducted on Pubmed, Scopus, Medline, Central, and the Web of Science using the query "pet ligands" and "tauopathies". Articles published from January 2018 to 9 February, 2023, were searched. Only studies on the development of novel PET radiotracers for imaging in tauopathies or comparative studies between existing PET tracers were included. (3) Results: A total of 126 articles were found, as follows: 96 were identified from PubMed, 27 from Scopus, one on Central, two on Medline, and zero on the Web of Science. Twenty-four duplicated works were excluded, and 63 articles did not satisfy the inclusion criteria. The remaining 40 articles were included for quality assessment. (4) Conclusions: PET imaging represents a valid instrument capable of helping clinicians in diagnosis, but it is not always perfect in differential diagnosis, even if further investigations on humans for novel promising ligands are needed.
Collapse
Affiliation(s)
- Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Silvia De Feo
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Marko Magdi Abdou Sidrak
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Ferdinando Corica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Joana Gorica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Giorgia Maria Granese
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Luca Filippi
- Department of Nuclear Medicine, Santa Maria Goretti Hospital, 00410 Latina, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
17
|
Al Khatib MHDO, Pinton G, Moro L, Porta C. Benefits and Challenges of Inhibiting EZH2 in Malignant Pleural Mesothelioma. Cancers (Basel) 2023; 15:1537. [PMID: 36900330 PMCID: PMC10000483 DOI: 10.3390/cancers15051537] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/25/2023] [Accepted: 02/26/2023] [Indexed: 03/04/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive thoracic cancer that is mainly associated with prior exposure to asbestos fibers. Despite being a rare cancer, its global rate is increasing and the prognosis remains extremely poor. Over the last two decades, despite the constant research of new therapeutic options, the combination chemotherapy with cisplatin and pemetrexed has remained the only first-line therapy for MPM. The recent approval of immune checkpoint blockade (ICB)-based immunotherapy has opened new promising avenues of research. However, MPM is still a fatal cancer with no effective treatments. Enhancer of zeste homolog 2 (EZH2) is a histone methyl transferase that exerts pro-oncogenic and immunomodulatory activities in a variety of tumors. Accordingly, a growing number of studies indicate that EZH2 is also an oncogenic driver in MPM, but its effects on tumor microenvironments are still largely unexplored. This review describes the state-of-the-art of EZH2 in MPM biology and discusses its potential use both as a diagnostic and therapeutic target. We highlight current gaps of knowledge, the filling of which will likely favor the entry of EZH2 inhibitors within the treatment options for MPM patients.
Collapse
Affiliation(s)
- MHD Ouis Al Khatib
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
- Center for Translational Research on Autoimmune & Allergic Diseases (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| | - Giulia Pinton
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| | - Laura Moro
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| | - Chiara Porta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
- Center for Translational Research on Autoimmune & Allergic Diseases (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| |
Collapse
|
18
|
Huang Y, Wang Z, Gong J, Zhu D, Chen W, Li F, Liang XJ, Liu X. Macrophages as potential targets in gene therapy for cancer treatment. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:89-101. [PMID: 36937317 PMCID: PMC10017190 DOI: 10.37349/etat.2023.00124] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/30/2022] [Indexed: 03/04/2023] Open
Abstract
Macrophages, as ubiquitous and functionally diverse immune cells, play a central role in innate immunity and initiate adaptive immunity. Especially, tumor-associated macrophages (TAMs) are crucial contributors to the tumorigenesis and development of cancer. Thus, macrophages are emerging potential targets for cancer treatment. Among the numerous targeted therapeutic options, gene therapy is one of the most potential therapeutic strategies via directly and specifically regulating biological functions of macrophages at the gene level for cancer treatment. This short review briefly introduces the characteristics of macrophage populations, the functions of TAM in the occurrence, and the progress of cancer. It also summarized some representative examples to highlight the current progress in TAM-targeted gene therapy. The review hopes to provide new insights into macrophage-targeted gene therapy for precision cancer therapy.
Collapse
Affiliation(s)
- Yuanzheng Huang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Zhihui Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Junni Gong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Dandan Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Wang Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Fangzhou Li
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- Nano Science and Technology Institute, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoxuan Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
19
|
Sun W, Mao Y, Xiong Y, Li J. The Clinical Effect of Topical Application of Tranexamic Acid in Arthroscopic Synovectomy of Pigmented Villonodular Synovitis of the Knee. Orthopedics 2023; 46:e179-e185. [PMID: 36779738 DOI: 10.3928/01477447-20230207-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
This study investigated the clinical effect of topical application of tranexamic acid (TXA) in arthroscopic synovectomy of pigmented villonodular synovitis (PVNS) of the knee. Eighty patients who were diagnosed with unilateral knee PVNS underwent arthroscopic synovectomies from January 2017 to January 2021 and were retrospectively investigated in this study. Patients were divided into two groups: group A was the control group, whereas patients in group B received an intra-articular injection of 50 mL of TXA (1 g:100 mL) immediately after the synovectomies. The primary outcome measurement was the drainage volume of the affected knee in each postoperative stage, as well as the hematological parameters. Secondary outcomes included visual analog scale score, knee range of motion, and Lysholm score. Postoperative complications were also recorded. A lower volume of drainage was found in group B than in group A on postoperative day (POD) 1, on POD 2, and in total (POD 1, P=.000; POD 2, P=.000; total, P=.000). A lower visual analog scale pain score in group B was noticed on POD 1 (P=.000) and POD 2 (P=.005). Range of motion (P=.019) and Lysholm score (P=.001) were significantly superior in group B compared with group A on POD 14. Regarding complications, 3 patients in group A and 1 patient in group B developed deep venous thrombosis after surgery. Hematomas were found intra-articularly in 5 patients in group A during follow-up. Intraarticular topical application of TXA was effective in reducing postoperative bleeding and early postoperative pain for patients who underwent arthroscopic synovectomy of PVNS. [Orthopedics. 20XX;XX(X):xx-xx.].
Collapse
|
20
|
Boruah P, Deka N. Interleukin 34 in Disease Progressions: A Comprehensive Review. Crit Rev Immunol 2023; 43:25-43. [PMID: 37943151 DOI: 10.1615/critrevimmunol.2023050326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
IL-34, a cytokine, discovered a decade before and is known to be a colony stimulating factor CSF-1 receptor (CSF-1R) ligand. Along with CSF-1R, it also interacts with syndecan-1 receptors and protein-tyrosine phosphatase (PTP-ζ). Hence, IL-34 takes part in a number of biological activities owing to its involvement in different signaling pathways. This review was done to analyze the recent studies on the functions of IL-34 in progression of diseases. The role of IL-34 under the physiological and pathological settings is studied by reviewing current data. In the last ten years, studies suggested that the IL-34 was involved in the regulation of morbid states such as inflammatory diseases, infections, transplant rejection, autoimmune diseases, neurologic diseases, and cancer. In general, the involvement of IL-34 is observed in many serious health ailments like metabolic diseases, heart diseases, infections and even cancer. As such, IL-34 can be regarded as a therapeutic target, potential biomarker or as a therapeutic tool, which ought to be assessed in future research activities.
Collapse
Affiliation(s)
- Prerona Boruah
- Shanghai Veterinary Research Institute, Shanghai, China; School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, Navi Mumbai, India
| | - Nikhita Deka
- Department of Life Sciences, Dibrugarh University, Assam, India
| |
Collapse
|
21
|
Shojaee P, Mornata F, Deutsch A, Locati M, Hatzikirou H. The impact of tumor associated macrophages on tumor biology under the lens of mathematical modelling: A review. Front Immunol 2022; 13:1050067. [PMID: 36439180 PMCID: PMC9685623 DOI: 10.3389/fimmu.2022.1050067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/18/2022] [Indexed: 09/10/2023] Open
Abstract
In this article, we review the role of mathematical modelling to elucidate the impact of tumor-associated macrophages (TAMs) in tumor progression and therapy design. We first outline the biology of TAMs, and its current application in tumor therapies, and their experimental methods that provide insights into tumor cell-macrophage interactions. We then focus on the mechanistic mathematical models describing the role of macrophages as drug carriers, the impact of macrophage polarized activation on tumor growth, and the role of tumor microenvironment (TME) parameters on the tumor-macrophage interactions. This review aims to identify the synergies between biological and mathematical approaches that allow us to translate knowledge on fundamental TAMs biology in addressing current clinical challenges.
