1
|
Castro F, Pinto ML, Leite Pereira C, Serre K, Costa ÂM, Cavadas B, Barbosa MA, Vermaelen K, León S, Serrano D, Gärtner F, Calvo A, Gonçalves RM, De Wever O, Oliveira MJ. Chitosan/γ-PGA nanoparticles and IFN-γ immunotherapy: A dual approach for triple-negative breast cancer treatment. J Control Release 2025; 379:621-635. [PMID: 39832747 DOI: 10.1016/j.jconrel.2025.01.042] [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: 09/23/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Interferon-γ (IFN-γ) is a key mediator in antitumor immunity and immunotherapy responses, yet its clinical applications remain restricted to chronic granulomatous disease and malignant osteopetrosis. IFN-γ effectiveness as a standalone treatment has shown limited success in clinical trials and its potential for synergistic effects when combined with immunotherapies is under clinical exploration. A particularly compelling combination is that of IFN-γ with Toll-like receptor (TLR) agonists that holds significant promise for cancer treatment. Previously, we demonstrated chitosan (Ch)/poly(γ-glutamic acid) (γ-PGA) nanoparticles (NPs), known to activate TLRs, as adjuvants to radiotherapy by remodeling breast tumor microenvironment and systemic immunosuppression. These immunomodulatory abilities make Ch/γ-PGA NPs promising adjuvants to IFN-γ-based therapies. Here, we addressed the synergistic therapeutic potential of combining Ch/γ-PGA NPs with IFN-γ therapy in the 4T1 orthotopic breast tumor mouse model. While control animals (placebo-treated) had progressive tumor growth and lung metastases, those treated with either NPs or IFN-γ alone had a significant slower tumor growth. Remarkably, primary tumor growth was halted throughout the duration of the treatment when both treatments were combined. Although the animals did not achieve durable complete responses upon treatment withdrawal, it was notable that the NPs plus IFN-γ group presented a lower lung metastatic burden compared to other groups. Systemically, the combination therapy slightly attenuated immunosuppression and the percentage of splenic myeloid cells, while increased the percentage of T helper 1 cells and of cytotoxic T cells. Overall, this proof-of-concept study suggests that Ch/γ-PGA NPs potentiate IFN-γ effects to reduce tumor progression, presenting a novel approach for anticancer strategies.
Collapse
Affiliation(s)
- Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Marta Laranjeiro Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Catarina Leite Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Karine Serre
- GIMM - Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas Moniz, Lisboa, Portugal
| | - Ângela Margarida Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Bruno Cavadas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Mário Adolfo Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Karim Vermaelen
- Tumor Immunology Laboratory, Department of Pulmonary Medicine and Immuno-Oncology Network Ghent, Ghent University Hospital, Ghent, Belgium; CRIG - Cancer Research Institute Ghent, Ghent University, Belgium
| | - Sergio León
- IdiSNA - Navarra Institute for Health Research, Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain; CIBERONC (-) Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Diego Serrano
- IdiSNA - Navarra Institute for Health Research, Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain
| | - Fátima Gärtner
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Alfonso Calvo
- IdiSNA - Navarra Institute for Health Research, Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain; CIBERONC (-) Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Raquel Madeira Gonçalves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Olivier De Wever
- CRIG - Cancer Research Institute Ghent, Ghent University, Belgium; LECR - Laboratory Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Belgium
| | - Maria José Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
2
|
Hsiao YJ, Hsieh MS, Chang GC, Hsu YC, Wang CY, Chen YM, Chen YL, Yang PC, Yu SL. Tp53 determines the spatial dynamics of M1/M2 tumor-associated macrophages and M1-driven tumoricidal effects. Cell Death Dis 2025; 16:38. [PMID: 39843434 PMCID: PMC11754596 DOI: 10.1038/s41419-025-07346-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/28/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025]
Abstract
The spatial role of M1 and M2 tumor-associated macrophages (M1/M2 TAMs) in precision medicine remains unclear. EGFR and TP53 are among the most frequently mutated genes in lung adenocarcinoma. We characterized the mutation status and density of M1/M2 TAMs within tumor islets and stroma in 117 lung adenocarcinomas using next-generation sequencing and immunohistochemistry, respectively. Stromal M1 TAMs were positively correlated with disease progression and smoking history. In contrast, islet M1/M2 TAMs were predominantly found in tumors with wild-type TP53 (wtp53) but not associated with EGFR status. The presence of wtp53 was associated with the spatial distribution of M1/M2 TAMs in tumor islets and stroma. Additionally, dominance of islet M1 TAMs and M1-signature were significantly associated with improved survival in patients with wtp53 lung adenocarcinoma, unlike in those with mutant TP53. Conditioned medium from M1 macrophages (M1 CM) induced apoptosis in wtp53 cells through increased p53 accumulation. We found that interferons in M1 CM activate JAK1/TYK2 via IFNARs, leading to enhanced STAT1 expression and Y701 phosphorylation. This activation facilitates p53-STAT1 interactions, reduces the interaction between p53 and MDM2, and subsequently decreases p53 ubiquitination. M1 CM inhibited tumorigenesis, and silencing p53 reduced the anti-tumor efficacy of polyinosinic:polycytidylic acid (poly I:C) in vivo. Furthermore, higher M1-signature was significantly associated with better responses and survival following anti-PD1 treatment in wtp53 melanomas. IFNs/STAT1/p53 signaling was critical for the anti-tumor activity of M1 macrophages. These findings suggest that p53 modulates the spatial balance of M1/M2 TAMs, and the tumoricidal effects of M1 TAMs depend on p53 status. Thus, p53 companion diagnostics could facilitate the development of M1-oriented therapies, which may be particularly beneficial for wtp53 patients when combined with immunotherapy.
Collapse
Affiliation(s)
- Yi-Jing Hsiao
- Department of Clinical and Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Gee-Chen Chang
- Division of Pulmonary Medicine, Department of Internal Medicine, Chung Shan Medical University Hospital Taichung, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yin-Chen Hsu
- Department of Clinical and Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Yu Wang
- Department of Clinical and Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yan-Ming Chen
- Department of Clinical and Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Ling Chen
- Department of Clinical and Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pan-Chyr Yang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Sung-Liang Yu
- Department of Clinical and Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan.
- Graduate Institute of Pathology, National Taiwan University College of Medicine, Taipei, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Graduate Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan.
- Graduate School of Advanced Technology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
3
|
Wang M, Wan Q, Wang C, Jing Q, Nie Y, Zhang X, Chen X, Yang D, Pan R, Li L, Zhu L, Gui H, Chen S, Deng Y, Chen T, Nie Y. Combinational delivery of TLR4 and TLR7/8 agonist enhanced the therapeutic efficacy of immune checkpoint inhibitors to colon tumor. Mol Cell Biochem 2025; 480:445-458. [PMID: 38507020 DOI: 10.1007/s11010-024-04966-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 02/11/2024] [Indexed: 03/22/2024]
Abstract
Immunotherapy is regarded as a potent cancer treatment, with DC vaccines playing a crucial role. Although clinical trials have demonstrated the safety and efficacy of DC vaccines, loading antigens in vitro is challenging, and their therapeutic effects remain unpredictable. Moreover, the diverse subtypes and maturity states of DCs in the body could induce both immune responses and immune tolerance, potentially affecting the vaccine's efficacy. Hence, the optimization of DC vaccines remains imperative. Our study discovered a new therapeutic strategy by using CT26 and MC38 mouse colon cancer models, as well as LLC mouse lung cancer models. The strategy involved the synergistic activation of DCs through intertumoral administration of TLR4 agonist high-mobility group nucleosome binding protein 1 (HMGN1) and TLR7/8 agonist (R848/resiquimod), combined with intraperitoneal administration of TNFR2 immunosuppressant antibody. The experimental results indicated that the combined use of HMGN1, R848, and α-TNFR2 had no effect on LLC cold tumors. However, it was effective in eradicating CT26 and MC38 colon cancer and inducing long-term immune memory. The combination of these three drugs altered the TME and promoted an increase in anti-tumor immune components. This may provide a promising new treatment strategy for colon cancer.
Collapse
Affiliation(s)
- Mengjiao Wang
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Quan Wan
- School of Preclinical Medicine of Zunyi Medical University, Zunyi, 563000, China
| | - Chenglv Wang
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Qianyu Jing
- School of Preclinical Medicine of Zunyi Medical University, Zunyi, 563000, China
| | - Yujie Nie
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Xiangyan Zhang
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, SAR, China
| | - De Yang
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute at Frederick, NIH, Frederick, MD, USA
| | - Runsang Pan
- Department of Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - Linzhao Li
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Lan Zhu
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Huan Gui
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Shuanghui Chen
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Yuezhen Deng
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Tao Chen
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Yingjie Nie
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China.
| |
Collapse
|
4
|
Bhaliya KR, Anwer M, Munn A, Wei MQ. New horizons in cancer immunotherapy: The evolving role of R848 and R837 (Review). Mol Clin Oncol 2025; 22:4. [PMID: 39563999 PMCID: PMC11574705 DOI: 10.3892/mco.2024.2799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/07/2024] [Indexed: 11/21/2024] Open
Abstract
Therapeutic approaches that increase the efficacy and safety of cancer treatments and improve disease outcomes have been developed worldwide. Immunotherapy uses the body's immune system to inhibit cancerous growth in tissues and organs. Various approaches have been developed to effectively control and inhibit cancerous growth, including checkpoint inhibitors, T-cell transfer therapy, monoclonal antibodies, vaccines and immunomodulators. Toll-like receptors (TLRs) target malignant cells by equipping the immune response. In addition, TLR agonists serve a key role in promoting the innate immune system and initiating antigen-specific T-cell responses. Notably, TLRs and TLR agonists have been utilized as monotherapies or in combination for the treatment of cancer. The present study aimed to review the use of R848 and R837 as TLR agonists, and outline their use as key immunomodulators in cancer therapy.
Collapse
Affiliation(s)
- Krupa R Bhaliya
- Menzies Health Institute, School of Medical Science, Griffith University, Southport, Queensland 4215, Australia
| | - Muneera Anwer
- Menzies Health Institute, School of Medical Science, Griffith University, Southport, Queensland 4215, Australia
| | - Alan Munn
- Menzies Health Institute, School of Medical Science, Griffith University, Southport, Queensland 4215, Australia
| | - Ming Q Wei
- Menzies Health Institute, School of Medical Science, Griffith University, Southport, Queensland 4215, Australia
| |
Collapse
|
5
|
Shu X, Xie Y, Shu M, Ou X, Yang J, Wu Z, Zhang X, Zhang J, Zeng H, Shao L. Acute effects of TLR3 agonist Poly(I:C) on bone marrow hematopoietic progenitor cells in mice. Immunol Lett 2024; 270:106927. [PMID: 39265918 DOI: 10.1016/j.imlet.2024.106927] [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: 08/07/2023] [Revised: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Hematopoietic progenitor cells (HPCs) in bone marrow with limited abilities for self-renewal and differentiation continuously supply hematopoietic cells through life. When suffering infection or inflammation, HPCs will actively proliferate to provide differentiated hematopoietic cells to maintain hematopoietic homeostasis. Poly(I:C), an agonist of TLR3, can specifically activate Type I interferon (IFN-I) signaling which exerts anti-inflammatory effects and influence hematopoiesis after infection. However, the effects of Poly(I:C)-induced IFN-I on the bone marrow hematopoietic system still deserve attention. In this study, our results revealed the efficacy of the IFN-I model, with a remarkably decrease in HPCs and a sharp elevation in LSKs numbers after single dose of Poly(I:C) injection. Apoptotic ratios of HPCs and LSKs significantly increased 48 h after Poly(I:C) treatment. Application of Poly(I:C) prompted the transition of HPCs and LSKs from G0 to G1 phases, potentially leading to the accelerated exhaustion of HPCs. From the cobblestone area-forming cell (CAFC) assay, we speculate that Poly(I:C) impairs the differentiation capacity of HPCs as well as their colony-forming ability. RT-qPCR and immunohistochemistry revealed significant upregulation of IFN-I associated genes and proteins following Poly(I:C) treatment. In conclusion, a single dose of Poly(I:C) induced an acute detrimental effect on HPCs within 48 h potentially due to TLR3 engagement. This activation cascaded into a robust IFN-I response emanating from the bone marrow, underscoring the intricate immunological dynamics at play following Poly(I:C) intervention.
Collapse
Affiliation(s)
- Xin Shu
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Yuxuan Xie
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Manling Shu
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Xiangying Ou
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Juan Yang
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Zhenyu Wu
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Xuan Zhang
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Jinfu Zhang
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Huihong Zeng
- Department of Histology and Embryology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, China; Basic Medical Experiment Center, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China.
| | - Lijian Shao
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, China; School of Public Health, Jiangxi Medical College, Nanchang University, China.
| |
Collapse
|
6
|
Pedersoli F, Mohammad IS, Patel AK, Kessler J, Chao C, Liu B, Lall C, Guerra C, Park JJ, Boas FE. Bioinspired intratumoral infusion port catheter improves local drug delivery in the liver. Sci Rep 2024; 14:27782. [PMID: 39538011 PMCID: PMC11561066 DOI: 10.1038/s41598-024-79694-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
Tumor immune modulation can be achieved using intratumoral injection of different immunomodulators during different phases of the cancer-immunity cycle. Intratumoral infusion catheters have been used in brain tumors, but these are not suitable outside the brain, where breathing motion results in catheter migration. Here, we use microstereolithography to manufacture a barbed sidehole catheter, modeled after the barbs in a bee stinger, where the barbs maintain the catheter position in the tumor, and sideholes within the barbs infuse drug into tumor tissue. In pig liver, we demonstrated 183-fold higher local drug concentration using the barbed sidehole catheter, compared to intravenous injection of water-soluble drug. High resistance sideholes and pulsatile injection both generate higher pressure in the catheter, which overcomes the tissue pressure, resulting in more drug delivery into tumor. A physical model of intratumoral infusion catheters accurately predicts the observed drug delivery results. Our catheter design is retained in the liver (and does not migrate out with breathing motion), and it preferentially infuses the drug into tumor tissue (not intratumoral vessels).
