1
|
Liu C, Li M, Liu L, Xu Q, Zheng L, Wu C, Ren J, Zhang T, Wang H, Lin Z. TGF-β1 induces autophagy and mediates the effect on macrophages differentiation in primary liver cancer. Int Immunopharmacol 2025; 157:114799. [PMID: 40339499 DOI: 10.1016/j.intimp.2025.114799] [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: 02/04/2025] [Revised: 05/01/2025] [Accepted: 05/01/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are closely associated with tumor development and patient outcomes due to their plasticity and polarization capacity. Several distinct TAMs have been proposed, but a complete understanding of heterogeneity and differentiation spectrum of macrophage in human primary liver cancer remains elusive. METHODS Deep single-cell RNA sequencing (scRNA-seq) data from 19 primary liver cancer patients were used to profile the transcriptomes of TAMs in liver cancer. Ingenuity pathway analysis (IPA) and in vitro experiments were used to explore possible mechanisms responsible for related signaling pathways altered at the transcriptional level. Finally, we analyzed the relationship between the abundance of the TAMs and the survival outcomes of the 428 patients in the Cancer Genome Atlas (TCGA). RESULTS Transcriptional profiles allowed us to identify four distinct TAMs cell subsets based on molecular and functional properties and to reconstruct their developmental trajectory. Specifically, TAM_c4 was preferentially enriched and potentially expanded in the advanced-stage patients or those receiving immune checkpoint blockade therapy (ICT). Gene pathway analysis revealed aberrant TGFB1 activation in TAM_c4, which was experimentally confirmed to drive TAM phenotypic transitions via autophagy signaling. High abundance of TAM_c4 is found to be related to a short survival time and low abundance of CD8+ T cells in primary liver cancers. CONCLUSIONS This integrated transcriptome compendium and experimental validation offer both mechanistic insights and a resource for understanding TAM heterogeneity in primary liver cancers.
Collapse
Affiliation(s)
- Chao Liu
- Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, People's Republic of China
| | - Mingjie Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lichao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qian Xu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Linlin Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Cailing Wu
- Faculty of Medicine, JiuJiang University, Jiujiang, People's Republic of China
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, People's Republic of China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
2
|
Li D, Rudloff U. Emerging therapeutics targeting tumor-associated macrophages for the treatment of solid organ cancers. Expert Opin Emerg Drugs 2025:1-39. [PMID: 40353504 DOI: 10.1080/14728214.2025.2504376] [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: 12/03/2024] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Over the last decade, immune checkpoint inhibitors (ICIs) like PD-1/PD-L1 or CTLA-4, which reinvigorate T cells for tumor control have become standard-of-care treatment options. In response to the increasingly recognized mechanisms of resistance to T cell activation in immunologically cold tumors, immuno-oncology drug development has started to shift beyond T cell approaches. These include tumor-associated macrophages (TAMs), a major pro-tumor immune cell population in the tumor microenvironment known to silence immune responses. AREAS COVERED Here we outline anti-TAM therapies in current development, either as monotherapy or in combination with other treatment modalities. We describe emerging drugs targeting TAMs under investigation in phase II and III testing with a focus on their distinguishing mechanism of action which include (1) reprogramming of TAMs toward anti-tumor function and immune surveillance, (2) blockade of recruitment, and (3) reduction and ablation of TAMs. EXPERT OPINION Several new immuno-oncology agents are under investigation to harness anti-tumor functions of TAMs. While robust anti-tumor efficacy of anti-TAM therapies across advanced solid organ cancers remains elusive to-date, TAM reprogramming therapies have yielded benefits in select cancers. The inherent heterogeneity of the diverse TAM population will require enhanced investments into biomarker-driven approaches to fully leverage its therapeutic potential.
Collapse
Affiliation(s)
- Dandan Li
- Developmental Therapeutics Branch (TDB), Biology Group, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Udo Rudloff
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
3
|
Saeed AF. Tumor-Associated Macrophages: Polarization, Immunoregulation, and Immunotherapy. Cells 2025; 14:741. [PMID: 40422244 DOI: 10.3390/cells14100741] [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: 04/01/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025] Open
Abstract
Tumor-associated macrophages' (TAMs) origin, polarization, and dynamic interaction in the tumor microenvironment (TME) influence cancer development. They are essential for homeostasis, monitoring, and immune protection. Cells from bone marrow or embryonic progenitors dynamically polarize into pro- or anti-tumor M2 or M1 phenotypes based on cytokines and metabolic signals. Recent advances in TAM heterogeneity, polarization, characterization, immunological responses, and therapy are described here. The manuscript details TAM functions and their role in resistance to PD-1/PD-L1 blockade. Similarly, TAM-targeted approaches, such as CSF-1R inhibition or PI3Kγ-driven reprogramming, are discussed to address anti-tumor immunity suppression. Furthermore, innovative biomarkers and combination therapy may enhance TAM-centric cancer therapies. It also stresses the relevance of this distinct immune cell in human health and disease, which could impact future research and therapies.