Collapse
Affiliation(s)
- Pejman Shojaee
- Centre for Information Services and High Performance Computing, Technische Universität (TU) Dresden, Dresden, Germany
| | - Federica Mornata
- Leukocyte Biology Lab, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Andreas Deutsch
- Centre for Information Services and High Performance Computing, Technische Universität (TU) Dresden, Dresden, Germany
| | - Massimo Locati
- Leukocyte Biology Lab, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine, Universitàdegli Studi di Milano, Milan, Italy
| | - Haralampos Hatzikirou
- Centre for Information Services and High Performance Computing, Technische Universität (TU) Dresden, Dresden, Germany
- Mathematics Department, Khalifa University, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Centre (HEIC), Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
22
|
Ye F, Wu P, Zhu Y, Huang G, Tao Y, Liao Z, Guan Y. Construction of the prognostic signature of alternative splicing revealed the prognostic predictor and immune microenvironment in head and neck squamous cell carcinoma. Front Genet 2022; 13:989081. [PMID: 36338975 PMCID: PMC9633855 DOI: 10.3389/fgene.2022.989081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/04/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Head and neck squamous cell carcinoma (HNSC) is a prevalent and heterogeneous malignancy with poor prognosis and high mortality rates. There is significant evidence of alternative splicing (AS) contributing to tumor development, suggesting its potential in predicting prognosis and therapeutic efficacy. This study aims to establish an AS-based prognostic signature in HNSC patients. Methods: The expression profiles and clinical information of 486 HNSC patients were downloaded from the TCGA database, and the AS data were downloaded from the TCGA SpliceSeq database. The survival-associated AS events were identified by conducting a Cox regression analysis and utilized to develop a prognostic signature by fitting into a LASSO-regularized Cox regression model. Survival analysis, univariate and multivariate Cox regression analysis, and receiver operating characteristic (ROC) curve analysis were performed to evaluate the signature and an independent cohort was used for validation. The immune cell function and infiltration were analyzed by CIBERSORT and the ssGSEA algorithm. Results: Univariate Cox regression analysis identified 2726 survival-associated AS events from 1714 genes. The correlation network reported DDX39B, PRPF39, and ARGLU1 as key splicing factors (SF) regulating these AS events. Eight survival-associated AS events were selected and validated by LASSO regression to develop a prognostic signature. It was confirmed that this signature could predict HNSC outcomes independent of other variables via multivariate Cox regression analysis. The risk score AUC was more than 0.75 for 3 years, highlighting the signature’s prediction capability. Immune infiltration analysis reported different immune cell distributions between the two risk groups. The immune cell content was higher in the high-risk group than in the low-risk group. The correlation analysis revealed a significant correlation between risk score, immune cell subsets, and immune checkpoint expression. Conclusion: The prognostic signature developed from survival-associated AS events could predict the prognosis of HNSC patients and their clinical response to immunotherapy. However, this signature requires further research and validation in larger cohort studies.
Collapse
Affiliation(s)
- Fan Ye
- Department of Surgery, Division of Otolaryngology, Head and Neck Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Pingan Wu
- Department of Surgery, Division of Otolaryngology, Head and Neck Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yaqiong Zhu
- Department of Otolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guan Huang
- Department of Surgery, Division of Otolaryngology, Head and Neck Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ying Tao
- Department of Surgery, Division of Otolaryngology, Head and Neck Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhencheng Liao
- Department of Surgery, Division of Otolaryngology, Head and Neck Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yafeng Guan
- Department of Surgery, Division of Otolaryngology, Head and Neck Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- *Correspondence: Yafeng Guan,
| |
Collapse
|
23
|
Foss CA, Ordonez AA, Naik R, Das D, Hall A, Wu Y, Dannals RF, Jain SK, Pomper MG, Horti AG. PET/CT imaging of CSF1R in a mouse model of tuberculosis. Eur J Nucl Med Mol Imaging 2022; 49:4088-4096. [PMID: 35713665 PMCID: PMC9922090 DOI: 10.1007/s00259-022-05862-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/03/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE Macrophages represent an essential means of sequestration and immune evasion for Mycobacterium tuberculosis. Pulmonary tuberculosis (TB) is characterized by dense collections of tissue-specific and recruited macrophages, both of which abundantly express CSF1R on their outer surface. 4-Cyano-N-(5-(1-(dimethylglycyl)piperidin-4-yl)-2',3',4',5'-tetrahydro-[1,1'-biphenyl]-2-yl)-1H-imidazole-2-carboxamide (JNJ-28312141) is a reported high affinity, CSF1R-selective antagonist. We report the radiosynthesis of 4-cyano-N-(5-(1-(N-methyl-N-([11C]methyl)glycyl)piperidin-4-yl)-2',3',4',5'-tetrahydro-[1,1'-biphenyl]-2-yl)-1H-imidazole-2-carboxamide ([11C]JNJ-28312141) and non-invasive detection of granulomatous and diffuse lesions in a mouse model of TB using positron emission tomography (PET). METHODS Nor-methyl-JNJ-28312141 precursor was radiolabeled with [11C]iodomethane to produce [11C]JNJ-28312141. PET/CT imaging was performed in the C3HeB/FeJ murine model of chronic pulmonary TB to co-localize radiotracer uptake with granulomatous lesions observed on CT. Additionally, CSF1R, Iba1 fluorescence immunohistochemistry was performed to co-localize CSF1R target with reactive macrophages in infected and healthy mice. RESULTS Radiosynthesis of [11C]JNJ-28312141 averaged a non-decay-corrected yield of 18.7 ± 2.1%, radiochemical purity of 99%, and specific activity averaging 658 ± 141 GBq/µmol at the end-of-synthesis. PET/CT imaging in healthy mice showed hepatobiliary [13.39-25.34% ID/g, percentage of injected dose per gram of tissue (ID/g)] and kidney uptake (12.35% ID/g) at 40-50 min post-injection. Infected mice showed focal pulmonary lesion uptake (5.58-12.49% ID/g), hepatobiliary uptake (15.30-40.50% ID/g), cervical node uptake, and renal uptake (11.66-29.33% ID/g). The ratio of infected lesioned lung/healthy lung uptake is 5.91:1, while the ratio of lesion uptake to adjacent infected radiolucent lung is 2.8:1. Pre-administration of 1 mg/kg of unlabeled JNJ-28312141 with [11C]JNJ-28312141 in infected animals resulted in substantial blockade. Fluorescence microscopy of infected and uninfected whole lung sections exclusively co-localized CSF1R staining with abundant Iba1 + macrophages. Healthy lung exhibited no CSF1R staining and very few Iba1 + macrophages. CONCLUSION [11C]JNJ-28312141 binds specifically to CSF1R + macrophages and delineates granulomatous foci of disease in a murine model of pulmonary TB.
Collapse
Affiliation(s)
- Catherine A Foss
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA.
- Department of Pediatrics, Center for Infection and Inflammation Imaging Research, Baltimore, MD, USA.
| | - Alvaro A Ordonez
- Department of Pediatrics, Center for Infection and Inflammation Imaging Research, Baltimore, MD, USA
| | - Ravi Naik
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Deepankar Das
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew Hall
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Yunkou Wu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Robert F Dannals
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Sanjay K Jain
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
- Department of Pediatrics, Center for Infection and Inflammation Imaging Research, Baltimore, MD, USA
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew G Horti
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
24
|
PD-L1 Status in Tenosynovial Giant Cell Tumors. Medicina (B Aires) 2022; 58:medicina58091270. [PMID: 36143947 PMCID: PMC9501118 DOI: 10.3390/medicina58091270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/16/2022] [Accepted: 09/08/2022] [Indexed: 11/24/2022] Open
Abstract
Background and Objectives: Tenosynovial giant cell tumors (TSGCTs) are benign soft tissue tumors that are divided into localized- and diffuse-type tumors, according to the World Health Organization classification of soft tissue tumours. The diffuse-type TSGCT sometimes behave aggressively and poses treatment challenges especially in patients with neurovascular involvement. Symptomatic patients who are not good candidates for surgery due to high morbidity risk may benefit from medical therapy. Objectives: Drugs that target programmed death ligand 1 (PD-L1) are among a new generation of medical therapy options, which, recently, have been explored and have displayed promising results in various cancer types; therefore, we aimed to investigate the PD-L1 status of TSGCTs as a possible therapeutic target. Materials and Methods: We assessed the PD-L1 status of 20 patients (15 men and 5 women, median age = 39 years) that had been diagnosed with TSGCTs in a single institution, between 2018 and 2020. The patients had localized- (n = 7) and diffuse-type (n = 13) TSGCTs. Formalin-fixed paraffin-embedded (FFPE) blocks were retrospectively retrieved from the pathology department. An immunohistochemical analysis was performed in sections of 3 micron thickness from these blocks. Results: Seventy-five percent of our patients with TSGCTs were immunopositive to PD-L1 staining. Conclusions: Taking into consideration the high positivity rate of PD-L1 staining in TSGCTs, PD-L1 blockage may be used as a valuable medical treatment for TSGCTs; however, further studies are needed.