Collapse
Affiliation(s)
- Federico Pedersoli
- Interventional Radiology, Imaging Institute of Southern Switzerland (IIMSI), Ente Ospedaliero Cantonale (EOC), Locarno, Switzerland
| | - Imran Shair Mohammad
- Department of Radiology / Interventional Radiology, City of Hope Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Anup Kumar Patel
- Department of Radiology / Interventional Radiology, City of Hope Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Jonathan Kessler
- Department of Radiology / Interventional Radiology, City of Hope Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Cherng Chao
- Department of Radiology / Interventional Radiology, City of Hope Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Bo Liu
- Department of Radiology / Interventional Radiology, City of Hope Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Chandana Lall
- Department of Radiology / Interventional Radiology, City of Hope Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | | | - John J Park
- Harbor-UCLA Medical Center, Torrance, CA, USA
| | - F Edward Boas
- Department of Radiology / Interventional Radiology, City of Hope Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
7
|
Newman MJ. Invention and characterization of a systemically administered, attenuated and killed bacteria-based multiple immune receptor agonist for anti-tumor immunotherapy. Front Immunol 2024; 15:1462221. [PMID: 39606250 PMCID: PMC11599860 DOI: 10.3389/fimmu.2024.1462221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Activation of immune receptors, such as Toll-like (TLR), NOD-like (NLR) and Stimulator of Interferon Genes (STING) is critical for efficient innate and adaptive immunity. Gram-negative bacteria (G-NB) contain multiple TLR, NOD and STING agonists. Potential utility of G-NB for cancer immunotherapy is supported by observations of tumor regression in the setting of infection and Coley's Toxins. Coley reported that intravenous (i.v.) administration was likely most effective but produced uncontrollable toxicity. The discovery of TLRs and their agonists, particularly the potent TLR4 agonist lipopolysaccharide (LPS)-endotoxin, comprising ~75% of the outer membrane of G-NB, suggests that LPS may be both a critical active ingredient and responsible for dose-limiting i.v. toxicity of G-NB. This communication reports the production of killed, stabilized, intact bacteria products from non-pathogenic G-NB with ~96% reduction of LPS-endotoxin activity. One resulting product candidate, Decoy10, was resistant to standard methods of cell disruption and contained TLR2,4,8,9, NOD2 and STING agonist activity. Decoy10 also exhibited reduced i.v. toxicity in mice and rabbits, and a largely uncompromised ability to induce cytokine and chemokine secretion by human immune cells in vitro, all relative to unprocessed, parental bacterial cells. Decoy10 and a closely related product, Decoy20, produced single agent anti-tumor activity or combination-mediated durable regression of established subcutaneous, metastatic or orthotopic colorectal, hepatocellular (HCC), pancreatic, and non-Hodgkin's lymphoma (NHL) tumors in mice, with induction of both innate and adaptive immunological memory (syngeneic and human tumor xenograft models). Decoy bacteria combination-mediated regressions were observed with a low-dose, oral non-steroidal anti-inflammatory drug (NSAID), anti-PD-1 checkpoint therapy, low-dose cyclophosphamide (LDC), and/or a targeted antibody (rituximab). Efficient tumor eradication was associated with plasma expression of 15-23 cytokines and chemokines, broad induction of cytokine, chemokine, innate and adaptive immune pathway genes in tumors, cold to hot tumor inflammation signature transition, and required NK, CD4+ and CD8+ T cells, collectively demonstrating a role for both innate and adaptive immune activation in the anti-tumor immune response.
Collapse
|
8
|
Mantovani A, Marchesi F, Di Mitri D, Garlanda C. Macrophage diversity in cancer dissemination and metastasis. Cell Mol Immunol 2024; 21:1201-1214. [PMID: 39402303 PMCID: PMC11528009 DOI: 10.1038/s41423-024-01216-z] [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: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 11/02/2024] Open
Abstract
Invasion and metastasis are hallmarks of cancer. In addition to the well-recognized hematogenous and lymphatic pathways of metastasis, cancer cell dissemination can occur via the transcoelomic and perineural routes, which are typical of ovarian and pancreatic cancer, respectively. Macrophages are a universal major component of the tumor microenvironment and, in established tumors, promote growth and dissemination to secondary sites. Here, we review the role of tumor-associated macrophages (TAMs) in cancer cell dissemination and metastasis, emphasizing the diversity of myeloid cells in different tissue contexts (lungs, liver, brain, bone, peritoneal cavity, nerves). The generally used models of lung metastasis fail to capture the diversity of pathways and tissue microenvironments. A better understanding of TAM diversity in different tissue contexts may pave the way for tailored diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Alberto Mantovani
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy.
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy.
- William Harvey Research Institute, Queen Mary University, London, UK.
| | - Federica Marchesi
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Diletta Di Mitri
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| |
Collapse
|
9
|
Wang J, Wang Y, Jiang X. Targeting anticancer immunity in melanoma tumour microenvironment: unleashing the potential of adjuvants, drugs, and phytochemicals. J Drug Target 2024; 32:1052-1072. [PMID: 39041142 DOI: 10.1080/1061186x.2024.2384071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Melanoma poses a challenge in oncology because of its aggressive nature and limited treatment modalities. The tumour microenvironment (TME) in melanoma contains unique properties such as an immunosuppressive and high-density environment, unusual vasculature, and a high number of stromal and immunosuppressive cells. In recent years, numerous experiments have focused on boosting the immune system to effectively remove malignant cells. Adjuvants, consisting of phytochemicals, toll-like receptor (TLR) agonists, and cytokines, have shown encouraging results in triggering antitumor immunity and augmenting the therapeutic effectiveness of anticancer therapy. These adjuvants can stimulate the maturation of dendritic cells (DCs) and infiltration of cytotoxic CD8+ T lymphocytes (CTLs). Furthermore, nanocarriers can help to deliver immunomodulators and antigens directly to the tumour stroma, thereby improving their efficacy against malignant cells. The remodelling of melanoma TME utilising phytochemicals, agonists, and other adjuvants can be combined with current modalities for improving therapy outcomes. This review article explores the potential of adjuvants, drugs, and their nanoformulations in enhancing the anticancer potency of macrophages, CTLs, and natural killer (NK) cells. Additionally, the capacity of these agents to repress the function of immunosuppressive components of melanoma TME, such as immunosuppressive subsets of macrophages, stromal and myeloid cells will be discussed.
Collapse
Affiliation(s)
- Jingping Wang
- Emergency Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Yaping Wang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Xiaofang Jiang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| |
Collapse
|
10
|
Sancho-Albero M, Fenaroli AL, Scaccaglia M, Matteo C, Grasselli C, Zucchetti M, Frapolli R, Nastasi C, De Cola L. Two Different Responsive Organosilica Nanocarriers to Combine Chemo- and Immunotherapy against Cancer. ACS OMEGA 2024; 9:41225-41235. [PMID: 39398182 PMCID: PMC11465578 DOI: 10.1021/acsomega.4c02838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/22/2024] [Accepted: 08/08/2024] [Indexed: 10/15/2024]
Abstract
The combination of chemo- and immunotherapy was recently demonstrated to improve a patient's response to therapy, giving rise to an emerging cancer treatment known as chemoimmunotherapy (CIT). Despite the promising benefits of CIT, the most important challenges are (i) the simultaneous or time-controlled delivery of two drugs and (ii) the selective uptake into different cells for each of the drugs: cancer cells for the chemotherapeutic and macrophages for the immunostimulation actives. Herein, a delivery strategy based on morphologically different stimuli-responsive breakable organosilica nanocarriers is exploited to transport two distinct drugs in the different cells using different times of delivery. We employ stimulus-sensitive, PEGylated organosilica nanocages to encapsulate the chemotherapeutic agent doxorubicin, which is preferentially taken up by tumor cells vs macrophages. On the other hand, similar size mesoporous organosilica nanoparticles, preferentially internalized by macrophages, are filled with the immunostimulator resiquimod. The administration in a sequential manner of the two different nanocarriers allowed us to assess the integrated effect of the combined therapy versus treatment with a single drug. In vitro work clearly shows an important reduction of tumor cell viability when both chemo- and immunotherapeutic agents are delivered.
Collapse
Affiliation(s)
- Maria Sancho-Albero
- Department
of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Alessia Lucrezia Fenaroli
- Department
of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Mirco Scaccaglia
- Department
of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Cristina Matteo
- Department
of Oncology, Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Chiara Grasselli
- Department
of Oncology, Immunopharmacology Unit, Istituto
di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Massimo Zucchetti
- Department
of Oncology, Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Roberta Frapolli
- Department
of Oncology, Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Claudia Nastasi
- Department
of Oncology, Immunopharmacology Unit, Istituto
di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Luisa De Cola
- Department
of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
- Department
of Pharmaceutical Science, DISFARM, Università
degli Studi di Milano, Milan 20133, Italy
| |
Collapse
|
11
|
Rwandamuriye FX, Wang T, Zhang H, Elaskalani O, Kuster J, Ye X, Vitali B, Schreurs J, Orozco Morales ML, Norret M, Evans CW, Zemek RM, Iyer KS, Lesterhuis WJ, Wylie B. Local therapy with combination TLR agonists stimulates systemic anti-tumor immunity and sensitizes tumors to immune checkpoint blockade. Oncoimmunology 2024; 13:2395067. [PMID: 39188754 PMCID: PMC11346538 DOI: 10.1080/2162402x.2024.2395067] [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: 03/19/2024] [Revised: 07/26/2024] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
Toll-like receptor (TLR) agonists are being developed as anti-cancer therapeutics due to their potent immunostimulatory properties. However, clinical trials testing TLR agonists as monotherapy have often failed to demonstrate significant improvement over standard of care. We hypothesized that the anti-cancer efficacy of TLR agonist immunotherapy could be improved by combinatorial approaches. To prevent increased toxicity, often seen with systemic combination therapies, we developed a hydrogel to deliver TLR agonist combinations at low doses, locally, during cancer debulking surgery. Using tumor models of WEHI 164 and bilateral M3-9-M sarcoma and CT26 colon carcinoma, we assessed the efficacy of pairwise combinations of poly(I:C), R848, and CpG in controlling local and distant tumor growth. We show that combination of the TLR3 agonist poly(I:C) and TLR7/8 agonist R848 drives anti-tumor immunity against local and distant tumors. In addition, combination of local poly(I:C) and R848 sensitized tumors to systemic immune checkpoint blockade, improving tumor control. Mechanistically, we demonstrate that local therapy with poly(I:C) and R848 recruits inflammatory monocytes to the tumor draining lymph nodes early in the anti-tumor response. Finally, we provide proof of concept for intraoperative delivery of poly(I:C) and R848 together via a surgically applicable biodegradable hydrogel.
Collapse
Affiliation(s)
| | - Tao Wang
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Hanfu Zhang
- School of Molecular Sciences, University of Western Australia, Crawley, WA, Australia
| | - Omar Elaskalani
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Jorren Kuster
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Xueting Ye
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Breana Vitali
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Juliët Schreurs
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | | | - Marck Norret
- School of Molecular Sciences, University of Western Australia, Crawley, WA, Australia
| | - Cameron W. Evans
- School of Molecular Sciences, University of Western Australia, Crawley, WA, Australia
| | - Rachael M. Zemek
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - K. Swaminathan Iyer
- School of Molecular Sciences, University of Western Australia, Crawley, WA, Australia
| | - W. Joost Lesterhuis
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Ben Wylie
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
12
|
López-Cuevas P, Oates TCL, Tong Q, McGowan LM, Cross SJ, Xu C, Zhao Y, Yin Z, Toye AM, Boussahel A, Hammond CL, Mann S, Martin P. Reprogramming macrophages with R848-loaded artificial protocells to modulate skin and skeletal wound healing. J Cell Sci 2024; 137:jcs262202. [PMID: 39078119 PMCID: PMC11385641 DOI: 10.1242/jcs.262202] [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: 04/16/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
After tissue injury, inflammatory cells are rapidly recruited to the wound where they clear microbes and other debris, and coordinate the behaviour of other cell lineages at the repair site in both positive and negative ways. In this study, we take advantage of the translucency and genetic tractability of zebrafish to evaluate the feasibility of reprogramming innate immune cells in vivo with cargo-loaded protocells and investigate how this alters the inflammatory response in the context of skin and skeletal repair. Using live imaging, we show that protocells loaded with R848 cargo (which targets TLR7 and TLR8 signalling), are engulfed by macrophages resulting in their switching to a pro-inflammatory phenotype and altering their regulation of angiogenesis, collagen deposition and re-epithelialization during skin wound healing, as well as dampening osteoblast and osteoclast recruitment and bone mineralization during fracture repair. For infected skin wounds, R848-reprogrammed macrophages exhibited enhanced bactericidal activities leading to improved healing. We replicated our zebrafish studies in cultured human macrophages, and showed that R848-loaded protocells similarly reprogramme human cells, indicating how this strategy might be used to modulate wound inflammation in the clinic.