Collapse
|
25
|
Zhan Y, Tong H, He S, Zhu H, Guo H, Sun H, Liu M. A New 7-azaindole Structure Analog: Molecular Docking, Synthesis and Preliminary Biological Activity in vitro for Anticancer. Chem Biodivers 2022; 19:e202200692. [PMID: 36082623 DOI: 10.1002/cbdv.202200692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 09/08/2022] [Indexed: 11/11/2022]
Abstract
In this work, a series of 7-azaindole analogs was designed by the bioisosteric principle based on pharmacodynamic parent nucleus. Moreover, 5-Chloro-3-{[2-({[6-(trifluoromethyl) pyridin-3-yl]methyl}amino)pyrimidin-5-yl]methyl}-1H-pyrrolo[2,3-b]pyridine (compound P1 ) with the strongest interaction with colony-stimulating factor 1 receptor (CSF-1R) was screened by molecular docking. Compound P1 was successfully prepared by the six-step reaction with HPLC purity of 99.26% and characterized by 1 H NMR and ESI-MS spectra. In vitro bioactivity study showed that compound P1 appeared the cytotoxicity to MCF-7 and A549 cells, especially to HOS cells (IC 50 = 88.79 ± 8.07 nM), while it had lower toxicity to normal L929 cells (IC 50 = 140.49 ± 8.03 μM). In addition, compound P1 could induce HOS cell death by apoptosis and blocking the G0/G1 phase at nanomolar concentrations. The obtained results indicated that compound P1 might be a promising candidate compound for anticancer drug.
Collapse
Affiliation(s)
- Yifeng Zhan
- Hubei University of Technology, Pharmaceutical Engineering, Nanli Road 28, 430068, Wuhan, CHINA
| | - Hang Tong
- Hubei University of Technology, Pharmaceutical Engineering, Nanli Road 28, 430068, Wuhan, CHINA
| | - Shibo He
- Hubei University of Technology, Pharmaceutical Engineering, Nanli Road 28, 430068, Wuhan, CHINA
| | - Hongda Zhu
- Hubei University of Technology, Pharmaceutical Engineering, Nanli Road 28, 430068, Wuhan, CHINA
| | - Huiling Guo
- Hubei University of Technology, Pharmaceutical Engineering, Nanli Road 28, 430068, Wuhan, CHINA
| | - Hongmei Sun
- Hubei University of Technology, Pharmaceutical Engineering, Nanli Road 28, 430068, Wuhan, CHINA
| | - Mingxing Liu
- Hubei University of Technology, Pharmaceutical Engineering, Nanli Road 28, 430068, Wuhan, CHINA
| |
Collapse
|
26
|
Xiong K, Qi M, Stoeger T, Zhang J, Chen S. The role of tumor-associated macrophages and soluble mediators in pulmonary metastatic melanoma. Front Immunol 2022; 13:1000927. [PMID: 36131942 PMCID: PMC9483911 DOI: 10.3389/fimmu.2022.1000927] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022] Open
Abstract
Skin malignant melanoma is a highly aggressive skin tumor, which is also a major cause of skin cancer-related mortality. It can spread from a relatively small primary tumor and metastasize to multiple locations, including lymph nodes, lungs, liver, bone, and brain. What’s more metastatic melanoma is the main cause of its high mortality. Among all organs, the lung is one of the most common distant metastatic sites of melanoma, and the mortality rate of melanoma lung metastasis is also very high. Elucidating the mechanisms involved in the pulmonary metastasis of cutaneous melanoma will not only help to provide possible explanations for its etiology and progression but may also help to provide potential new therapeutic targets for its treatment. Increasing evidence suggests that tumor-associated macrophages (TAMs) play an important regulatory role in the migration and metastasis of various malignant tumors. Tumor-targeted therapy, targeting tumor-associated macrophages is thus attracting attention, particularly for advanced tumors and metastatic tumors. However, the relevant role of tumor-associated macrophages in cutaneous melanoma lung metastasis is still unclear. This review will present an overview of the origin, classification, polarization, recruitment, regulation and targeting treatment of tumor-associated macrophages, as well as the soluble mediators involved in these processes and a summary of their possible role in lung metastasis from cutaneous malignant melanoma. This review particularly aims to provide insight into mechanisms and potential therapeutic targets to readers, interested in pulmonary metastasis melanoma.
Collapse
Affiliation(s)
- Kaifen Xiong
- The Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People’s Hospital (The Second Clinical Medical College), Jinan University, Guangdong, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Department of Dermatology, Xiangya Hospital of Central South University, Changsha, China
| | - Min Qi
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Tobias Stoeger
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jianglin Zhang
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Department of Dermatology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Guangdong, China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, China
- *Correspondence: Jianglin Zhang, ; Shanze Chen,
| | - Shanze Chen
- The Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People’s Hospital (The Second Clinical Medical College), Jinan University, Guangdong, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Jianglin Zhang, ; Shanze Chen,
| |
Collapse
|
27
|
Immune Cell Infiltration Characteristics of Pigmented Villous Nodular Synovitis and Prediction of Potential Diagnostic Markers Based on Bioinformatics. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8708692. [PMID: 35711523 PMCID: PMC9197622 DOI: 10.1155/2022/8708692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/21/2022] [Indexed: 11/21/2022]
Abstract
Background Pigmented villous nodular synovitis (PVNS) is a tumor-like proliferative disease characterized by impairment of daily activities, decreased quality of life, and a high recurrence rate. However, the specific pathological mechanisms are still ill-defined and controversial. The purpose of this study was to define potential diagnostic markers and evaluate immune cell infiltration in the pathogenesis of PVNS. Method The expression profile of GSE3698 was reanalyzed in the Gene Expression Omnibus (GEO) database. First, differentially expressed genes (DEGs) were identified using the R package “limma” and analyzed by Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. Next, the DEGs were imported into the STRING database and Cytoscape to construct a protein–protein interaction (PPI) network. Then, cytoHubba and ROC curve analyses were used to determine potential diagnostic biomarkers of PVNS. Finally, we used CIBERSORT to estimate the proportions of 22 immune cell subtypes in PVNS and analyzed the correlation between diagnostic markers and infiltrating immune cells. Result We found 139 DEGs (including 93 upregulated genes and 46 downregulated genes). TYROBP, FCER1G, LAPTM5, and HLA-DPB1 were identified as potential diagnostic biomarkers of PVNS. Immune cell infiltration analysis indicated that neutrophils and M2 macrophages might be associated with the genesis and progression of PVNS. Furthermore, our correlation analysis of diagnostic markers and infiltrating immune cells found that TYROBP, FCER1G, LAPTM5, and HLA-DPB1 were positively correlated with M2 macrophage infiltration and that neutrophils, TYROBP, FCER1G, and LAPTM5 were negatively correlated with plasma cell infiltration. Conclusions We identified TYROBP, FCER1G, LAPTM5, and HLA-DPB1 as potential diagnostic markers for PVNS and concluded that immune cell infiltration plays an important role in the genesis and progression of PVNS.
Collapse
|
28
|
Rodriguez-Perdigon M, Jimaja S, Haen L, Bruns N, Rothen-Rutishauser B, Rüegg C. Polymersomes-mediated Delivery of CSF1R Inhibitor to Tumor Associated Macrophages Promotes M2 to M1-like Macrophage Repolarization. Macromol Biosci 2022; 22:e2200168. [PMID: 35624036 DOI: 10.1002/mabi.202200168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Indexed: 11/07/2022]
Abstract
The crosstalk between cancer cells and tumor associated macrophages (TAMs) within the tumor environment modulates tumor progression at all stages of cancer disease. TAMs are predominantly M2-like polarized macrophages with tumor-promoting activities. Nonetheless, they can be repolarized to tumoricidal M1-like macrophages through macrophage colony stimulating factor 1 receptor inhibition (CSF1Ri). CSF1Ri is being explored as multifaced therapeutic approach to suppress TAMs tumor-promoting functions and reduce cancer cell aggressiveness and viability. However, treatment with CSF1Ri results in significant TAMs death, thereby extinguishing the possibility of generating tumoricidal M1-like macrophages. Immunotherapy has improved overall patient's survival in some cancer types, but also caused frequent off-target toxicity. Approaches to balance efficacy versus toxicity are needed. Herein, a CSF1Ri loaded polymersomes (PM) based delivery platform is developed to promote M2-like macrophage repolarization. When testing in vitro on primary human monocyte-derived macrophages (MDMs), CSF1Ri loaded PM are preferentially taken up by M2-like macrophages and enhance M2 to M1-like macrophage repolarization while minimizing cytotoxicity in comparison to the free drug. When testing in a MDMs-MDA-MB-231 breast cancer cell co-culture model, CSF1Ri loaded PM further retain their M2 to M1-like macrophages polarization capacity. This CSF1Ri loaded PM-based platform system represents a promising tool for macrophage-based immunotherapy approaches. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Manuel Rodriguez-Perdigon
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, Fribourg, 1700, Switzerland
| | - Sètuhn Jimaja
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, 1700, Switzerland
| | - Laetitia Haen
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, 1700, Switzerland
| | - Nico Bruns
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, 1700, Switzerland.,Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow, UK
| | | | - Curzio Rüegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, Fribourg, 1700, Switzerland
| |
Collapse
|
29
|
Haddad R, Zlotnik O, Goshen-Lago T, Levi M, Brook E, Brenner B, Kundel Y, Ben-Aharon I, Kashtan H. Tumor Lymphocyte Infiltration Is Correlated with a Favorable Tumor Regression Grade after Neoadjuvant Treatment for Esophageal Adenocarcinoma. J Pers Med 2022; 12:jpm12040627. [PMID: 35455743 PMCID: PMC9029859 DOI: 10.3390/jpm12040627] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/23/2022] [Accepted: 04/11/2022] [Indexed: 01/01/2023] Open
Abstract
(1) Background: We aimed to explore the association between neoadjuvant treatment, tumor-infiltrating immune lymphocyte (TIL), and tumor-associated macrophage (TAM) and survival in patients with esophageal adenocarcinoma. (2) Methods: Patients who underwent esophagectomy were divided into three groups according to their treatment modality and tumor regression grade (TRG): (i) surgery-only group (SG), (ii) good responders (GR) group (TRG 0−1), and (iii) bad responders (BR) group (TRG 2−3). We then carried out statistical correlations of the immunofluorescence analysis of the immune infiltrate in the esophageal surgical specimens with several clinical and pathological parameters. In addition, we analyzed The Cancer Genomic Atlas (TCGA) dataset for differences in TILs, TAMs, and protein expression in immune pathways. (3) Results: Forty-three patients (SG—15, GR—13, and BR—13) were evaluated. The highest enrichment of CD3+ (p < 0.001), CD8+ (p = 0.001) and CD4+ (p = 0.009) was observed in the stroma of GR patients. On multivariate analysis, only CD8+ T cell and signet-ring features were independent prognostic factors for overall survival. In TCGA analysis, we identified overexpression of TAM and colony-stimulating factor 1 receptor (CSF-1R). (4) Conclusions: High enrichment of lymphocyte subpopulations in the microenvironment of esophageal adenocarcinoma is associated with a favorable response to neoadjuvant treatment and an improved patient outcome.
Collapse
Affiliation(s)
- Riad Haddad
- Department of Surgery, Carmel Medical Center, Haifa 3436212, Israel; (R.H.); (H.K.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 3525433, Israel
| | - Oran Zlotnik
- Department of Surgery, Rabin Medical Center, Petach Tikva 4941492, Israel;
| | - Tal Goshen-Lago
- Division of Oncology, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Mattan Levi
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 6329302, Israel; (M.L.); (B.B.); (Y.K.)
| | - Elena Brook
- Department of Pathology, Rabin Medical Center, Petach Tikva 4941492, Israel;
| | - Baruch Brenner
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 6329302, Israel; (M.L.); (B.B.); (Y.K.)
- Institute of Oncology, Davidoff Center, Rabin Medical Center, Petach Tikva 4941492, Israel
| | - Yulia Kundel
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 6329302, Israel; (M.L.); (B.B.); (Y.K.)
- Institute of Oncology, Davidoff Center, Rabin Medical Center, Petach Tikva 4941492, Israel
| | - Irit Ben-Aharon
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 3525433, Israel
- Division of Oncology, Rambam Health Care Campus, Haifa 3109601, Israel;
- Correspondence:
| | - Hanoch Kashtan
- Department of Surgery, Carmel Medical Center, Haifa 3436212, Israel; (R.H.); (H.K.)
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 6329302, Israel; (M.L.); (B.B.); (Y.K.)
| |
Collapse
|
30
|
Song Y, Huang Y, Zhou F, Ding J, Zhou W. Macrophage-targeted nanomedicine for chronic diseases immunotherapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.08.090] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
31
|
Hourani T, Holden JA, Li W, Lenzo JC, Hadjigol S, O’Brien-Simpson NM. Tumor Associated Macrophages: Origin, Recruitment, Phenotypic Diversity, and Targeting. Front Oncol 2021; 11:788365. [PMID: 34988021 PMCID: PMC8722774 DOI: 10.3389/fonc.2021.788365] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022] Open
Abstract
The tumor microenvironment (TME) is known to have a strong influence on tumorigenesis, with various components being involved in tumor suppression and tumor growth. A protumorigenic TME is characterized by an increased infiltration of tumor associated macrophages (TAMs), where their presence is strongly associated with tumor progression, therapy resistance, and poor survival rates. This association between the increased TAMs and poor therapeutic outcomes are stemming an increasing interest in investigating TAMs as a potential therapeutic target in cancer treatment. Prominent mechanisms in targeting TAMs include: blocking recruitment, stimulating repolarization, and depletion methods. For enhancing targeting specificity multiple nanomaterials are currently being explored for the precise delivery of chemotherapeutic cargo, including the conjugation with TAM-targeting peptides. In this paper, we provide a focused literature review of macrophage biology in relation to their role in tumorigenesis. First, we discuss the origin, recruitment mechanisms, and phenotypic diversity of TAMs based on recent investigations in the literature. Then the paper provides a detailed review on the current methods of targeting TAMs, including the use of nanomaterials as novel cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Neil M. O’Brien-Simpson
- Antimicrobial, Cancer Therapeutics and Vaccines (ACTV) Research Group, Melbourne Dental School, Centre for Oral Health Research, Royal Dental Hospital, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
32
|
Is intra-articular infliximab therapy a good alternative to radionuclide synovectomy for a patient with refractory pigmented villonodular synovitis? Reumatologia 2021; 59:340-345. [PMID: 34819709 PMCID: PMC8609374 DOI: 10.5114/reum.2021.110601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/01/2021] [Indexed: 12/02/2022] Open
Abstract
Pigmented villonodular synovitis (PVNS) is a rare disease that has clinical and histopathological characteristics of both benign proliferative disorder and a chronic inflammatory process of the synovial tissue. The primary mode of treatment is surgery followed by an adjuvant radiotherapy; however, the risk of recurrence is a significant (40–70%). Several publications suggest that the TNF-α inhibitors might be a treatment option. We present a case of a 29-year-old female diagnosed with PVNS of the knee joint, refractory to surgery and 3 radionuclide synovectomies. Because the possibilities of conventional therapy were exhausted, treatment with an intra-articular anti-TNF-α monoclonal antibody (infliximab) was performed. Despite a high safety profile and a good tolerance of that therapy we did not observe significant clinical and radiological improvement. To assess the effectiveness of intra-articular TNF-α inhibitors as an adjuvant treatment in PVNS, prospective studies are needed.
Collapse
|
33
|
Conte E. Targeting monocytes/macrophages in fibrosis and cancer diseases: Therapeutic approaches. Pharmacol Ther 2021; 234:108031. [PMID: 34774879 DOI: 10.1016/j.pharmthera.2021.108031] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/19/2021] [Accepted: 11/02/2021] [Indexed: 02/08/2023]
Abstract
Over almost 140 years since their identification, the knowledge about macrophages has unbelievably evolved. The 'big eaters' from being thought of as simple phagocytic cells have been recognized as master regulators in immunity, homeostasis, healing/repair and organ development. Long considered to originate exclusively from bone marrow-derived circulating monocytes, macrophages have been also demonstrated to be the first immune cells colonizing tissues in the developing embryo and persisting in adult life by self-renewal, as long-lived tissue resident macrophages. Therefore, heterogeneous populations of macrophages with different ontogeny and functions co-exist in tissues. Macrophages act as sentinels of homeostasis and are intrinsically programmed to lead the wound healing and repair processes that occur after injury. However, in certain pathological circumstances macrophages get dysfunctional, and impaired or aberrant macrophage activities become key features of diseases. For instance, in both fibrosis and cancer, that have been defined 'wounds that do not heal', dysfunctional monocyte-derived macrophages overall play a key detrimental role. On the other hand, due to their plasticity these cells can be 're-educated' and exert anti-fibrotic and anti-cancer functions. Therefore macrophages represent an important therapeutic target in both fibrosis and cancer diseases. The current review will illustrate new insights into the role of monocytes/macrophages in these devastating diseases and summarize new therapeutic strategies and applications of macrophage-targeted drug development in their clinical setting.