Collapse
Affiliation(s)
- Paco López-Cuevas
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Tiah C L Oates
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Qiao Tong
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Stephen J Cross
- Wolfson Bioimaging Facility, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Can Xu
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Yu Zhao
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Zhuping Yin
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell Products, University of Bristol, Bristol BS34 7QH, UK
| | - Asme Boussahel
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Stephen Mann
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- Max Planck Bristol Centre for Minimal Biology, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
13
|
Zhang W, Wang M, Ji C, Liu X, Gu B, Dong T. Macrophage polarization in the tumor microenvironment: Emerging roles and therapeutic potentials. Biomed Pharmacother 2024; 177:116930. [PMID: 38878638 DOI: 10.1016/j.biopha.2024.116930] [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: 04/14/2024] [Revised: 05/31/2024] [Accepted: 06/09/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) is a combination of tumor cells and indigenous host stroma, which consists of tumor-infiltrating immune cells, endothelial cells, fibroblasts, pericytes, and non-cellular elements. Tumor-associated macrophages (TAMs) represent the major tumor-infiltrating immune cell type and are generally polarized into two functionally contradictory subtypes, namely classical activated M1 macrophages and alternatively activated M2 macrophages. Macrophage polarization refers to how macrophages are activated at a given time and space. The interplay between the TME and macrophage polarization can influence tumor initiation and progression, making TAM a potential target for cancer therapy. Here, we review the latest investigations on factors orchestrating macrophage polarization in the TME, how macrophage polarization affects tumor progression, and the perspectives in modulating macrophage polarization for cancer immunotherapy.
Collapse
Affiliation(s)
- Wenru Zhang
- Department of Natural Products Chemistry, Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Mengmeng Wang
- Department of Natural Products Chemistry, Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Chonghao Ji
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaohui Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 2A Nanwei Road, Xicheng District, Beijing 100050, China
| | - Bowen Gu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, United States.
| | - Ting Dong
- Department of Natural Products Chemistry, Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
14
|
Alessandroni L, Sagratini G, Gagaoua M. Proteomics and bioinformatics analyses based on two-dimensional electrophoresis and LC-MS/MS for the primary characterization of protein changes in chicken breast meat from divergent farming systems: Organic versus antibiotic-free. FOOD CHEMISTRY. MOLECULAR SCIENCES 2024; 8:100194. [PMID: 38298469 PMCID: PMC10828576 DOI: 10.1016/j.fochms.2024.100194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/02/2024] [Accepted: 01/13/2024] [Indexed: 02/02/2024]
Abstract
Proteomics is a key analytical method in meat research thanks to its potential in investigating the proteins at interplay in post-mortem muscles. This study aimed to characterize for the first time the differences in early post-mortem muscle proteomes of chickens raised under two farming systems: organic versus antibiotic-free. Forty post-mortem Pectoralis major muscle samples from two chicken strains (Ross 308 versus Ranger Classic) reared under organic versus antibiotic-free farming systems were characterized and compared using two-dimensional electrophoresis and LC-MS/MS mass spectrometry. Within antibiotic-free and organic farming systems, 14 and 16 proteins were differentially abundant between Ross 308 and Ranger Classic, respectively. Within Ross 308 and Ranger Classic chicken strains, 12 and 18 proteins were differentially abundant between organic and antibiotic-free, respectively. Bioinformatics was applied to investigate the molecular pathways at interplay, which highlighted the key role of muscle structure and energy metabolism. Antibiotic-free and organic farming systems were found to significantly impact the muscle proteome of chicken breast meat. This paper further proposes a primary list of putative protein biomarkers that can be used for chicken meat or farming system authenticity.
Collapse
Affiliation(s)
- Laura Alessandroni
- School of Pharmacy, Chemistry Interdisciplinary Project (CHIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Gianni Sagratini
- School of Pharmacy, Chemistry Interdisciplinary Project (CHIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | | |
Collapse
|
15
|
Ramsey HE, Gorska AE, Smith BN, Monteith AJ, Fuller L, Arrate MP, Savona MR. TLR3 agonism augments CD47 inhibition in acute myeloid leukemia. Haematologica 2024; 109:2111-2121. [PMID: 38152031 PMCID: PMC11215363 DOI: 10.3324/haematol.2023.283850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/15/2023] [Indexed: 12/29/2023] Open
Abstract
CD47-SIRPa is a myeloid check point pathway that promotes phagocytosis of cells lacking markers for self-recognition. Tumor cells can overexpress CD47 and bind to SIRPa on macrophages, preventing phagocytosis. CD47 expression is enhanced and correlated with a negative prognosis in acute myeloid leukemia (AML), with its blockade leading to cell clearance. ALX90 is an engineered fusion protein with high affinity for CD47. Composed of the N-terminal D1 domain of SIRPα genetically linked to an inactive Fc domain from human immunoglobulin (Ig) G, ALX90 is designed to avoid potential toxicity of CD47-expressing red blood cells. Venetoclax (VEN) is a specific B-cell lymphoma-2 (BCL-2) inhibitor that can restore apoptosis in malignant cells. In AML, VEN is combined with azanucleosides to induce superior remission rates, however treatment for refractory/relapse is an unmet need. We questioned whether the anti-tumor activity of a VENbased regimen can be augmented through CD47 inhibition (CD47i) in AML and how this triplet may be enhanced. Human AML cell lines were sensitive to ALX90 and its addition increased efficacy of a VEN plus azacitidin (VEN+AZA) regimen in vivo. However, CD47i failed to clear bone marrow tumor burden in PDX models. We hypothesized that the loss of resident macrophages in the bone marrow in AML reduced efficiency of CD47i. Therefore, we attempted to enhance this medullary macrophage population with agonism of TLR3 via polyinosinic:polycytidylic acid (poly(I:C)), which led to expansion and activation of medullary macrophages in in vivo AML PDX models and potentiated CD47i. In summary, the addition of poly(I:C) can enhance medullary macrophage populations to potentiate the phagocytosis merited by therapeutic inhibition of CD47.
Collapse
MESH Headings
- CD47 Antigen/metabolism
- CD47 Antigen/antagonists & inhibitors
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Humans
- Animals
- Mice
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Macrophages/metabolism
- Macrophages/drug effects
- Sulfonamides/pharmacology
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/antagonists & inhibitors
- Antigens, Differentiation/metabolism
- Phagocytosis/drug effects
- Poly I-C/pharmacology
Collapse
Affiliation(s)
- Haley E Ramsey
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN; Program in Cancer Biology
| | - Agnieszka E Gorska
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Brianna N Smith
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Andrew J Monteith
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Londa Fuller
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Maria P Arrate
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Michael R Savona
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN; Center for Immunobiology; Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN.
| |
Collapse
|
16
|
Xiao X, Zheng Y, Wang T, Zhang X, Fang G, Zhang Z, Zhang Z, Zhao J. Enhancing anti-angiogenic immunotherapy for melanoma through injectable metal-organic framework hydrogel co-delivery of combretastatin A4 and poly(I:C). NANOSCALE ADVANCES 2024; 6:3135-3145. [PMID: 38868828 PMCID: PMC11166098 DOI: 10.1039/d4na00079j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024]
Abstract
The interplay between vascularization and macrophage-induced immune suppression plays a crucial role in melanoma treatment. In this study, we propose a novel combination approach to combat melanoma by simultaneously inhibiting tumor vascularization and enhancing macrophage-mediated anti-tumor responses. We investigate the potential of combining combretastatin A4 (CA4), a vascular-disrupting agent, with poly(I:C) (PIC), an immunostimulatory adjuvant. This combination approach effectively suppresses melanoma cell proliferation, disrupts vascularization, and promotes macrophage polarization towards the M1 phenotype for melanoma suppression. To facilitate efficient co-delivery of CA4 and PIC for enhanced anti-angiogenic immunotherapy, we develop an injectable metal-organic framework hydrogel using Zeolitic Imidazolate Framework-8 (ZIF-8) and hyaluronic acid (HA) (ZIF-8/HA). Our findings demonstrate that ZIF-8 enables efficient loading of CA4 and enhances the stability of PIC against RNAase degradation in vitro. Furthermore, the developed co-delivery hydrogel system, PIC/CA4@ZIF-8/HA, exhibits improved rheological properties, good injectability and prolonged drug retention. Importantly, in vivo experiments demonstrate that the PIC/CA4@ZIF-8/HA formulation significantly reduces the dosage and administration frequency while achieving a more pronounced therapeutic effect. It effectively inhibits melanoma growth by suppressing angiogenesis, destroying blood vessels, promoting M1 macrophage infiltration, and demonstrating excellent biocompatibility. In conclusion, our study advances anti-angiogenic immunotherapy for melanoma through the potent combination of PIC/CA4, particularly when administered using the PIC/CA4@ZIF-8/HA formulation. These findings provide a new perspective on clinical anti-angiogenic immunotherapy for melanoma, emphasizing the importance of targeting tumor vascularization and macrophage-mediated immune suppression simultaneously.
Collapse
Affiliation(s)
- Xufeng Xiao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Yunuo Zheng
- Department of Obstetrics and Gynecology, Xuzhou Central Hospital Xuzhou 221009 Jiangsu China
| | - Tianlong Wang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Xiaoqing Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Gaochuan Fang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Zhonghai Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| | - Zhengkui Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University Xuzhou 221002 Jiangsu China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University Xuzhou 221002 Jiangsu China
| | - Jiaojiao Zhao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu International Joint Center of Genomics, School of Life Sciences, Jiangsu Normal University Xuzhou 221116 Jiangsu China
| |
Collapse
|
17
|
Soufihasanabad S, Mahmoudi M, Taghavi-Farahabadi M, Mirsanei Z, Mahmoudi Lamouki R, Mirza Abdalla JK, Babaei E, Hashemi SM. In vivo polarization of M2 macrophages by mesenchymal stem cell-derived extracellular vesicles: A novel approach to macrophage polarization and its potential in treating inflammatory diseases. Med Hypotheses 2024; 187:111353. [DOI: 10.1016/j.mehy.2024.111353] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
18
|
Zhang S, Liu Y, Zhang XL, Sun Y, Lu ZH. ANKRD22 aggravates sepsis-induced ARDS and promotes pulmonary M1 macrophage polarization. J Transl Autoimmun 2024; 8:100228. [PMID: 38225946 PMCID: PMC10788270 DOI: 10.1016/j.jtauto.2023.100228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 01/17/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is independently associated with a poor prognosis in patients with sepsis. Macrophage M1 polarization plays an instrumental role in this process. Therefore, the exploration of key molecules affecting acute lung injury and macrophage M1 polarization may provide therapeutic targets for the treatment of septic ARDS. Here, we identified that elevated levels of Ankyrin repeat domain-containing protein 22 (ANKRD22) were associated with poor prognosis and more pronounced M1 macrophage polarization in septic patients by analyzing high-throughput data. ANKRD22 expression was also significantly upregulated in the alveolar lavage fluid, peripheral blood, and lung tissue of septic ARDS model mice. Knockdown of ANKRD22 significantly attenuated acute lung injury in mice with sepsis-induced ARDS and reduced the M1 polarization of lung macrophages. Furthermore, deletion of ANKRD22 in macrophages inhibited M1 macrophage polarization and reduced levels of phosphorylated IRF3 and intracellular interferon regulatory factor 3 (IRF3) expression, while re-expression of ANKRD22 reversed these changes. Further experiments revealed that ANKRD22 promotes IRF3 activation by binding to mitochondrial antiviral-signaling protein (MAVS). In conclusion, these findings suggest that ANKRD22 promotes the M1 polarization of lung macrophages and exacerbates sepsis-induced ARDS.
Collapse
Affiliation(s)
- Shi Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, ZhongdaHospital, Southeast University, Nanjing, Jiangsu, China
- Department of Pulmonary and Critical Care Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yao Liu
- Emergency Department of Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Gulou District, Nanjing, China
| | - Xiao-Long Zhang
- Department of Ultrasound, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yun Sun
- The First Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province, 230601, China
| | - Zhong-Hua Lu
- The First Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province, 230601, China
| |
Collapse
|
19
|
Qin S, Xie B, Wang Q, Yang R, Sun J, Hu C, Liu S, Tao Y, Xiao D. New insights into immune cells in cancer immunotherapy: from epigenetic modification, metabolic modulation to cell communication. MedComm (Beijing) 2024; 5:e551. [PMID: 38783893 PMCID: PMC11112485 DOI: 10.1002/mco2.551] [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: 10/16/2023] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 05/25/2024] Open
Abstract
Cancer is one of the leading causes of death worldwide, and more effective ways of attacking cancer are being sought. Cancer immunotherapy is a new and effective therapeutic method after surgery, radiotherapy, chemotherapy, and targeted therapy. Cancer immunotherapy aims to kill tumor cells by stimulating or rebuilding the body's immune system, with specific efficiency and high safety. However, only few tumor patients respond to immunotherapy and due to the complex and variable characters of cancer immune escape, the behavior and regulatory mechanisms of immune cells need to be deeply explored from more dimensions. Epigenetic modifications, metabolic modulation, and cell-to-cell communication are key factors in immune cell adaptation and response to the complex tumor microenvironment. They collectively determine the state and function of immune cells through modulating gene expression, changing in energy and nutrient demands. In addition, immune cells engage in complex communication networks with other immune components, which are mediated by exosomes, cytokines, and chemokines, and are pivotal in shaping the tumor progression and therapeutic response. Understanding the interactions and combined effects of such multidimensions mechanisms in immune cell modulation is important for revealing the mechanisms of immunotherapy failure and developing new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Sha Qin
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Bin Xie
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Qingyi Wang
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Rui Yang
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Jingyue Sun
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Chaotao Hu
- Regenerative Medicine, Medical SchoolUniversity of Chinese Academy of SciencesBeijingChina
| | - Shuang Liu
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha, Hunan, China. UniversityChangshaHunanChina
| | - Yongguang Tao
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South universityChangshaHunanChina
| | - Desheng Xiao
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic Medical ScienceXiangya School of MedicineCentral South UniversityChangshaHunanChina
| |
Collapse
|
20
|
Li S, Sheng J, Zhang D, Qin H. Targeting tumor-associated macrophages to reverse antitumor drug resistance. Aging (Albany NY) 2024; 16:10165-10196. [PMID: 38787372 PMCID: PMC11210230 DOI: 10.18632/aging.205858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
Currently, antitumor drugs show limited clinical outcomes, mainly due to adaptive resistance. Clinical evidence has highlighted the importance of the tumor microenvironment (TME) and tumor-associated macrophages (TAMs) in tumor response to conventional antitumor drugs. Preclinical studies show that TAMs following antitumor agent can be reprogrammed to an immunosuppressive phenotype and proangiogenic activities through different mechanisms, mediating drug resistance and poor prognosis. Potential extrinsic inhibitors targeting TAMs repolarize to an M1-like phenotype or downregulate proangiogenic function, enhancing therapeutic efficacy of anti-tumor therapy. Moreover, pharmacological modulation of macrophages that restore the immune stimulatory characteristics is useful to reshaping the tumor microenvironment, thus further limiting tumor growth. This review aims to introduce macrophage response in tumor therapy and provide a potential therapeutic combination strategy of TAM-targeting immunomodulation with conventional antitumor drugs.