Collapse
|
34
|
Wang S, Yang X, Xie W, Fu S, Chen Q, Li Z, Zhang Z, Sun T, Gong B, Ma M. LncRNA GAPLINC Promotes Renal Cell Cancer Tumorigenesis by Targeting the miR-135b-5p/CSF1 Axis. Front Oncol 2021; 11:718532. [PMID: 34722262 PMCID: PMC8551964 DOI: 10.3389/fonc.2021.718532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Background Long noncoding RNAs (lncRNAs) are closely related to the occurrence and development of cancer. Gastric adenocarcinoma-associated, positive CD44 regulator, long intergenic noncoding RNA (GAPLINC) is a recently identified lncRNA that can actively participate in the tumorigenesis of various cancers. Here, we investigated the functional roles and mechanism of GAPLINC in renal cell carcinoma (RCC) development. Methods Differentially expressed lncRNAs between RCC tissues and normal kidney tissues were detected by using a microarray technique. RNA sequencing was applied to explore the mRNA expression profile changes after GAPLINC silencing. After gain- and loss-of-function approaches were implemented, the effect of GAPLINC on RCC in vitro and in vivo was assessed by cell proliferation and migration assays. Moreover, rescue experiments and luciferase reporter assays were used to study the interactions between GAPLINC, miR-135b-5p and CSF1. Results GAPLINC was significantly upregulated in RCC tissues and cell lines and was associated with a poor prognosis in RCC patients. Knockdown of GAPLINC repressed RCC growth in vitro and in vivo, while overexpression of GAPLINC exhibited the opposite effect. Mechanistically, we found that GAPLINC upregulates oncogene CSF1 expression by acting as a sponge of miR-135b-5p. Conclusion Taken together, our results suggest that GAPLINC is a novel prognostic marker and molecular therapeutic target for RCC.
Collapse
Affiliation(s)
- Siyuan Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaorong Yang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenjie Xie
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shengqiang Fu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qiang Chen
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhilong Li
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhicheng Zhang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Sun
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Binbin Gong
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ming Ma
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
35
|
Chamseddine AN, Assi T, Mir O, Chouaib S. Modulating tumor-associated macrophages to enhance the efficacy of immune checkpoint inhibitors: A TAM-pting approach. Pharmacol Ther 2021; 231:107986. [PMID: 34481812 DOI: 10.1016/j.pharmthera.2021.107986] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAM) plasticity and diversity are both essential hallmarks of the monocyte-macrophage lineage and the tumor-derived inflammation. TAM exemplify the perfect adaptable cell with dynamic phenotypic modifications that reflect changes in their functional polarization status. Under several tumor microenvironment (TME)-related cues, TAM shift their polarization, hence promoting or halting cancer progression. Immune checkpoint inhibitors (ICI) displayed unprecedented clinical responses in various refractory cancers; but only approximately a third of patients experienced durable responses. It is, therefore, crucial to enhance the response rate of immunotherapy. Several mechanisms of resistance to ICI have been elucidated including TAM role with its essential immunosuppressive functions that reduce both anti-tumor immunity and the subsequent ICI efficacy. In the past few years, thorough research has led to a better understanding of TAM biology and innovative approaches can now be adapted through targeting macrophages' recruitment axis as well as TAM activation and polarization status within the TME. Some of these therapeutic strategies are currently being evaluated in several clinical trials in association with ICI agents. This combination between TAM modulation and ICI allows targeting TAM intrinsic immunosuppressive functions and tumor-promoting factors as well as overcoming ICI resistance. Hence, such strategies, with a better understanding of the mechanisms driving TAM modulation, may have the potential to optimize ICI efficacy.
Collapse
Affiliation(s)
- Ali N Chamseddine
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Biostatistics and Epidemiology, CESP INSERM U1018, OncoStat, Gustave Roussy, F-94805, Villejuif, France.
| | - Tarek Assi
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France
| | - Olivier Mir
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Pharmacology, Gustave Roussy, F-94805, Villejuif, France; Department of Ambulatory Care, Gustave Roussy, F-94805, Villejuif, France
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, F-94805, Villejuif, France
| |
Collapse
|
36
|
van der Wildt B, Miao Z, Reyes ST, Park JH, Klockow JL, Zhao N, Romero A, Guo SG, Shen B, Windhorst AD, Chin FT. BLZ945 derivatives for PET imaging of colony stimulating factor-1 receptors in the brain. Nucl Med Biol 2021; 100-101:44-51. [PMID: 34174546 DOI: 10.1016/j.nucmedbio.2021.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/04/2021] [Accepted: 06/17/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND The kinase colony stimulating factor-1 receptor (CSF-1R) has recently been identified as a novel therapeutic target for decreasing tumor associated macrophages and microglia load in cancer treatment. In glioblastoma multiforme (GBM), a high-grade cancer in the brain with extremely poor prognosis, macrophages and microglia can make up to 50% of the total tumor mass. Currently, no non-invasive methods are available for measuring CSF-1R expression in vivo. The aim of this work is to develop a PET tracer for imaging of CSF-1R receptor expression in the brain for future GBM patient selection and treatment monitoring. METHODS BLZ945 and a derivative that potentially allows for fluorine-18 labeling were synthesized and evaluated in vitro to determine their affinity towards CSF-1R. BLZ945 was radiolabeled with carbon-11 by N-methylation of des-methyl-BLZ945 using [11C]CH3I. Following administration to healthy mice, metabolic stability of [11C]BLZ945 in blood and brain and activity distribution were determined ex vivo. PET scanning was performed at baseline, efflux transporter blocking, and CSF-1R blocking conditions. Finally, [11C]BLZ945 binding was evaluated in vitro by autoradiography on mouse brain sections. RESULTS BLZ945 was the most potent compound in our series with an IC50 value of 6.9 ± 1.4 nM. BLZ945 was radiolabeled with carbon-11 in 20.7 ± 1.1% decay corrected radiochemical yield in a 60 min synthesis procedure with a radiochemical purity of >95% and a molar activity of 153 ± 34 GBq·μmol-1. Ex vivo biodistribution showed moderate brain uptake and slow wash-out, in addition to slow blood clearance. The stability of BLZ945 in blood plasma and brain was >99% at 60 min post injection. PET scanning demonstrated BLZ945 to be a substrate for efflux transporters. High brain uptake was observed, which was shown to be mostly non-specific. In accordance, in vitro autoradiography on brain sections revealed high non-specific binding. CONCLUSIONS [11C]BLZ945, a CSF-1R PET tracer, was synthesized in high yield and purity. The tracer has high potency for the target, however, future studies are warranted to address non-specific binding and tracer efflux before BLZ945 or derivatives could be translated into humans for brain imaging.