Collapse
Affiliation(s)
- Sheng Li
- The Second Hospital of Jilin University, Changchun, China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China
| | - Dan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China
| | - Hanjiao Qin
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Liu D, He W, Yang LL. Revitalizing antitumor immunity: Leveraging nucleic acid sensors as therapeutic targets. Cancer Lett 2024; 588:216729. [PMID: 38387757 DOI: 10.1016/j.canlet.2024.216729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/30/2024] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
Nucleic acid sensors play a critical role in recognizing and responding to pathogenic nucleic acids as danger signals. Upon activation, these sensors initiate downstream signaling cascades that lead to the production and release of pro-inflammatory cytokines, chemokines, and type I interferons. These immune mediators orchestrate diverse effector responses, including the activation of immune cells and the modulation of the tumor microenvironment. However, careful consideration must be given to balancing the activation of nucleic acid sensors to avoid unwanted autoimmune or inflammatory responses. In this review, we provide an overview of nucleic acid sensors and their role in combating cancer through the perception of various aberrant nucleic acids and activation of the immune system. We discuss the connections between different programmed cell death modes and nucleic acid sensors. Finally, we outline the development of nucleic acid sensor agonists, highlighting how their potential as therapeutic targets opens up new avenues for cancer immunotherapy.
Collapse
Affiliation(s)
- Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
22
|
Yuan H, Gui H, Chen S, Zhu L, Wang C, Jing Q, Lv H, Wan Q, Wang S, Zhou S, Ren X, Nie Y, Li L. Regulating Tumor-Associated Macrophage Polarization by Cyclodextrin-Modified PLGA Nanoparticles Loaded with R848 for Treating Colon Cancer. Int J Nanomedicine 2024; 19:3589-3605. [PMID: 38645464 PMCID: PMC11032718 DOI: 10.2147/ijn.s450205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/10/2024] [Indexed: 04/23/2024] Open
Abstract
Purpose This study aimed to develop a novel and feasible modification strategy to improve the solubility and antitumor activity of resiquimod (R848) by utilizing the supramolecular effect of 2-hydroxypropyl-beta-cyclodextrin (2-HP-β-CD). Methods R848-loaded PLGA nanoparticles modified with 2-HP-β-CD (CD@R848@NPs) were synthesized using an enhanced emulsification solvent-evaporation technique. The nanoparticles were then characterized in vitro by several methods, such as scanning electron microscopy (SEM), differential scanning calorimetry (DSC), Fourier transform infrared (FTIR) spectroscopy, particle size analysis, and zeta potential analysis. Then, the nanoparticles were loaded with IR-780 dye and imaged using an in vivo imaging device to evaluate their biodistribution. Additionally, the antitumor efficacy and underlying mechanism of CD@R848@NPs in combination with an anti-TNFR2 antibody were investigated using an MC-38 colon adenocarcinoma model in vivo. Results The average size of the CD@R848@NPs was 376 ± 30 nm, and the surface charge was 21 ± 1 mV. Through this design, the targeting ability of 2-HP-β-CD can be leveraged and R848 is delivered to tumor-supporting M2-like macrophages in an efficient and specific manner. Moreover, we used an anti-TNFR2 antibody to reduce the proportion of Tregs. Compared with plain PLGA nanoparticles or R848, CD@R848@NPs increased penetration in tumor tissues, dramatically reprogrammed M1-like macrophages, removed tumors and prolonged patient survival. Conclusion The new nanocapsule system is a promising strategy for targeting tumor, reprogramming tumor -associated macrophages, and enhancement immunotherapy.
Collapse
Affiliation(s)
- Haohua Yuan
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| | - Huan Gui
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| | - Shuanghui Chen
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| | - Lan Zhu
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| | - Chenglv Wang
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| | - Qianyu Jing
- School of Preclinical Medicine of Zunyi Medical University, Zunyi, Guizhou Province, 563000, People’s Republic of China
| | - Hang Lv
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| | - Quan Wan
- School of Preclinical Medicine of Zunyi Medical University, Zunyi, Guizhou Province, 563000, People’s Republic of China
| | - Shuyi Wang
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| | - Shengwen Zhou
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| | - Xiaodong Ren
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| | - Yingjie Nie
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People’s Hospital, Guiyang, Guizhou Province, 550002, People’s Republic of China
| | - Linzhao Li
- Medical College, Guizhou University, Guizhou Province, 550025, People’s Republic of China
| |
Collapse
|
23
|
Sarkar B, Arlauckas SP, Cuccarese MF, Garris CS, Weissleder R, Rodell CB. Host-functionalization of macrin nanoparticles to enable drug loading and control tumor-associated macrophage phenotype. Front Immunol 2024; 15:1331480. [PMID: 38545103 PMCID: PMC10965546 DOI: 10.3389/fimmu.2024.1331480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
Macrophages are critical regulators of the tumor microenvironment and often present an immuno-suppressive phenotype, supporting tumor growth and immune evasion. Promoting a robust pro-inflammatory macrophage phenotype has emerged as a therapeutic modality that supports tumor clearance, including through synergy with immune checkpoint therapies. Polyglucose nanoparticles (macrins), which possess high macrophage affinity, are useful vehicles for delivering drugs to macrophages, potentially altering their phenotype. Here, we examine the potential of functionalized macrins, synthesized by crosslinking carboxymethyl dextran with L-lysine, as effective carriers of immuno-stimulatory drugs to tumor-associated macrophages (TAMs). Azide groups incorporated during particle synthesis provided a handle for click-coupling of propargyl-modified β-cyclodextrin to macrins under mild conditions. Fluorescence-based competitive binding assays revealed the ability of β-cyclodextrin to non-covalently bind to hydrophobic immuno-stimulatory drug candidates (Keq ~ 103 M-1), enabling drug loading within nanoparticles. Furthermore, transcriptional profiles of macrophages indicated robust pro-inflammatory reprogramming (elevated Nos2 and Il12; suppressed Arg1 and Mrc1 expression levels) for a subset of these immuno-stimulatory agents (UNC2025 and R848). Loading of R848 into the modified macrins improved the drug's effect on primary murine macrophages by three-fold in vitro. Intravital microscopy in IL-12-eYFP reporter mice (24 h post-injection) revealed a two-fold enhancement in mean YFP fluorescence intensity in macrophages targeted with R848-loaded macrins, relative to vehicle controls, validating the desired pro-inflammatory reprogramming of TAMs in vivo by cell-targeted drug delivery. Finally, in an intradermal MC38 tumor model, cyclodextrin-modified macrin NPs loaded with immunostimulatory drugs significantly reduced tumor growth. Therefore, efficient and effective repolarization of tumor-associated macrophages to an M1-like phenotype-via drug-loaded macrins-inhibits tumor growth and may be useful as an adjuvant to existing immune checkpoint therapies.
Collapse
Affiliation(s)
- Biplab Sarkar
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Sean P. Arlauckas
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
| | - Michael F. Cuccarese
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
| | - Christopher S. Garris
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
- Department of Pathology, Harvard Medical School, Boston, MA, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Christopher B. Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
24
|
Anfray C, Varela CF, Ummarino A, Maeda A, Sironi M, Gandoy S, Brea J, Loza MI, León S, Calvo A, Correa J, Fernandez-Megia E, Alonso MJ, Allavena P, Crecente-Campo J, Andón FT. Polymeric nanocapsules loaded with poly(I:C) and resiquimod to reprogram tumor-associated macrophages for the treatment of solid tumors. Front Immunol 2024; 14:1334800. [PMID: 38259462 PMCID: PMC10800412 DOI: 10.3389/fimmu.2023.1334800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Background In the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play a key immunosuppressive role that limits the ability of the immune system to fight cancer. Toll-like receptors (TLRs) ligands, such as poly(I:C) or resiquimod (R848) are able to reprogram TAMs towards M1-like antitumor effector cells. The objective of our work has been to develop and evaluate polymeric nanocapsules (NCs) loaded with poly(I:C)+R848, to improve drug stability and systemic toxicity, and evaluate their targeting and therapeutic activity towards TAMs in the TME of solid tumors. Methods NCs were developed by the solvent displacement and layer-by-layer methodologies and characterized by dynamic light scattering and nanoparticle tracking analysis. Hyaluronic acid (HA) was chemically functionalized with mannose for the coating of the NCs to target TAMs. NCs loaded with TLR ligands were evaluated in vitro for toxicity and immunostimulatory activity by Alamar Blue, ELISA and flow cytometry, using primary human monocyte-derived macrophages. For in vivo experiments, the CMT167 lung cancer model and the MN/MCA1 fibrosarcoma model metastasizing to lungs were used; tumor-infiltrating leukocytes were evaluated by flow cytometry and multispectral immunophenotyping. Results We have developed polymeric NCs loaded with poly(I:C)+R848. Among a series of 5 lead prototypes, protamine-NCs were selected based on their physicochemical properties (size, charge, stability) and in vitro characterization, showing good biocompatibility on primary macrophages and ability to stimulate their production of T-cell attracting chemokines (CXCL10, CCL5) and to induce M1-like macrophages cytotoxicity towards tumor cells. In mouse tumor models, the intratumoral injection of poly(I:C)+R848-protamine-NCs significantly prevented tumor growth and lung metastasis. In an orthotopic murine lung cancer model, the intravenous administration of poly(I:C)+R848-prot-NCs, coated with an additional layer of HA-mannose to improve TAM-targeting, resulted in good antitumoral efficacy with no apparent systemic toxicity. While no significant alterations were observed in T cell numbers (CD8, CD4 or Treg), TAM-reprogramming in treated mice was confirmed by the relative decrease of interstitial versus alveolar macrophages, having higher CD86 expression but lower CD206 and Arg1 expression in the same cells, in treated mice. Conclusion Mannose-HA-protamine-NCs loaded with poly(I:C)+R848 successfully reprogram TAMs in vivo, and reduce tumor progression and metastasis spread in mouse tumors.
Collapse
Affiliation(s)
- Clément Anfray
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Carmen Fernández Varela
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Aldo Ummarino
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Akihiro Maeda
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Marina Sironi
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Sara Gandoy
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- BioFarma Research Group, CIMUS, Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Jose Brea
- BioFarma Research Group, CIMUS, Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - María Isabel Loza
- BioFarma Research Group, CIMUS, Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Sergio León
- Navarra Institute for Health Research (IdiSNA), Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Alfonso Calvo
- Navarra Institute for Health Research (IdiSNA), Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Juan Correa
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Eduardo Fernandez-Megia
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Paola Allavena
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - José Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Fernando Torres Andón
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Oncology Department, Complexo Hospitalario de A Coruña (CHUAC), A Coruña, Spain
| |
Collapse
|
25
|
Li X, Hattori S, Ebara M, Shirahata N, Hanagata N. A facile approach to preparing personalized cancer vaccines using iron-based metal organic framework. Front Immunol 2024; 14:1328379. [PMID: 38259474 PMCID: PMC10800499 DOI: 10.3389/fimmu.2023.1328379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Background Considering the diversity of tumors, it is of great significance to develop a simple, effective, and low-cost method to prepare personalized cancer vaccines. Methods In this study, a facile one-pot synthetic route was developed to prepare cancer vaccines using model antigen or autologous tumor antigens based on the coordination interaction between Fe3+ ions and endogenous fumarate ligands. Results Herein, Fe-based metal organic framework can effectively encapsulate tumor antigens with high loading efficiency more than 80%, and act as both delivery system and adjuvants for tumor antigens. By adjusting the synthesis parameters, the obtained cancer vaccines are easily tailored from microscale rod-like morphology with lengths of about 0.8 μm (OVA-ML) to nanoscale morphology with sizes of about 50~80 nm (OVA-MS). When cocultured with antigen-presenting cells, nanoscale cancer vaccines more effectively enhance antigen uptake and Th1 cytokine secretion than microscale ones. Nanoscale cancer vaccines (OVA-MS, dLLC-MS) more effectively enhance lymph node targeting and cross-presentation of tumor antigens, mount antitumor immunity, and inhibit the growth of established tumor in tumor-bearing mice, compared with microscale cancer vaccines (OVA-ML, dLLC-ML) and free tumor antigens. Conclusions Our work paves the ways for a facile, rapid, and low-cost preparation approach for personalized cancer vaccines.
Collapse
Affiliation(s)
- Xia Li
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki, Japan
| | - Shinya Hattori
- Bioanalysis Unit, Research Network and Facility Services Division, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki, Japan
| | - Mitsuhiro Ebara
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki, Japan
| | - Naoto Shirahata
- Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Tsukuba, Ibaraki, Japan
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo, Japan
| | - Nobutaka Hanagata
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki, Japan
| |
Collapse
|
26
|
Gu J, Wang J, Sun Y, Mao X, Qian C, Tang X, Wang J, Xie H, Ling L, Zhao Y, Liu X, Zhang K, Pan H, Wang S, Wang C, Zhou W. Immune cells within tertiary lymphoid structures are associated with progression-free survival in patients with locoregional recurrent breast cancer. Cancer Med 2024; 13:e6864. [PMID: 38133211 PMCID: PMC10807640 DOI: 10.1002/cam4.6864] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
INTRODUCTION Locoregional recurrent breast cancers have a poor prognosis. Little is known about the prognostic impact of immune microenvironment, and tertiary lymphoid structures (TLSs) in particular have not been reported. Thus, we aimed to characterize the immune microenvironment in locoregional recurrent breast tumors and to investigate its relationship with prognosis. METHODS We retrospectively included 112 patients with locoregional recurrent breast cancer, and hematoxylin-eosin staining and immunohistochemical staining (CD3, CD4, CD8, CD19, CD38, and CD68) were performed on locoregional recurrent tumor samples. The association of immune cells and TLSs with progression-free survival (PFS) were analyzed by survival analysis. RESULTS We found more immune cells in the peritumor than stroma. After grouping according to estrogen receptor (ER) status, a low level of peritumoral CD3+ cells in ER+ subgroup (p = 0.015) and a low level of stromal CD68+ cells in ER- subgroup (p = 0.047) were both associated with longer PFS. TLSs were present in 68% of recurrent tumors, and CD68+ cells within TLSs were significantly associated with PFS as an independent prognostic factor (p = 0.035). TLSs and immune cells (CD3, CD38, and CD68) within TLSs were associated with longer PFS in ER- recurrent tumors (p = 0.044, p = 0.012, p = 0.050, p < 0.001, respectively), whereas CD38+ cells within TLSs were associated with shorter PFS in ER+ recurrent tumors (p = 0.037). CONCLUSION Our study proposes potential predictors for the clinical prognosis of patients with locoregional recurrent breast cancer, emphasizing the prognostic value of immune cells within TLSs, especially CD68+ cells.
Collapse
Affiliation(s)
- Jinyuan Gu
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Jiaming Wang
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Yue Sun
- Department of OncologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Xinrui Mao
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Chao Qian
- Department of General SurgerySir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Xinyu Tang
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Ji Wang
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Hui Xie
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Lijun Ling
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Yi Zhao
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Xiaoan Liu
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Kai Zhang
- Pancreas Center & Department of General SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsuChina
- Pancreas Institute of Nanjing Medical UniversityNanjingJiangsuChina
| | - Hong Pan
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Shui Wang
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Cong Wang
- Department of PathologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Wenbin Zhou
- Department of Breast SurgeryThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center For Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
| |
Collapse
|
27
|
Liu L, Chen G, Gong S, Huang R, Fan C. Targeting tumor-associated macrophage: an adjuvant strategy for lung cancer therapy. Front Immunol 2023; 14:1274547. [PMID: 38022518 PMCID: PMC10679371 DOI: 10.3389/fimmu.2023.1274547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
The emergence of immunotherapy has revolutionized the treatment landscape for various types of cancer. Nevertheless, lung cancer remains one of the leading causes of cancer-related mortality worldwide due to the development of resistance in most patients. As one of the most abundant groups of immune cells in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play crucial and complex roles in the development of lung cancer, including the regulation of immunosuppressive TME remodeling, metabolic reprogramming, neoangiogenesis, metastasis, and promotion of tumoral neurogenesis. Hence, relevant strategies for lung cancer therapy, such as inhibition of macrophage recruitment, TAM reprograming, depletion of TAMs, and engineering of TAMs for drug delivery, have been developed. Based on the satisfactory treatment effect of TAM-targeted therapy, recent studies also investigated its synergistic effect with current therapies for lung cancer, including immunotherapy, radiotherapy, chemotherapy, anti-epidermal growth factor receptor (anti-EGFR) treatment, or photodynamic therapy. Thus, in this article, we summarized the key mechanisms of TAMs contributing to lung cancer progression and elaborated on the novel therapeutic strategies against TAMs. We also discussed the therapeutic potential of TAM targeting as adjuvant therapy in the current treatment of lung cancer, particularly highlighting the TAM-centered strategies for improving the efficacy of anti-programmed cell death-1/programmed cell death-ligand 1 (anti-PD-1/PD-L1) treatment.
Collapse
Affiliation(s)
| | | | | | | | - Chunmei Fan
- *Correspondence: Chunmei Fan, ; Rongfu Huang,
| |
Collapse
|
28
|
Tian L, Tan Z, Yang Y, Liu S, Yang Q, Tu Y, Chen J, Guan H, Fan L, Yu B, Chen X, Hu Y. In situ sprayed hydrogels containing resiquimod-loaded liposomes reduce chronic osteomyelitis recurrence by intracellular bacteria clearance. Acta Biomater 2023; 169:209-227. [PMID: 37516419 DOI: 10.1016/j.actbio.2023.07.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 07/31/2023]
Abstract
At present, surgical debridement and systematic administration of antibiotics represent the mainstay of treatment for chronic osteomyelitis. However, it is now understood that Staphylococcus aureus (S. aureus) can survive within excessively polarized M2 macrophages and evade antibiotics, accounting for the high recurrence of chronic osteomyelitis. Effective treatments for intracellular infection have rarely been reported. Herein, we designed an in situ sprayed liposomes hydrogels spray with macrophage-targeted effects and the ability to reverse polarization and eradicate intracellular bacteria to reduce the recurrence of osteomyelitis. Resiquimod (R848)-loaded and phosphatidylserine (PS)-coating nanoliposomes were introduced into fibrinogen and thrombin to form the PSL-R848@Fibrin spray. Characterization and phagocytosis experiments were performed to confirm the successful preparation of the PSL-R848@Fibrin spray. Meanwhile, in vitro cell experiments validated its ability to eliminate intracellular S. aureus by reprogramming macrophages from the M2 to the M1 phenotype. Additionally, we established a chronic osteomyelitis rat model to simulate the treatment and recurrence process. Histological analysis demonstrated a significant increase in M1 macrophages and the elimination of intracellular bacteria. Imaging revealed a significant decrease in osteomyelitis recurrence. Overall, the liposome hydrogels could target macrophages to promote antibacterial properties against intracellular infection and reduce the recurrence of chronic osteomyelitis, providing the foothold for improving the outcomes of this patient population. STATEMENT OF SIGNIFICANCE: Chronic osteomyelitis remains a high recurrence although undergoing traditional treatment of debridement and antibiotics. S. aureus can survive within the excessively polarized M2 macrophages to evade the effects of antibiotics. However, few studies have sought to investigate effective intracellular bacteria eradication. Herein, we designed a macrophage-targeted R848-containing liposomes fibrin hydrogels spray (PSL-R848@Fibrin) that can reprogram polarization of macrophages and eradicate intracellular bacteria for osteomyelitis treatment. With great properties of rapid gelation, strong adhesion, high flexibility and fit-to-shape capacity, the facile-operated immunotherapeutic in-situ-spray fibrin hydrogels exhibited huge promise of reversing polarization and fighting intracellular infections. Importantly, we revealed a hitherto undocumented treatment strategy for reducing the recurrence of chronic osteomyelitis and potentially improving the prognosis of chronic osteomyelitis patients.
Collapse
Affiliation(s)
- Liangjie Tian
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Zilin Tan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Yusheng Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Shencai Liu
- Division of Orthopaedics Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Qingfeng Yang
- Division of Orthopaedics Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Yuesheng Tu
- Division of Orthopaedics Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Jialan Chen
- Division of Orthopaedics Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Hongye Guan
- Department of Orthopedic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Lei Fan
- Division of Orthopaedics Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China.
| | - Xianhui Chen
- Department of Orthopedic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong 528000, China.
| | - Yanjun Hu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
29
|
Tang M, Chen B, Xia H, Pan M, Zhao R, Zhou J, Yin Q, Wan F, Yan Y, Fu C, Zhong L, Zhang Q, Wang Y. pH-gated nanoparticles selectively regulate lysosomal function of tumour-associated macrophages for cancer immunotherapy. Nat Commun 2023; 14:5888. [PMID: 37735462 PMCID: PMC10514266 DOI: 10.1038/s41467-023-41592-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
Tumour-associated macrophages (TAMs), as one of the most abundant tumour-infiltrating immune cells, play a pivotal role in tumour antigen clearance and immune suppression. M2-like TAMs present a heightened lysosomal acidity and protease activity, limiting an effective antigen cross-presentation. How to selectively reprogram M2-like TAMs to reinvigorate anti-tumour immune responses is challenging. Here, we report a pH-gated nanoadjuvant (PGN) that selectively targets the lysosomes of M2-like TAMs in tumours rather than the corresponding organelles from macrophages in healthy tissues. Enabled by the PGN nanotechnology, M2-like TAMs are specifically switched to a M1-like phenotype with attenuated lysosomal acidity and cathepsin activity for improved antigen cross-presentation, thus eliciting adaptive immune response and sustained tumour regression in tumour-bearing female mice. Our findings provide insights into how to specifically regulate lysosomal function of TAMs for efficient cancer immunotherapy.
Collapse
Affiliation(s)
- Mingmei Tang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Meijie Pan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ruiyang Zhao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jiayi Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Qingqing Yin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Fangjie Wan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yue Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Chuanxun Fu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Lijun Zhong
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- Chemical Biology Center, Peking University, Beijing, China.
| |
Collapse
|
30
|
Ward J, Martin P. Live-imaging studies reveal how microclots and the associated inflammatory response enhance cancer cell extravasation. J Cell Sci 2023; 136:jcs261225. [PMID: 37671502 PMCID: PMC10561694 DOI: 10.1242/jcs.261225] [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: 04/11/2023] [Accepted: 08/31/2023] [Indexed: 09/07/2023] Open
Abstract
Previous clinical studies and work in mouse models have indicated that platelets and microclots might enable the recruitment of immune cells to the pre-metastatic cancer niche, leading to efficacious extravasation of cancer cells through the vessel wall. Here, we investigated the interaction between platelets, endothelial cells, inflammatory cells, and engrafted human and zebrafish cancer cells by live-imaging studies in translucent zebrafish larvae, and show how clotting (and clot resolution) act as foci and as triggers for extravasation. Fluorescent tagging in each lineage revealed their dynamic behaviour and potential roles in these events, and we tested function by genetic and drug knockdown of the contributing players. Morpholino knockdown of fibrinogen subunit α (fga) and warfarin treatment to inhibit clotting both abrogated extravasation of cancer cells. The inflammatory phenotype appeared fundamental, and we show that forcing a pro-inflammatory, tnfa-positive phenotype is inhibitory to extravasation of cancer cells.
Collapse
Affiliation(s)
- Juma Ward
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
31
|
Exposito F, Redrado M, Houry M, Hastings K, Molero-Abraham M, Lozano T, Solorzano JL, Sanz-Ortega J, Adradas V, Amat R, Redin E, Leon S, Legarra N, Garcia J, Serrano D, Valencia K, Robles-Oteiza C, Foggetti G, Otegui N, Felip E, Lasarte JJ, Paz-Ares L, Zugazagoitia J, Politi K, Montuenga L, Calvo A. PTEN Loss Confers Resistance to Anti-PD-1 Therapy in Non-Small Cell Lung Cancer by Increasing Tumor Infiltration of Regulatory T Cells. Cancer Res 2023; 83:2513-2526. [PMID: 37311042 DOI: 10.1158/0008-5472.can-22-3023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 04/11/2023] [Accepted: 06/08/2023] [Indexed: 06/15/2023]
Abstract
Immunotherapy resistance in non-small cell lung cancer (NSCLC) may be mediated by an immunosuppressive microenvironment, which can be shaped by the mutational landscape of the tumor. Here, we observed genetic alterations in the PTEN/PI3K/AKT/mTOR pathway and/or loss of PTEN expression in >25% of patients with NSCLC, with higher frequency in lung squamous carcinomas (LUSC). Patients with PTEN-low tumors had higher levels of PD-L1 and PD-L2 and showed worse progression-free survival when treated with immunotherapy. Development of a Pten-null LUSC mouse model revealed that tumors with PTEN loss were refractory to antiprogrammed cell death protein 1 (anti-PD-1), highly metastatic and fibrotic, and secreted TGFβ/CXCL10 to promote conversion of CD4+ lymphocytes into regulatory T cells (Treg). Human and mouse PTEN-low tumors were enriched in Tregs and expressed higher levels of immunosuppressive genes. Importantly, treatment of mice bearing Pten-null tumors with TLR agonists and anti-TGFβ antibody aimed to alter this immunosuppressive microenvironment and led to tumor rejection and immunologic memory in 100% of mice. These results demonstrate that lack of PTEN causes immunotherapy resistance in LUSCs by establishing an immunosuppressive tumor microenvironment that can be reversed therapeutically. SIGNIFICANCE PTEN loss leads to the development of an immunosuppressive microenvironment in lung cancer that confers resistance to anti-PD-1 therapy, which can be overcome by targeting PTEN loss-mediated immunosuppression.