Collapse
Affiliation(s)
- Berend van der Wildt
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA; Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, de Boelelaan 1117, Amsterdam, Netherlands
| | - Zheng Miao
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Samantha T Reyes
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Jun H Park
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Jessica L Klockow
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Ning Zhao
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Alex Romero
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Scarlett G Guo
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Bin Shen
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Albert D Windhorst
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, de Boelelaan 1117, Amsterdam, Netherlands
| | - Frederick T Chin
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA.
| |
Collapse
|
37
|
van der Wildt B, Nezam M, Kooijman EJ, Reyes ST, Shen B, Windhorst AD, Chin FT. Evaluation of carbon-11 labeled 5-(1-methyl-1H-pyrazol-4-yl)-N-(2-methyl-5-(3-(trifluoromethyl)benzamido)phenyl)nicotinamide as PET tracer for imaging of CSF-1R expression in the brain. Bioorg Med Chem 2021; 42:116245. [DOI: 10.1016/j.bmc.2021.116245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/14/2022]
|
38
|
Fujiwara T, Yakoub MA, Chandler A, Christ AB, Yang G, Ouerfelli O, Rajasekhar VK, Yoshida A, Kondo H, Hata T, Tazawa H, Dogan Y, Moore MAS, Fujiwara T, Ozaki T, Purdue E, Healey JH. CSF1/CSF1R Signaling Inhibitor Pexidartinib (PLX3397) Reprograms Tumor-Associated Macrophages and Stimulates T-cell Infiltration in the Sarcoma Microenvironment. Mol Cancer Ther 2021; 20:1388-1399. [PMID: 34088832 DOI: 10.1158/1535-7163.mct-20-0591] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 03/10/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022]
Abstract
Colony-stimulating factor 1 (CSF1) is a primary regulator of the survival, proliferation, and differentiation of monocyte/macrophage that sustains the protumorigenic functions of tumor-associated macrophages (TAMs). Considering current advances in understanding the role of the inflammatory tumor microenvironment, targeting the components of the sarcoma microenvironment, such as TAMs, is a viable strategy. Here, we investigated the effect of PLX3397 (pexidartinib) as a potent inhibitor of the CSF1 receptor (CSF1R). PLX3397 was recently approved by the Food and Drug Administration (FDA) to treat tenosynovial giant cell tumor and reprogram TAMs whose infiltration correlates with unfavorable prognosis of sarcomas. First, we confirmed by cytokine arrays of tumor-conditioned media (TCM) that cytokines including CSF1 are secreted from LM8 osteosarcoma cells and NFSa fibrosarcoma cells. The TCM, like CSF1, stimulated ERK1/2 phosphorylation in bone marrow-derived macrophages (BMDMs), polarized BMDMs toward an M2 (TAM-like) phenotype, and strikingly promoted BMDM chemotaxis. In vitro administration of PLX3397 suppressed pERK1/2 stimulation by CSF1 or TCM, and reduced M2 polarization, survival, and chemotaxis in BMDMs. Systemic administration of PLX3397 to the osteosarcoma orthotopic xenograft model significantly suppressed the primary tumor growth and lung metastasis, and thus improved metastasis-free survival. PLX3397 treatment concurrently depleted TAMs and FOXP3+ regulatory T cells and, surprisingly, enhanced infiltration of CD8+ T cells into the microenvironments of both primary and metastatic osteosarcoma sites. Our preclinical results show that PLX3397 has strong macrophage- and T-cell-modulating effects that may translate into cancer immunotherapy for bone and soft-tissue sarcomas.
Collapse
Affiliation(s)
- Tomohiro Fujiwara
- Department of Surgery, Orthopaedic Service, Memorial Sloan Kettering Cancer Center, New York, New York.,Hospital for Special Surgery, New York, New York.,Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Mohamed A Yakoub
- Department of Surgery, Orthopaedic Service, Memorial Sloan Kettering Cancer Center, New York, New York.,Hospital for Special Surgery, New York, New York
| | - Andrew Chandler
- Department of Surgery, Orthopaedic Service, Memorial Sloan Kettering Cancer Center, New York, New York.,Hospital for Special Surgery, New York, New York
| | - Alexander B Christ
- Department of Surgery, Orthopaedic Service, Memorial Sloan Kettering Cancer Center, New York, New York.,Hospital for Special Surgery, New York, New York
| | - Guangli Yang
- Organic Synthesis Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ouathek Ouerfelli
- Organic Synthesis Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vinagolu K Rajasekhar
- Department of Surgery, Orthopaedic Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Aki Yoshida
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroya Kondo
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiaki Hata
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yildirim Dogan
- Cell Biology, Memorial Sloan Kettering Cancer Center, New York, New York.,AVROBIO Inc., One Kendall Square, Cambridge, Massachusetts
| | - Malcolm A S Moore
- Cell Biology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Ed Purdue
- Hospital for Special Surgery, New York, New York
| | - John H Healey
- Department of Surgery, Orthopaedic Service, Memorial Sloan Kettering Cancer Center, New York, New York. .,Hospital for Special Surgery, New York, New York
| |
Collapse
|
39
|
Baseline Frequency of Inflammatory Cxcl9-Expressing Tumor-Associated Macrophages Predicts Response to Avelumab Treatment. Cell Rep 2021; 32:107873. [PMID: 32640238 DOI: 10.1016/j.celrep.2020.107873] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/30/2020] [Accepted: 06/15/2020] [Indexed: 12/25/2022] Open
Abstract
The tumor microenvironment is rich with immune-suppressive macrophages that are associated with cancer progression and resistance to immune checkpoint therapy. Using pre-treatment tumor biopsies complemented with single-cell RNA sequencing (RNA-seq), we characterize intratumoral immune heterogeneity to unveil potential mechanisms of resistance to avelumab (anti-PD-L1). We identify a proinflammatory F480+MHCII+Ly6Clo macrophage population that is associated with response rather than resistance to avelumab. These macrophages are the primary source of the interferon-inducible chemokine Cxcl9, which facilitates the recruitment of protective Cxcr3+ T cells. Consequently, the efficacy of avelumab in mouse tumor models is dependent on Cxcr3 and Cxcl9, and baseline levels of Cxcl9 in patients treated with avelumab are associated with clinical response and overall survival. These data suggest that, within the broadly immune-suppressive macrophage compartment, a pro-inflammatory population exists that promotes responsiveness to PD-L1 blockade.
Collapse
|
40
|
Mola S, Pinton G, Erreni M, Corazzari M, De Andrea M, Grolla AA, Martini V, Moro L, Porta C. Inhibition of the Histone Methyltransferase EZH2 Enhances Protumor Monocyte Recruitment in Human Mesothelioma Spheroids. Int J Mol Sci 2021; 22:ijms22094391. [PMID: 33922336 PMCID: PMC8122808 DOI: 10.3390/ijms22094391] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a highly aggressive cancer with a long latency period and dismal prognosis. Recently, tazemetostat (EPZ-6438), an inhibitor of the histone methyltransferase EZH2, has entered clinical trials due to the antiproliferative effects reported on MPM cells. However, the direct and indirect effects of epigenetic reprogramming on the tumor microenvironment are hitherto unexplored. To investigate the impact of tumor-associated macrophages (TAMs) on MPM cell responsiveness to tazemetostat, we developed a three-dimensional MPM spheroid model that recapitulates in vitro, both monocytes’ recruitment in tumors and their functional differentiation toward a TAM-like phenotype (Mo-TAMs). Along with an increased expression of genes for monocyte chemoattractants, inhibitory immune checkpoints, immunosuppressive and M2-like molecules, Mo-TAMs promote tumor cell proliferation and spreading. Prolonged treatment of MPM spheroids with tazemetostat enhances both the recruitment of Mo-TAMs and the expression of their protumor phenotype. Therefore, Mo-TAMs profoundly suppress the antiproliferative effects due to EZH2 inhibition in MPM cells. Overall, our findings indicate that TAMs are a driving force for MPM growth, progression, and resistance to tazemetostat; therefore, strategies of TAM depletion might be evaluated to improve the therapeutic efficacy of pharmacological inhibition of EZH2.
Collapse
Affiliation(s)
- Silvia Mola
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (S.M.); (G.P.); (A.A.G.); (L.M.)
- Center for Translational Research on Autoimmune & Allergic Diseases (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (M.C.); (M.D.A.); (V.M.)
| | - Giulia Pinton
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (S.M.); (G.P.); (A.A.G.); (L.M.)
| | - Marco Erreni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy;
| | - Marco Corazzari
- Center for Translational Research on Autoimmune & Allergic Diseases (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (M.C.); (M.D.A.); (V.M.)
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Marco De Andrea
- Center for Translational Research on Autoimmune & Allergic Diseases (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (M.C.); (M.D.A.); (V.M.)
- Department of Public Health and Pediatric Sciences, Medical School, University of Turin, 10126 Turin, Italy
| | - Ambra A. Grolla
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (S.M.); (G.P.); (A.A.G.); (L.M.)
| | - Veronica Martini
- Center for Translational Research on Autoimmune & Allergic Diseases (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (M.C.); (M.D.A.); (V.M.)
- Department of Translational Medicine (DIMET), University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy
| | - Laura Moro
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (S.M.); (G.P.); (A.A.G.); (L.M.)
| | - Chiara Porta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (S.M.); (G.P.); (A.A.G.); (L.M.)
- Center for Translational Research on Autoimmune & Allergic Diseases (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (M.C.); (M.D.A.); (V.M.)