Collapse
Affiliation(s)
- Francisco Exposito
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Miriam Redrado
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IDISNA, Pamplona, Spain
| | - Maeva Houry
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Katherine Hastings
- Yale Cancer Center, New Haven, Connecticut
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
- Department of Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, Connecticut
| | - Magdalena Molero-Abraham
- Department of Medical Oncology and Tumor Microenvironment and Immunotherapy Research Group, 12 de Octubre Hospital, Madrid, Spain
| | - Teresa Lozano
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Jose Luis Solorzano
- Anatomic Pathology and Molecular Diagnostics, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Julian Sanz-Ortega
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Vera Adradas
- Department of Medical Oncology and Tumor Microenvironment and Immunotherapy Research Group, 12 de Octubre Hospital, Madrid, Spain
| | - Ramon Amat
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Esther Redin
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Sergio Leon
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Naroa Legarra
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Javier Garcia
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Diego Serrano
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IDISNA, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Karmele Valencia
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
| | | | - Giorgia Foggetti
- Yale Cancer Center, New Haven, Connecticut
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
- Department of Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, Connecticut
| | - Nerea Otegui
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Enriqueta Felip
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Juan J Lasarte
- IDISNA, Pamplona, Spain
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Luis Paz-Ares
- CIBERONC, ISCIII, Madrid, Spain
- Department of Medical Oncology and Tumor Microenvironment and Immunotherapy Research Group, 12 de Octubre Hospital, Madrid, Spain
| | - Jon Zugazagoitia
- CIBERONC, ISCIII, Madrid, Spain
- Department of Medical Oncology and Tumor Microenvironment and Immunotherapy Research Group, 12 de Octubre Hospital, Madrid, Spain
| | - Katerina Politi
- Yale Cancer Center, New Haven, Connecticut
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
- Department of Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, Connecticut
| | - Luis Montuenga
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Alfonso Calvo
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- CIBERONC, ISCIII, Madrid, Spain
- IDISNA, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| |
Collapse
|
32
|
Zhang YY, Li J, Li F, Xue S, Xu QY, Zhang YQ, Feng L. Palmitic acid combined with γ-interferon inhibits gastric cancer progression by modulating tumor-associated macrophages' polarization via the TLR4 pathway. J Cancer Res Clin Oncol 2023; 149:7053-7067. [PMID: 36862159 DOI: 10.1007/s00432-023-04655-9] [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: 01/17/2023] [Accepted: 02/14/2023] [Indexed: 03/03/2023]
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) constitute the main infiltrating immune cells in the solid tumor microenvironment. Amounting studies have analyzed the antitumor effect on immune response induced by Toll-like receptor (TLR) agonists, such as lipopolysaccharide (LPS), γ-interferon (γ-IFN), and palmitic Acid (PA). However, their combined treatment for gastric cancer (GC) has not been illuminated. METHODS We investigated the relevance of macrophage polarization and the effect of PA and γ-IFN in GC in vitro and in vivo. M1 and M2 macrophage-associated markers were measured by real-time quantitative PCR and flow cytometry, and the activation level of the TLR4 signaling pathways was evaluated by western blot analysis. The effect of PA and γ-IFN on the proliferation, migration, and invasion of GC cells (GCCs) was evaluated by Cell-Counting Kit-8, transwell assays, and wound-healing assays. In vivo animal models were used to verify the effect of PA and γ-IFN on tumor progression, and the M1 and M2 macrophage markers, CD8 + T lymphocytes, regulatory T cells (Treg) cells, and the myeloid-derived suppressor cells (MDSCs) in tumor tissues were analyzed by flow cytometry and immunohistochemical (IHC). RESULTS The results showed that this combination strategy enhanced M1-like macrophages and diminished M2-like macrophages through the TLR4 signaling pathway in vitro. In addition, the combination strategy impairs the proliferative and migratory activity of GCC in vitro and in vivo. While, the antitumor effect was abolished using the TAK-424 (a specific TLR-4 signaling pathway inhibitor) in vitro. CONCLUSIONS The combined treatment of PA and γ-IFN inhibited GC progression by modulating macrophages polarization via the TLR4 pathway.
Collapse
Affiliation(s)
- Yan-Yan Zhang
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Jian Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Fan Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Shuai Xue
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Qing-Yu Xu
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Ya-Qiong Zhang
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China.
| |
Collapse
|
33
|
Jia D, Lu Y, Lv M, Wang F, Lu X, Zhu W, Wei J, Guo W, Liu R, Li G, Wang R, Li J, Yuan F. Targeted co-delivery of resiquimod and a SIRPα variant by liposomes to activate macrophage immune responses for tumor immunotherapy. J Control Release 2023; 360:858-871. [PMID: 37473808 DOI: 10.1016/j.jconrel.2023.07.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/25/2023] [Accepted: 07/18/2023] [Indexed: 07/22/2023]
Abstract
Tumor-associated macrophages (TAMs) are the major immune cells infiltrating the tumor microenvironment (TME) and typically exhibit an immunosuppressive M2-like phenotype, which facilitates tumor growth and promotes resistance to immunotherapy. Additionally, tumor cells tend to express high levels of CD47, a "don't eat me" signal, that obstructs macrophage phagocytosis. Consequently, re-educating TAMs in combination with CD47 blockage is promising to trigger intense macrophage immune responses against tumors. As a toll-like receptor 7/8 agonist, resiquimod (R848) possesses the capacity to re-educate TAMs from M2 type to M1 type. We found that intratumoral administration of R848 synergistically improved the antitumor immunotherapeutic effect of CV1 protein (a SIRPα variant with high antagonism to CD47). However, the poor bioavailability and potential toxicity of this combo strategy remain a challenge. Here, a TAMs-targeted liposome (named: R-LS/M/CV1) co-delivering R848 and CV1 protein was constructed via decorating mannose on the liposomal surface. R-LS/M/CV1 exhibited high abilities of targeting, re-education and pro-phagocytosis of tumor cells to M2 macrophages in vitro. Intratumoral administration of R-LS/M/CV1 remarkedly eliminated tumor burden in the MC38 tumor model via repolarization of TAMs to M1 type, pro-phagocytosis of TAMs against tumors, and recruitment of tumor-infiltrating T cells. More encouragingly, due to the double targeting to TAMs and tumor cells of mannose and CV1 protein, R-LS/M/CV1 effectively accumulated at the tumor site, thereby not only remarkedly inhibiting tumors, but also exerting no hematological and histopathological toxicity when administered systemically. Our integrated strategy based on re-educating TAMs and CD47 blockade provides a promising approach to trigger macrophage immune responses against tumors for immunotherapy.
Collapse
Affiliation(s)
- Dianlong Jia
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Yue Lu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China.
| | - Mingjia Lv
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Feifei Wang
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China
| | - Xiaomeng Lu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Weifan Zhu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Jianmei Wei
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China
| | - Wen Guo
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Renmin Liu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Guangyong Li
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Rui Wang
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Jun Li
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China.
| | - Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China.
| |
Collapse
|
34
|
Fang G, Zhang Z, Jiang B, Zheng Y, Xiao X, Wang T, Zhang Z, Zhao J. Immunologically active ferumoxytol-poly(I : C) nanomaterials inhibit metastatic melanoma by regulating myeloid-derived suppressor cell differentiation. Biomater Sci 2023. [PMID: 37366334 DOI: 10.1039/d3bm00416c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Nanomaterials have been identified as a potential therapeutic option for targeting myeloid-derived suppressor cells (MDSCs), which are known to play a crucial role in tumor metastasis and treatment resistance. Here, we report a unique immunologically active nanomaterial composed of ferumoxytol and poly(I : C) (FP-NPs) and investigate its immunoregulatory activities on MDSCs in metastatic melanoma. In vivo assays demonstrated that FP-NPs had the ability to significantly impede the progression of metastatic melanoma and decrease the MDSC population in the lungs, spleen, and bone marrow of mice. Both in vivo and in vitro experiments revealed that FP-NPs reduced the number of granulocytic MDSCs and promoted the differentiation of monocytic MDSCs into anti-tumor M1 macrophages. Transcriptome sequencing indicated that FP-NPs significantly altered the expression of several genes involved in immunity. Analysis of Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and quantitative real-time PCR revealed that FP-NPs significantly increased the expression of the myeloid cell differentiation-related gene interferon regulatory factor 7 and activated interferon beta-related signaling pathways, which stimulated the differentiation of MDSCs into M1 macrophages. These findings suggest that FP-NPs, a unique nanomaterial with immunological properties, can induce MDSCs to differentiate into M1 macrophages, potentially offering new treatment prospects for metastatic melanoma in the future.
Collapse
Affiliation(s)
- Gaochuan Fang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Zhonghai Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Bo Jiang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
- Department of Urology, Xuzhou Central Hospital, Xuzhou, 221009, China
| | - Yunuo Zheng
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Xufeng Xiao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Tianlong Wang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| | - Zhengkui Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China.
| | - Jiaojiao Zhao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu Province, China.
| |
Collapse
|
35
|
Xiao Z, Wang R, Wang X, Yang H, Dong J, He X, Yang Y, Guo J, Cui J, Zhou Z. Impaired function of dendritic cells within the tumor microenvironment. Front Immunol 2023; 14:1213629. [PMID: 37441069 PMCID: PMC10333501 DOI: 10.3389/fimmu.2023.1213629] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Dendritic cells (DCs), a class of professional antigen-presenting cells, are considered key factors in the initiation and maintenance of anti-tumor immunity due to their powerful ability to present antigen and stimulate T-cell responses. The important role of DCs in controlling tumor growth and mediating potent anti-tumor immunity has been demonstrated in various cancer models. Accordingly, the infiltration of stimulatory DCs positively correlates with the prognosis and response to immunotherapy in a variety of solid tumors. However, accumulating evidence indicates that DCs exhibit a significantly dysfunctional state, ultimately leading to an impaired anti-tumor immune response due to the effects of the immunosuppressive tumor microenvironment (TME). Currently, numerous preclinical and clinical studies are exploring immunotherapeutic strategies to better control tumors by restoring or enhancing the activity of DCs in tumors, such as the popular DC-based vaccines. In this review, an overview of the role of DCs in controlling tumor progression is provided, followed by a summary of the current advances in understanding the mechanisms by which the TME affects the normal function of DCs, and concluding with a brief discussion of current strategies for DC-based tumor immunotherapy.
Collapse
Affiliation(s)
- Zhihua Xiao
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Ruiqi Wang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Xuyan Wang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Haikui Yang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Jiamei Dong
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Xin He
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Yang Yang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Jiahao Guo
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Jiawen Cui
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhiling Zhou
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| |
Collapse
|
36
|
Zhang Y, Yang J, Zhang T, Gu H. Emerging advances in nanobiomaterials-assisted chimeric antigen receptor (CAR)-macrophages for tumor immunotherapy. Front Bioeng Biotechnol 2023; 11:1211687. [PMID: 37388769 PMCID: PMC10301827 DOI: 10.3389/fbioe.2023.1211687] [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: 04/25/2023] [Accepted: 06/06/2023] [Indexed: 07/01/2023] Open
Abstract
Adoptive cell immunotherapy, especially chimeric antigen receptor (CAR)-T-cells therapy, has made great progress in the clinical treatment of hematological malignancies. However, restricted by the complex tumor microenvironment, the potential efficiency of T-cell infiltration and activated immune cells are limited, thus failure prevented the progression of the solid tumor. Alternatively, tumor-associated macrophages (TAMs), one sustentacular and heterogeneous cellular population within the tumor microenvironment, are regarded as potential therapeutic targets. Recently, CARs have shown tremendous promise in treating malignancies by equipping macrophages. This novel therapeutic strategy circumvents the tumor microenvironment's limitations and provides a safer therapeutic approach. Meanwhile, nanobiomaterials as gene delivery carriers not only substantially reduce the treatment cost of this novel therapeutic strategy, but also set the foundation for in vivo CAR-M therapy. Here, we highlight the major strategies prepared for CAR-M, emphasizing the challenges and opportunities of these approaches. First, the common therapeutic strategies for macrophages are summarized in clinical and preclinical trials. Namely, TAM-targeted therapeutic strategies: 1) Inhibit monocyte or macrophage recruitment into tumors, 2) deplete TAMs, and 3) reprogramme TAMs to antitumor M1 phenotype. Second, the current development and progress of CAR-M therapy are reviewed, including the researchers' attempts in CAR structure design, cell origin, and gene delivery vectors, especially nanobiomaterials as an alternative to viral vectors, as well as some challenges faced by current CAR-M therapy are also summarized and discussed. Finally, the field of genetically engineered macrophages integration with nanotechnology for the future in oncology has been prospected.
Collapse
Affiliation(s)
- Yanan Zhang
- Nano Biomedical Research Center, School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jingxing Yang
- Nano Biomedical Research Center, School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | | | - Hongchen Gu
- Nano Biomedical Research Center, School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
37
|
Velasco WV, Khosravi N, Castro-Pando S, Torres-Garza N, Grimaldo MT, Krishna A, Clowers MJ, Umer M, Tariq Amir S, Del Bosque D, Daliri S, De La Garza MM, Ramos-Castaneda M, Evans SE, Moghaddam SJ. Toll-like receptors 2, 4, and 9 modulate promoting effect of COPD-like airway inflammation on K-ras-driven lung cancer through activation of the MyD88/NF-ĸB pathway in the airway epithelium. Front Immunol 2023; 14:1118721. [PMID: 37283745 PMCID: PMC10240392 DOI: 10.3389/fimmu.2023.1118721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/02/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction Toll-like receptors (TLRs) are an extensive group of proteins involved in host defense processes that express themselves upon the increased production of endogenous damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) due to the constant contact that airway epithelium may have with pathogenic foreign antigens. We have previously shown that COPD-like airway inflammation induced by exposure to an aerosolized lysate of nontypeable Haemophilus influenzae (NTHi) promotes tumorigenesis in a K-ras mutant mouse model of lung cancer, CCSPCre/LSL-K-rasG12D (CC-LR) mouse. Methods In the present study, we have dissected the role of TLRs in this process by knocking out TLR2, 4, and 9 and analyzing how these deletions affect the promoting effect of COPD-like airway inflammation on K-ras-driven lung adenocarcinoma. Results We found that knockout of TLR 2, 4, or 9 results in a lower tumor burden, reduced angiogenesis, and tumor cell proliferation, accompanied by increased tumor cell apoptosis and reprogramming of the tumor microenvironment to one that is antitumorigenic. Additionally, knocking out of downstream signaling pathways, MyD88/NF-κB in the airway epithelial cells further recapitulated this initial finding. Discussion Our study expands the current knowledge of the roles that TLR signaling plays in lung cancer, which we hope, can pave the way for more reliable and efficacious prevention and treatment modalities for lung cancer.
Collapse
Affiliation(s)
- Walter V. Velasco
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nasim Khosravi
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Susana Castro-Pando
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nelly Torres-Garza
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
| | - Maria T. Grimaldo
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Avantika Krishna
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Michael J. Clowers
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Misha Umer
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sabah Tariq Amir
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Diana Del Bosque
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Soudabeh Daliri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Miguelina De La Garza
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
| | - Marco Ramos-Castaneda
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
| | - Scott E. Evans
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
38
|
Gilman KE, Matiatos AP, Cracchiolo MJ, Moon AG, Davini DW, Simpson RJ, Katsanis E. Multiagent Intratumoral Immunotherapy Can Be Effective in A20 Lymphoma Clearance and Generation of Systemic T Cell Immunity. Cancers (Basel) 2023; 15:cancers15071951. [PMID: 37046612 PMCID: PMC10093573 DOI: 10.3390/cancers15071951] [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: 02/03/2023] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
The use of immunotherapies has shown promise against selective human cancers. Identifying novel combinations of innate and adaptive immune cell-activating agents that can work synergistically to suppress tumor growth and provide additional protection against resistance or recurrence is critical. The A20 murine lymphoma model was used to evaluate the effect of various combination immunotherapies administered intratumorally. We show that single-modality treatment with Poly(I:C) or GM-CSF-secreting allogeneic cells only modestly controls tumor growth, whereas when given together there is an improved benefit, with 50% of animals clearing tumors and surviving long-term. Neither heat nor irradiation of GM-CSF-secreting cells enhanced the response over use of live cells. The use of a TIM-3 inhibitory antibody and an OX40 agonist in combination with Poly(I:C) allowed for improved tumor control, with 90% of animals clearing tumors with or without a combination of GM-CSF-secreting cells. Across all treatment groups, mice rejecting their primary A20 tumors were immune to subsequent challenge with A20, and this longstanding immunity was T-cell dependent. The results herein support the use of combinations of innate and adaptive immune activating agents for immunotherapy against lymphoma and should be investigated in other cancer types.
Collapse
Affiliation(s)
- Kristy E Gilman
- Department of Pediatrics, University of Arizona, Tucson, AZ 85721, USA
| | - Andrew P Matiatos
- Department of Pediatrics, University of Arizona, Tucson, AZ 85721, USA
| | | | - Amanda G Moon
- Department of Cell and Molecular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Dan W Davini
- Department of Pediatrics, University of Arizona, Tucson, AZ 85721, USA
| | - Richard J Simpson
- Department of Pediatrics, University of Arizona, Tucson, AZ 85721, USA
- Department of Immunobiology, University of Arizona, Tucson, AZ 85721, USA
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ 85721, USA
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ 85721, USA
- Department of Immunobiology, University of Arizona, Tucson, AZ 85721, USA
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
- Department of Medicine, University of Arizona, Tucson, AZ 85721, USA
- Department of Pathology, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
39
|
Mohapatra A, Rajendrakumar SK, Cherukula K, Park MS, Padmanaban S, Vasukuty A, Mohanty A, Lee JY, Bae WK, Park IK. A sugar modified amphiphilic cationic nano-adjuvant ceased tumor immune suppression and rejuvenated peptide vaccine induced antitumor immunity in cervical cancer. Biomater Sci 2023; 11:1853-1866. [PMID: 36655902 DOI: 10.1039/d2bm01715f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Human papilloma virus (HPV), one of the most common cancer-causing viruses, accounts for more than 90% of human anal and cervical cancers. Clinical studies have focused on adjuvant therapy with vaccines to improve therapeutic outcomes in patients with late-stage HPV-related cancers. In the present study, a mannose receptor (CD206) targeting a lithocholic acid-modified polyethylenimine (PEI) nano-adjuvant delivering the toll-like receptor 7/8 agonist, resiquimod (R848) (mLAPMi-R848), in a HPV E6- and E7-expressing TC-1 tumor murine model was developed. Peritumoral administration of mLAPMi resulted in enhanced accumulation in tumor/tumor-draining lymph nodes and significantly targeted antigen presenting cells like macrophage and dendritic cells. PEI-based nanocarriers can exploit the adjuvant potency of R848 and improve the antitumor immunity. Hence, co-administration of mLAPMi-R848 along with an E6E7 peptide in TC-1 tumor mice eradicated tumor burden and elicited splenocyte-induced cytotoxicity in TC-1 cancer cells. In a bilateral TC-1 tumor model, administration of mLAPMi-R848 and E6E7 peptide significantly suppressed both primary and secondary tumor burdens and improved the overall survival rate. Immune cell profiling revealed elevated levels of mature DCs and CD8+ T cells but reduced levels of tumor-associated immunosuppressive cells (TAICs) like myeloid derived suppressor cells (MDSCs) and regulatory T (Treg) cells in distal tumors. Overall, this study demonstrated that mLAPMi-R848 has improved the antitumor immunity of the peptide antigen against HPV-induced cancers by targeted immunodulation of antigen presenting cells (APCs) and reducing TAICs. Furthermore, this nano-adjuvant has the potential to offer a new treatment option for patients with cervical cancer and can be applied for the treatment of other HPV induced cancers.
Collapse
Affiliation(s)
- Adityanarayan Mohapatra
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Santhosh Kalash Rajendrakumar
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Kondareddy Cherukula
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Myong-Suk Park
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Sathiyamoorthy Padmanaban
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Arathy Vasukuty
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Ayeskanta Mohanty
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Engineering, Gwangju 61005, Korea
| | - Woo Kyun Bae
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - In-Kyu Park
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| |
Collapse
|
40
|
Shen W, Wang X, Xiang H, Shichi S, Nakamoto H, Kimura S, Sugiyama K, Taketomi A, Kitamura H. IFN-γ-STAT1-mediated NK2R expression is involved in the induction of antitumor effector CD8 + T cells in vivo. Cancer Sci 2023; 114:1816-1829. [PMID: 36715504 PMCID: PMC10154869 DOI: 10.1111/cas.15738] [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: 06/13/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
The induction of antitumor effector T cells in the tumor microenvironment is a crucial event for cancer immunotherapy. Neurokinin receptor 2 (NK2R), a G protein-coupled receptor for neurokinin A (NKA), regulates diverse physiological functions. However, the precise role of NKA-NK2R signaling in antitumor immunity is unclear. Here, we found that an IFN-γ-STAT1 cascade augmented NK2R expression in CD8+ T cells, and NK2R-mediated NKA signaling was involved in inducing antitumor effector T cells in vivo. The administration of a synthetic analog of double-stranded RNA, polyinosinic-polycytidylic acid (poly I:C), into a liver cancer mouse model induced type I and type II IFNs and significantly suppressed the tumorigenesis of Hepa1-6 liver cancer cells in a STAT1-dependent manner. The reduction in tumor growth was diminished by the depletion of CD8+ T cells. IFN-γ stimulation significantly induced NK2R and tachykinin precursor 1 (encodes NKA) gene expression in CD8+ T cells. NKA stimulation combined with anti-CD3 monoclonal antibody (mAb) treatment significantly augmented IFN-γ and granzyme B production by CD8+ T cells compared with the anti-CD3 mAb alone in vitro. ERK1/2 phosphorylation and IκBα degradation in activated CD8+ T cells were suppressed under NK2R deficiency. Finally, we confirmed that tumor growth was significantly increased in NK2R-deficient mice compared with that in wild-type mice, and the antitumor effects of poly I:C were abolished by NK2R absence. These findings suggest that IFN-γ-STAT1-mediated NK2R expression is involved in the induction of antitumor effector T cells in the tumor microenvironment, which contributes to the suppression of cancer cell tumorigenesis in vivo. In this study, we revealed that IFN-γ-STAT1-mediated NK2R expression is involved in the induction of antitumor effector CD8+ T cells in the tumor microenvironment, which contributes to suppressing the tumorigenesis of liver cancer cells in vivo.
Collapse
Affiliation(s)
- Weidong Shen
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Xiangdong Wang
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Huihui Xiang
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Shunsuke Shichi
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroki Nakamoto
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Saori Kimura
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ko Sugiyama
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hidemitsu Kitamura
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
41
|
Effects of Bacillus subtilis Natto Strains on Antiviral Responses in Resiquimod-Stimulated Human M1-Phenotype Macrophages. Foods 2023; 12:foods12020313. [PMID: 36673407 PMCID: PMC9858497 DOI: 10.3390/foods12020313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Bacillus subtilis natto is used in the production of natto, a traditional fermented soy food, and has beneficial immunomodulatory effects in humans. Single-stranded RNA (ssRNA) viruses, including influenza and coronavirus, often cause global pandemics. We proposed a human cell culture model mimicking ssRNA viral infection and investigated the ability of B. subtilis natto to induce antiviral effects in the model. The gene expressions were analyzed using quantitative real-time reverse transcription PCR. M1-phenotype macrophages derived from THP-1 cells strongly express the Toll-like receptor 8 (76.2-hold), CD80 (64.2-hold), and CCR7 (45.7-hold) mRNA compared to M0 macrophages. One µg/mL of resiquimod (RSQ)-stimulation induced the expression of IRF3 (1.9-hold), CXCL10 (14.5-hold), IFNβ1 (3.5-hold), ISG20 (4.4-hold), and MxA (1.7-hold) mRNA in the M1-phenotype macrophages. Based on these results, the RSQ-stimulated M1-phenotype macrophages were used as a cell culture model mimicking ssRNA viral infection. Moreover, the B. subtilis natto XF36 strain induced the expression of genes associated with antiviral activities (IFNβ1, IFNλ1, ISG20, and RNase L) and anti-inflammatory activities (IL-10) in the cell culture model. Thus, it is suggested that the XF36 suppresses viral infections and excessive inflammation by inducing the expression of genes involved in antiviral and anti-inflammatory activities.
Collapse
|
42
|
Abstract
In most solid cancers, tumor-associated macrophages (TAMs) infiltrating the tumor microenvironment (TME) represent a major population of immunosuppressive cells. This correlates with poor prognosis and resistance to antitumoral therapies, including immune checkpoint inhibitors. Although initial preclinical studies were primarily meant to deplete macrophages in the TME or prevent their recruitment at tumor sites, recent evidence has indicated that the reprogramming of macrophages into cytotoxic effectors might be more beneficial in eliciting an effective antitumor immune response. Taking this into consideration, the comprehensive analysis of the phenotype and function of macrophages in the TME, and their interaction with cancer cells or other immune cells, has become of paramount importance in oncological research. Accordingly, here we explain the experimental procedures for the in vivo evaluation of tumor progression and response to therapy, with a particular focus on the detailed analysis of TAMs and related immune cells in the TME by flow cytometry, RNA analysis, and multiplex immunophenotyping. The output generated through these experiments allow researchers to test the efficacy of new therapeutic strategies on targeting.
Collapse
|
43
|
Ummarino A, Anfray C, Maeda A, Andón FT, Allavena P. In Vitro Methods to Evaluate Macrophage Polarization and Function in Cancer. Methods Mol Biol 2023; 2614:81-91. [PMID: 36587120 DOI: 10.1007/978-1-0716-2914-7_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tumor-associated macrophages (TAMs) play a key immunosuppressive role that limits the ability of the immune system to fight cancer and hinder the anti-tumoral efficacy of most treatments currently applied in the clinic. However, a key feature of macrophages is their phenotypical and functional plasticity, which called their attention as promising targets for therapeutic intervention based on their elimination or reprogramming toward M1-like cytotoxic effector cells, with anti-tumor functions. This polarization status of macrophages can be studied in terms of molecular markers and functional activities, using an appropriate combination of experimental methodologies, both in vitro and in vivo. Here we focus on describing in vitro protocols to isolate primary monocytes from buffy coats and to study macrophage phenotype and function, after exposure to new therapies, by a combination of flow cytometry, RT-PCR, and ELISA analysis. We also provide the methodology to evaluate in vitro the cytotoxic activity of treated macrophages toward cancer cells.