- Correspondence: ; Tel.: +39-0321-375883; Fax: +39-0321-375821
| |
Collapse
|
41
|
Kinoshita M, Ogawa Y, Honobe A, Tomita O, Oishi N, Shimada S, Kondo T, Kawamura T. Expression of CD1 molecules and colony-stimulating factor 1 receptor in indeterminate cell histiocytosis. J Dermatol 2021; 48:1086-1089. [PMID: 33774864 DOI: 10.1111/1346-8138.15857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 02/17/2021] [Accepted: 03/04/2021] [Indexed: 01/15/2023]
Abstract
Indeterminate cell histiocytosis (ICH) and Langerhans cell histiocytosis (LCH) are rare histiocyte proliferating disorders with unknown etiologies. However, both tumor cells immunophenotypically share some features of Langerhans cells (LC), thereby expressing CD1a. Recent transcriptome analysis revealed that circulating CD1c+ myeloid dendritic cells are the potential precursor of LCH tumor cells. LC express CD1a as well as CD1c, but not CD1b. We discovered that both tumor cells express CD1c, but not CD1b, similar to LC. Moreover, like LC, both tumor cells express colony-stimulating factor 1 receptor (CSF-1R). Considering the crucial role of the interleukin (IL)-34/CSF-1R axis for the development and survival of LC, CSF-1R on both tumor cells might facilitate their survival and proliferation in situ. These data provide additional evidence to support the fact that ICH and LCH share immunophenotypical features with LC. In addition, we hypothesized that tumor cells in ICH and LCH survive and proliferate through IL-34-mediated CSF-1R signaling.
Collapse
Affiliation(s)
- Manao Kinoshita
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Youichi Ogawa
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Akiko Honobe
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Ozumi Tomita
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Naoki Oishi
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Shinji Shimada
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Tetsuo Kondo
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Tatsuyoshi Kawamura
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
42
|
Mo H, Hao Y, Lv Y, Chen Z, Shen J, Zhou S, Yin M. Overexpression of macrophage-colony stimulating factor-1 receptor as a prognostic factor for survival in cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e25218. [PMID: 33761709 PMCID: PMC9282102 DOI: 10.1097/md.0000000000025218] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 02/23/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The relation between the expression of macrophage-colony stimulating factor-1 receptor (CSF-1R) and prognosis of cancer patients has been evaluated in multiple studies, but the results remain controversial. We, therefore, performed a meta-analysis and systematic review to figure out the role of CSF-1R in the prognosis of patients with cancer. METHODS Several databases were searched, including Web of Science, PubMed, and EMBASE. All human studies were published as full text. The Newcastle-Ottawa risk of bias scale was applied to evaluate the research. We extracted hazard ratios (HRs) with 95% confidence interval (95% CI) which assessed progression-free survival (PFS) and overall survival (OS) in order to assess the impacts of CSF-1R on the prognosis of cancer patients. RESULTS A total of 12 citations were identified, with studies including 2260 patients in different cancer types that met the eligibility criteria. It was suggested in a pooled analysis that the over-expression of CSF-1R was significantly related to worse PFS (HR: 1.68; P < .001, 1.25-2.10, 95% CI) and also poorer OS (HR=1.28; P < .001, 1.03-1.54, 95% CI). Analysis in subgroups indicated over-expressed CSF-1R was significantly associated with worse OS in hematological malignancy (HR = 2.29; P < .001, 1.49-3.09, 95% CI; model of fixed-effects; I2 = 0.0%, P < .001). Sensitivity analysis suggested that there was no study influencing the stability of the results. CONCLUSIONS The overexpression of CSF-1R was significantly predictive of worse prognosis in those who suffer from different kinds of malignancies, particularly in hematological malignancy, which indicates that it might be a potential biomarker of prognosis in cancer survival and a potential molecular target in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Huaqing Mo
- Cancer Center, The People's Hospital of Guangxi Zhuang Autonomous Region, China
| | - Yanrong Hao
- Cancer Center, The People's Hospital of Guangxi Zhuang Autonomous Region, China
| | - Yanru Lv
- Cancer Center, The People's Hospital of Guangxi Zhuang Autonomous Region, China
| | - Zenan Chen
- Cancer Center, The People's Hospital of Guangxi Zhuang Autonomous Region, China
| | - Jingyi Shen
- Cancer Center, The People's Hospital of Guangxi Zhuang Autonomous Region, China
| | - Shu Zhou
- Cancer Center, The People's Hospital of Guangxi Zhuang Autonomous Region, China
| | - MengJie Yin
- Cancer Center, The People's Hospital of Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
43
|
Huang XL, Khan MI, Wang J, Ali R, Ali SW, Zahra QUA, Kazmi A, Lolai A, Huang YL, Hussain A, Bilal M, Li F, Qiu B. Role of receptor tyrosine kinases mediated signal transduction pathways in tumor growth and angiogenesis-New insight and futuristic vision. Int J Biol Macromol 2021; 180:739-752. [PMID: 33737188 DOI: 10.1016/j.ijbiomac.2021.03.075] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/13/2021] [Accepted: 03/13/2021] [Indexed: 12/18/2022]
Abstract
In the past two decades, significant progress has been made in the past two decades towards the understanding of the basic mechanisms underlying cancer growth and angiogenesis. In this context, receptor tyrosine kinases (RTKs) play a pivotal role in cell proliferation, differentiation, growth, motility, invasion, and angiogenesis, all of which contribute to tumor growth and progression. Mutations in RTKs lead to abnormal signal transductions in several pathways such as Ras-Raf, MEK-MAPK, PI3K-AKT and mTOR pathways, affecting a wide range of biological functions including cell proliferation, survival, migration and vascular permeability. Increasing evidence demonstrates that multiple kinases are involved in angiogenesis including RTKs such as vascular endothelial growth factor, platelet derived growth factor, epidermal growth factor, insulin-like growth factor-1, macrophage colony-stimulating factor, nerve growth factor, fibroblast growth factor, Hepatocyte Growth factor, Tie 1 & 2, Tek, Flt-3, Flt-4 and Eph receptors. Overactivation of RTKs and its downstream regulation is implicated in tumor initiation and angiogenesis, representing one of the hallmarks of cancer. This review discusses the role of RTKs, PI3K, and mTOR, their involvement, and their implication in pro-oncogenic cellular processes and angiogenesis with effective approaches and newly approved drugs to inhibit their unrestrained action.
Collapse
Affiliation(s)
- Xiao Lin Huang
- School of Computer Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Muhammad Imran Khan
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Jing Wang
- First Affiliated Hospital of University of Science and Technology of China Hefei, Anhui 230036, China
| | - Rizwan Ali
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Syed Wajahat Ali
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Qurat-Ul-Ain Zahra
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ahsan Kazmi
- Department of Pathology, Al-Nafees Medical College and Hospital, Isra University, Islamabad 45600, Pakistan
| | - Arbelo Lolai
- School of Computer Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yu Lin Huang
- School of Computer Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Alamdar Hussain
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska Hospital, Huddinge, SE 141 86 Stockholm, Sweden; Department of Biosciences, COMSATS Institute of Information Technology, Chak Shahzad Campus, Islamabad 44000, Pakistan
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China
| | - Fenfen Li
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Bensheng Qiu
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
44
|
Role of Tumor-Associated Macrophages in Sarcomas. Cancers (Basel) 2021; 13:cancers13051086. [PMID: 33802565 PMCID: PMC7961818 DOI: 10.3390/cancers13051086] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Recent studies have shown the pro-tumoral role of tumor-associated macrophages (TAMs) not only in major types of carcinomas but also in sarcomas. Several types of TAM-targeted drugs have been investigated under clinical trials, which may represent a novel therapeutic approach for bone and soft-tissue sarcomas. Abstract Sarcomas are complex tissues in which sarcoma cells maintain intricate interactions with their tumor microenvironment. Tumor-associated macrophages (TAMs) are a major component of tumor-infiltrating immune cells in the tumor microenvironment and have a dominant role as orchestrators of tumor-related inflammation. TAMs promote tumor growth and metastasis, stimulate angiogenesis, mediate immune suppression, and limit the antitumor activity of conventional chemotherapy and radiotherapy. Evidence suggests that the increased infiltration of TAMs and elevated expression of macrophage-related genes are associated with poor prognoses in most solid tumors, whereas evidence of this in sarcomas is limited. Based on these findings, TAM-targeted therapeutic strategies, such as inhibition of CSF-1/CSF-1R, CCL2/CCR2, and CD47/SIRPα, have been developed and are currently being evaluated in clinical trials. While most of the therapeutic challenges that target sarcoma cells have been unsuccessful and the prognosis of sarcomas has plateaued since the 1990s, several clinical trials of these strategies have yielded promising results and warrant further investigation to determine their translational benefit in sarcoma patients. This review summarizes the roles of TAMs in sarcomas and provides a rationale and update of TAM-targeted therapy as a novel treatment approach for sarcomas.