Collapse
Affiliation(s)
- Aldo Ummarino
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy.,Humanitas University, Rozzano-Milano, Italy
| | - Clément Anfray
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano-Milano, Italy
| | - Akihiro Maeda
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Fernando Torres Andón
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy.,Center for Research in Molecular Medicine & Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Paola Allavena
- Department of Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy. .,Humanitas University, Rozzano-Milano, Italy.
| |
Collapse
|
44
|
Chaintreuil P, Kerreneur E, Bourgoin M, Savy C, Favreau C, Robert G, Jacquel A, Auberger P. The generation, activation, and polarization of monocyte-derived macrophages in human malignancies. Front Immunol 2023; 14:1178337. [PMID: 37143666 PMCID: PMC10151765 DOI: 10.3389/fimmu.2023.1178337] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/04/2023] [Indexed: 05/06/2023] Open
Abstract
Macrophages are immune cells that originate from embryogenesis or from the differentiation of monocytes. They can adopt numerous phenotypes depending on their origin, tissue distribution and in response to different stimuli and tissue environment. Thus, in vivo, macrophages are endowed with a continuum of phenotypes that are rarely strictly pro-inflammatory or anti-inflammatory and exhibit a broad expression profile that sweeps over the whole polarization spectrum. Schematically, three main macrophage subpopulations coexist in human tissues: naïve macrophages also called M0, pro-inflammatory macrophages referred as M1 macrophages, and anti-inflammatory macrophages also known as M2 macrophages. Naïve macrophages display phagocytic functions, recognize pathogenic agents, and rapidly undergo polarization towards pro or anti-inflammatory macrophages to acquire their full panel of functions. Pro-inflammatory macrophages are widely involved in inflammatory response, during which they exert anti-microbial and anti-tumoral functions. By contrast, anti-inflammatory macrophages are implicated in the resolution of inflammation, the phagocytosis of cell debris and tissue reparation following injuries. Macrophages also play important deleterious or beneficial roles in the initiation and progression of different pathophysiological settings including solid and hematopoietic cancers. A better understanding of the molecular mechanisms involved in the generation, activation and polarization of macrophages is a prerequisite for the development of new therapeutic strategies to modulate macrophages functions in pathological situations.
Collapse
Affiliation(s)
- Paul Chaintreuil
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Emeline Kerreneur
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Maxence Bourgoin
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Coline Savy
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Cécile Favreau
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Guillaume Robert
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Arnaud Jacquel
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- *Correspondence: Arnaud Jacquel, ; Patrick Auberger,
| | - Patrick Auberger
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale, Nice, France
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- *Correspondence: Arnaud Jacquel, ; Patrick Auberger,
| |
Collapse
|
45
|
Abstract
Accumulating evidence indicates that Toll-like receptor (TLR) agonists proficiently (re)instore cancer immunosurveillance as immunological adjuvants. So far, three TLR agonists have been approved by regulatory agencies for use in oncological applications. Additionally, these immunotherapeutics have been extensively investigated over the past few years. Multiple clinical trials are currently evaluating the combination of TLR agonists with chemotherapy, radiotherapy, or different immunotherapies. Moreover, antibodies targeting tumor-enriched surface proteins that have been conjugated to TLR agonists are being developed to stimulate anticancer immune responses specifically within the tumor microenvironment. Solid preclinical and translational results support the favorable immune-activating effects of TLR agonists. Here, we summarize recent preclinical and clinical advances in the development of TLR agonists for anticancer immunotherapy.
Collapse
Affiliation(s)
- Julie Le Naour
- Centre de Recherche Des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Guido Kroemer
- Centre de Recherche Des Cordeliers, Equipe Labellisée Par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.,Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
46
|
Pereira-Veiga T, Bravo S, Gómez-Tato A, Yáñez-Gómez C, Abuín C, Varela V, Cueva J, Palacios P, Dávila-Ibáñez AB, Piñeiro R, Vilar A, Chantada-Vázquez MDP, López-López R, Costa C. Red Blood Cells Protein Profile Is Modified in Breast Cancer Patients. Mol Cell Proteomics 2022; 21:100435. [PMID: 36519745 PMCID: PMC9713370 DOI: 10.1016/j.mcpro.2022.100435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
Metastasis is the primary cause of death for most breast cancer (BC) patients who succumb to the disease. During the hematogenous dissemination, circulating tumor cells interact with different blood components. Thus, there are microenvironmental and systemic processes contributing to cancer regulation. We have recently published that red blood cells (RBCs) that accompany circulating tumor cells have prognostic value in metastatic BC patients. RBC alterations are related to several diseases. Although the principal known role is gas transport, it has been recently assigned additional functions as regulatory cells on circulation. Hence, to explore their potential contribution to tumor progression, we characterized the proteomic composition of RBCs from 53 BC patients from stages I to III and IV, compared with 33 cancer-free controls. In this work, we observed that RBCs from BC patients showed a different proteomic profile compared to cancer-free controls and between different tumor stages. The differential proteins were mainly related to extracellular components, proteasome, and metabolism. Embryonic hemoglobins, not expected in adults' RBCs, were detected in BC patients. Besides, lysosome-associated membrane glycoprotein 2 emerge as a new RBCs marker with diagnostic and prognostic potential for metastatic BC patients. Seemingly, RBCs are acquiring modifications in their proteomic composition that probably represents the systemic cancer disease, conditioned by the tumor microenvironment.
Collapse
Affiliation(s)
- Thais Pereira-Veiga
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Susana Bravo
- Proteomic Unit, Instituto de Investigaciones Sanitarias-IDIS, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Antonio Gómez-Tato
- CITMAga, University of Santiago de Compostela (Campus Vida), Santiago de Compostela, Spain
| | - Celso Yáñez-Gómez
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Carmen Abuín
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Vanesa Varela
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Juan Cueva
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Patricia Palacios
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Ana B Dávila-Ibáñez
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Roberto Piñeiro
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Ana Vilar
- Department of Gynecology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - María Del Pilar Chantada-Vázquez
- Proteomic Unit, Instituto de Investigaciones Sanitarias-IDIS, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Rafael López-López
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain.
| | - Clotilde Costa
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain.
| |
Collapse
|
47
|
Zhang Z, Kuo JCT, Zhang C, Huang Y, Lee RJ. Ivermectin Enhanced Antitumor Activity of Resiquimod in a Co-Loaded Squalene Emulsion. J Pharm Sci 2022; 111:3038-3046. [PMID: 35697319 DOI: 10.1016/j.xphs.2022.06.005] [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: 02/18/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 12/14/2022]
Abstract
Immunogenic cell death (ICD) plays an important role in sensitizing tumor cells to antigen-presenting cells followed by antitumor immunity. However, a successful antitumor response by ICD requires both apoptotic tumor microenvironments and activated immune systems. Ivermectin (IVM) has been shown to induce cell apoptosis through autophagy which can be a great candidate for ICD therapy. However, a single treatment of IVM-free drug is not an ideal anticancer therapy due to its anti-inflammatory effects and cytotoxicity. In the present study, IVM was shown to enhance the ICD process in addition to the treatment of resiquimod (R848), a TLR7/8 agonist, when co-loaded in a squalene-based nanoemulsion (NE). R848-IVM co-loaded NE was developed and characterized in vitro. Antitumor activity of R848-IVM NE was also evaluated in vitro and in vivo. R848-IVM NE exhibited long-term stability and reduced cytotoxicity by IVM. In vivo studies demonstrated that IVM significantly augments the ICD by upregulating Cd8a and releasing HMGB1 in tumor tissue, which could enhance R848-driven antitumor immunity. R848-IVM NE treatment showed strong antitumor activity with over 80% tumor growth inhibition, suggesting a potential combination therapy of systemic co-delivering IVM with TLR agonists against solid cancer.
Collapse
Affiliation(s)
- Zhongkun Zhang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH, 43210, USA.
| | - Jimmy Chun-Tien Kuo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH, 43210, USA.
| | - Chi Zhang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH, 43210, USA.
| | - Yirui Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH, 43210, USA
| | - Robert J Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
48
|
Wang Z, Zhong H, Liang X, Ni S. Targeting tumor-associated macrophages for the immunotherapy of glioblastoma: Navigating the clinical and translational landscape. Front Immunol 2022; 13:1024921. [PMID: 36311702 PMCID: PMC9606568 DOI: 10.3389/fimmu.2022.1024921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/03/2022] [Indexed: 12/05/2022] Open
Abstract
Tumor-associated macrophages (TAMs) can directly clear tumor cells and enhance the phagocytic ability of immune cells. An abundance of TAMs at the site of the glioblastoma tumor indicates that TAM-targeting immunotherapy could represent a potential form of treatment for this aggressive cancer. Herein, we discuss: i) the dynamic role of TAMs in glioblastoma; ii) describe the formation of the immunosuppressive tumor microenvironment; iii) summarize the latest clinical trial data that reveal how TAM function can be regulated in favor tumor eradication; and lastly, iv) evaluate the implications of existing and novel translational approaches for treating glioblastoma in clinical practice.
Collapse
Affiliation(s)
- Zide Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Hanlin Zhong
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College of Shandong University, Jinan, China
- *Correspondence: Xiaohong Liang, ; Shilei Ni,
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- *Correspondence: Xiaohong Liang, ; Shilei Ni,
| |
Collapse
|
49
|
Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov 2022; 21:799-820. [PMID: 35974096 PMCID: PMC9380983 DOI: 10.1038/s41573-022-00520-5] [Citation(s) in RCA: 785] [Impact Index Per Article: 261.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2022] [Indexed: 12/11/2022]
Abstract
Tumour-associated macrophages are an essential component of the tumour microenvironment and have a role in the orchestration of angiogenesis, extracellular matrix remodelling, cancer cell proliferation, metastasis and immunosuppression, as well as in resistance to chemotherapeutic agents and checkpoint blockade immunotherapy. Conversely, when appropriately activated, macrophages can mediate phagocytosis of cancer cells and cytotoxic tumour killing, and engage in effective bidirectional interactions with components of the innate and adaptive immune system. Therefore, they have emerged as therapeutic targets in cancer therapy. Macrophage-targeting strategies include inhibitors of cytokines and chemokines involved in the recruitment and polarization of tumour-promoting myeloid cells as well as activators of their antitumorigenic and immunostimulating functions. Early clinical trials suggest that targeting negative regulators (checkpoints) of myeloid cell function indeed has antitumor potential. Finally, given the continuous recruitment of myelomonocytic cells into tumour tissues, macrophages are candidates for cell therapy with the development of chimeric antigen receptor effector cells. Macrophage-centred therapeutic strategies have the potential to complement, and synergize with, currently available tools in the oncology armamentarium. Macrophages can promote tumorigenesis and enhance the antitumour response. This Review discusses the molecular mechanisms underlying the reprogramming of macrophages in the tumour microenvironment and provides an overview of macrophage-targeted therapies for the treatment of cancer.
Collapse
Affiliation(s)
- Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy. .,IRCCS- Humanitas Research Hospital, Milan, Italy. .,The William Harvey Research Institute, Queen Mary University of London, London, UK.
| | - Paola Allavena
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS- Humanitas Research Hospital, Milan, Italy
| | - Federica Marchesi
- IRCCS- Humanitas Research Hospital, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS- Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
50
|
Andón FT, Leon S, Ummarino A, Redin E, Allavena P, Serrano D, Anfray C, Calvo A. Innate and Adaptive Responses of Intratumoral Immunotherapy with Endosomal Toll-Like Receptor Agonists. Biomedicines 2022; 10:1590. [PMID: 35884895 PMCID: PMC9313389 DOI: 10.3390/biomedicines10071590] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
Toll-like receptors (TLRs) are natural initial triggers of innate and adaptive immune responses. With the advent of cancer immunotherapy, nucleic acids engineered as ligands of endosomal TLRs have been investigated for the treatment of solid tumors. Despite promising results, their systemic administration, similarly to other immunotherapies, raises safety issues. To overcome these problems, recent studies have applied the direct injection of endosomal TLR agonists in the tumor and/or draining lymph nodes, achieving high local drug exposure and strong antitumor response. Importantly, intratumoral delivery of TLR agonists showed powerful effects not only against the injected tumors but also often against uninjected lesions (abscopal effects), resulting in some cases in cure and antitumoral immunological memory. Herein, we describe the structure and function of TLRs and their role in the tumor microenvironment. Then, we provide our vision on the potential of intratumor versus systemic delivery or vaccination approaches using TLR agonists, also considering the use of nanoparticles to improve their targeting properties. Finally, we collect the preclinical and clinical studies applying intratumoral injection of TLR agonists as monotherapies or in combination with: (a) other TLR or STING agonists; (b) other immunotherapies; (c) radiotherapy or chemotherapy; (d) targeted therapies.
Collapse
Affiliation(s)
- Fernando Torres Andón
- Center for Research in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy;
| | - Sergio Leon
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, 31008 Pamplona, Spain; (S.L.); (E.R.); (D.S.)
| | - Aldo Ummarino
- Laboratory of Cellular Immunology, Humanitas University, 20089 Pieve Emanuele, Italy; (A.U.); (C.A.)
| | - Esther Redin
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, 31008 Pamplona, Spain; (S.L.); (E.R.); (D.S.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Navarra Institute for Health Research (IdiSNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Paola Allavena
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy;
- Laboratory of Cellular Immunology, Humanitas University, 20089 Pieve Emanuele, Italy; (A.U.); (C.A.)
| | - Diego Serrano
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, 31008 Pamplona, Spain; (S.L.); (E.R.); (D.S.)
- Navarra Institute for Health Research (IdiSNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Clément Anfray
- Laboratory of Cellular Immunology, Humanitas University, 20089 Pieve Emanuele, Italy; (A.U.); (C.A.)
| | - Alfonso Calvo
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Department of Pathology and Histology, University of Navarra, 31008 Pamplona, Spain; (S.L.); (E.R.); (D.S.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Navarra Institute for Health Research (IdiSNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| |
Collapse
|