Collapse
|
45
|
Mantovani A, Marchesi F, Jaillon S, Garlanda C, Allavena P. Tumor-associated myeloid cells: diversity and therapeutic targeting. Cell Mol Immunol 2021; 18:566-578. [PMID: 33473192 PMCID: PMC8027665 DOI: 10.1038/s41423-020-00613-4] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022] Open
Abstract
Myeloid cells in tumor tissues constitute a dynamic immune population characterized by a non-uniform phenotype and diverse functional activities. Both tumor-associated macrophages (TAMs), which are more abundantly represented, and tumor-associated neutrophils (TANs) are known to sustain tumor cell growth and invasion, support neoangiogenesis and suppress anticancer adaptive immune responses. In recent decades, several therapeutic approaches have been implemented in preclinical cancer models to neutralize the tumor-promoting roles of both TAMs and TANs. Some of the most successful strategies have now reached the clinic and are being investigated in clinical trials. In this review, we provide an overview of the recent literature on the ever-growing complexity of the biology of TAMs and TANs and the development of the most promising approaches to target these populations therapeutically in cancer patients.
Collapse
Affiliation(s)
- Alberto Mantovani
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy.
- Department of Biomedical Science, Humanitas University, Rozzano, Italy.
- The William Harvey Research Institute, Queen Mary University of London, London, UK.
| | - Federica Marchesi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sebastien Jaillon
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
- Department of Biomedical Science, Humanitas University, Rozzano, Italy
| | - Cecilia Garlanda
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
- Department of Biomedical Science, Humanitas University, Rozzano, Italy
| | - Paola Allavena
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| |
Collapse
|
46
|
Rodriguez E, Boelaars K, Brown K, Eveline Li RJ, Kruijssen L, Bruijns SCM, van Ee T, Schetters STT, Crommentuijn MHW, van der Horst JC, van Grieken NCT, van Vliet SJ, Kazemier G, Giovannetti E, Garcia-Vallejo JJ, van Kooyk Y. Sialic acids in pancreatic cancer cells drive tumour-associated macrophage differentiation via the Siglec receptors Siglec-7 and Siglec-9. Nat Commun 2021; 12:1270. [PMID: 33627655 PMCID: PMC7904912 DOI: 10.1038/s41467-021-21550-4] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/25/2021] [Indexed: 11/09/2022] Open
Abstract
Changes in glycosylation during tumour progression are a key hallmark of cancer. One of the glycan moieties generally overexpressed in cancer are sialic acids, which can induce immunomodulatory properties via binding to Siglec receptors. We here show that Pancreatic Ductal Adenocarcinoma (PDAC) tumour cells present an increased sialylation that can be recognized by Siglec-7 and Siglec-9 on myeloid cells. We identified the expression of the α2,3 sialyltransferases ST3GAL1 and ST3GAL4 as main contributor to the synthesis of ligands for Siglec-7 and Siglec-9 in tumour cells. Analysing the myeloid composition in PDAC, using single cell and bulk transcriptomics data, we identified monocyte-derived macrophages as contributors to the poor clinical outcome. Tumour-derived sialic acids dictate monocyte to macrophage differentiation via signalling through Siglec-7 and Siglec-9. Moreover, triggering of Siglec-9 in macrophages reduce inflammatory programmes, while increasing PD-L1 and IL-10 expression, illustrating that sialic acids modulate different myeloid cells. This work highlights a critical role for sialylated glycans in controlling immune suppression and provides new potential targets for cancer immunotherapy in PDAC.
Collapse
Affiliation(s)
- Ernesto Rodriguez
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Kelly Boelaars
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Kari Brown
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - R J Eveline Li
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Laura Kruijssen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Sven C M Bruijns
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Thomas van Ee
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Sjoerd T T Schetters
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Matheus H W Crommentuijn
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Joost C van der Horst
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Nicole C T van Grieken
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, Netherlands
| | - Sandra J van Vliet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Geert Kazemier
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Elisa Giovannetti
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Juan J Garcia-Vallejo
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands.
| |
Collapse
|
47
|
Freuchet A, Salama A, Remy S, Guillonneau C, Anegon I. IL-34 and CSF-1, deciphering similarities and differences at steady state and in diseases. J Leukoc Biol 2021; 110:771-796. [PMID: 33600012 DOI: 10.1002/jlb.3ru1120-773r] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/04/2021] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Although IL-34 and CSF-1 share actions as key mediators of monocytes/macrophages survival and differentiation, they also display differences that should be identified to better define their respective roles in health and diseases. IL-34 displays low sequence homology with CSF-1 but has a similar general structure and they both bind to a common receptor CSF-1R, although binding and subsequent intracellular signaling shows differences. CSF-1R expression has been until now mainly described at a steady state in monocytes/macrophages and myeloid dendritic cells, as well as in some cancers. IL-34 has also 2 other receptors, protein-tyrosine phosphatase zeta (PTPζ) and CD138 (Syndecan-1), expressed in some epithelium, cells of the central nervous system (CNS), as well as in numerous cancers. While most, if not all, of CSF-1 actions are mediated through monocyte/macrophages, IL-34 has also other potential actions through PTPζ and CD138. Additionally, IL-34 and CSF-1 are produced by different cells in different tissues. This review describes and discusses similarities and differences between IL-34 and CSF-1 at steady state and in pathological situations and identifies possible ways to target IL-34, CSF-1, and its receptors.
Collapse
Affiliation(s)
- Antoine Freuchet
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Apolline Salama
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Séverine Remy
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|
48
|
Allavena P, Anfray C, Ummarino A, Andón FT. Therapeutic Manipulation of Tumor-associated Macrophages: Facts and Hopes from a Clinical and Translational Perspective. Clin Cancer Res 2021; 27:3291-3297. [PMID: 33531428 DOI: 10.1158/1078-0432.ccr-20-1679] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/09/2020] [Accepted: 01/21/2021] [Indexed: 11/16/2022]
Abstract
The stroma of most solid tumors is populated by myeloid cells, which mostly represent macrophages. Tumor-associated macrophages (TAMs), strongly influenced by cancer cell-derived factors, are key drivers of immunosuppression and support tumor growth and spread to distant sites. Their accurate quantification and characterization in the tumor microenvironment are gaining prognostic value: increasing evidence demonstrates their ability to hamper cancer patients' response to chemotherapy, as well as to immunotherapies based on checkpoint inhibition. Therefore, strategies to counteract their negative effects are nowadays gaining momentum at preclinical, translational, and clinical levels. Our knowledge of the biology of TAMs has greatly advanced in the last years; several strategies to target and reprogram their functions to become antitumor effectors have proven successful in experimental preclinical tumor models; on the other hand, few approaches have so far been effectively translated into clinic practice. A growing interest in the therapeutic manipulation of TAMs is evidenced by numerous early-phase clinical trials, which are continuously fueled by new discoveries from basic research. This gives us hope that the targeting and sustained reprogramming of TAMs will be more specific to synergize with current therapies and maximize antitumor responses in patients.
Collapse
Affiliation(s)
- Paola Allavena
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, Italy.
| | - Clément Anfray
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, Italy
| | - Aldo Ummarino
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, Italy
| | - Fernando Torres Andón
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, Italy.,Center for Research in Molecular Medicine & Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
49
|
Yan S, Wan G. Tumor-associated macrophages in immunotherapy. FEBS J 2021; 288:6174-6186. [PMID: 33492779 DOI: 10.1111/febs.15726] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/23/2020] [Accepted: 01/22/2021] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAMs) are essential components of the tumor microenvironment involved in the progression and metastasis of cancer. They are intimately involved in angiogenesis and immunosuppression in normal and malignant tissues, as well as pro-fibrotic activities. With the development of immunotherapy, eradication of cancer cells through activation of the innate immune system has achieved inspiring results, whereas only a handful of patients show a durable response. The tumor-suppressive environment has been investigated with respect to playing a vital role in cancer relapse. In this review, we uncover the heterogeneity of the origin of TAMs, as well as the functions of TAMs in tumor progression associated with intricate regulatory networks in the tumor microenvironment, aiming to inspire therapeutic insight for tumor immunotherapy.
Collapse
Affiliation(s)
- Shijia Yan
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (Cultivation), Guangdong Province Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Guohui Wan
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (Cultivation), Guangdong Province Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
50
|
|