Collapse
|
4
|
Mongeon B, Craig M. Virtual Clinical Trial Reveals Significant Clinical Potential of Targeting Tumor-Associated Macrophages and Microglia to Treat Glioblastoma. CPT Pharmacometrics Syst Pharmacol 2025. [PMID: 40347051 DOI: 10.1002/psp4.70033] [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: 01/20/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 05/12/2025] Open
Abstract
Glioblastoma is the most aggressive primary brain tumor, with a median survival of 15 months with treatment. Standard-of-care (SOC) consists of resection, radio- and chemotherapy. Clinical trials involving PD-1 inhibition with nivolumab combined with SOC failed to increase survival. A quantitative understanding of the interactions between the tumor and its immune environment that drive treatment outcomes is currently lacking. As such, we developed a mathematical model of tumor growth that considers CD8+ T cells, pro- and antitumoral tumor-associated macrophages and microglia (TAMs), SOC, and nivolumab. Using our model, we studied five TAM-targeting strategies currently under investigation for solid tumors. Our results show that PD-1 inhibition fails due to a lack of CD8+ T cell recruitment during treatment, explained by TAM-driven immunosuppressive mechanisms. Our model predicts that while reducing TAM numbers does not improve prognosis, altering their functions to counter their protumoral properties has the potential to considerably reduce post-treatment tumor burden. In particular, restoring antitumoral TAM phagocytic activity through anti-CD47 treatment in combination with SOC was predicted to nearly eradicate the tumor. By studying time-varying efficacy with the same half-life as the anti-CD47 antibody Hu5F9-G4, our model predicts that repeated dosing of anti-CD47 provides sustained control of tumor growth. We propose that targeting TAMs by enhancing their antitumoral properties is a highly promising avenue to treat glioblastoma and warrants future clinical development. Together, our results provide proof-of-concept that mechanistic mathematical modeling can uncover the mechanisms driving treatment outcomes and explore the potential of novel treatment strategies for hard-to-treat tumors like glioblastoma.
Collapse
Affiliation(s)
- Blanche Mongeon
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, Quebec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Quebec, Canada
| | - Morgan Craig
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, Quebec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
5
|
Summer M, Riaz S, Ali S, Noor Q, Ashraf R, Khan RRM. Understanding the Dual Role of Macrophages in Tumor Growth and Therapy: A Mechanistic Review. Chem Biodivers 2025; 22:e202402976. [PMID: 39869825 DOI: 10.1002/cbdv.202402976] [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: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
Macrophages are heterogeneous cells that are the mediators of tissue homeostasis. These immune cells originated from monocytes and are classified into two basic categories, M1 and M2 macrophages. M1 macrophages exhibit anti-tumorous inflammatory reactions due to the behavior of phagocytosis. M2 macrophages or tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) and have a basic role in tumor progression by interacting with other immune cells in TME. By the expression of various cytokines, chemokines, and growth factors, TAMs lead to strengthening tumor cell proliferation, angiogenesis, and suppression of the immune system which further support invasion and metastasis. This review discusses recent and updated mechanisms regarding tumor progression by M2 macrophages. Moreover, the current therapeutic approaches targeting TAMs, their advantages, and limitations are also summarized, and further treatment approaches are outlined along with an elaboration of the tumor regression role of macrophages. This comprehensive review article possibly helps to understand the mechanisms underlying the tumor progression and regression role of macrophages in a comparative way from a basic level to the advanced one.
Collapse
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Saima Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Qudsia Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rimsha Ashraf
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rana Rashad Mahmood Khan
- Faculty of Chemistry and Life Sciences, Department of Chemistry, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|
6
|
Fu ZD, Wang Y, Yan HL, Wu JH. Mendelian Randomization Analysis Supports a Causal Relationship Between Circulating Inflammatory Proteins and Basal Cell Carcinoma. Clin Cosmet Investig Dermatol 2025; 18:767-779. [PMID: 40190471 PMCID: PMC11970273 DOI: 10.2147/ccid.s521068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/22/2025] [Indexed: 04/09/2025]
Abstract
Background It has been shown that the Basal cell carcinoma (BCC) is associated with chronic inflammation of skin conditions, the circulating inflammatory protein levels may be a more intuitive index in response to inflammation, however, the cause-and-effect relationship between circulating inflammatory proteins and BCC is currently unknown. Methods This study performed a Mendelian randomization (MR) analysis using the plasma inflammatory protein levels from a large genome-wide protein quantitative trait loci study as the exposure data, and the outcome data from a GWAS for BCC. Inverse variance weighed, MR-Egger, maximum likelihood ratio, and weighted median for assessing causality between inflammatory proteins and BCC. MR-Egger regression and Cochran's Q statistic were applied for sensitivity analysis and MRPRESSO was applied to exclude outliers. Inverse MR analysis was performed on inflammatory proteins found to be causally associated with BCC. Results Six circulating inflammatory proteins with a causal relationship with BCC were obtained, including CCL4, was of a significant protective effect on BCC development. IL-18 and CCL28, were of suggestive protective effects on BCC development. CX3CL1, IL-17A, and CSF-1 were potential risk factors in the development of BCC. According to the results of reverse MR analysis, there is no significant causal relationship between BCC and the above-mentioned inflammatory proteins. Conclusion This two-sample MR study revealed a strong association between circulating inflammatory proteins and the development of BCC. Specifically, CCL4, CCL28, IL-18, CX3CL1, IL-17A, and CSF-1 emerged as potential targets for prognostic evaluation and treatment of BCC. However, further experimental studies are needed to elucidate the specific mechanisms.
Collapse
Affiliation(s)
- Zhi-da Fu
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
- Center for Reproductive Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Yao Wang
- Shanghai International Travel Healthcare Center, Shanghai Customs District, Shanghai, People’s Republic of China
| | - Hong-li Yan
- Center for Reproductive Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Jian-hua Wu
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
7
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
8
|
Suliman M, Saleh RO, Chandra M, Rasool KH, Jabir M, Jawad SF, Hasan TF, Singh M, Singh M, Singh A. Macrophage-derived lncRNAs in cancer: regulators of tumor progression and therapeutic targets. Med Oncol 2025; 42:91. [PMID: 40048034 DOI: 10.1007/s12032-025-02643-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Macrophages are key tumor microenvironment (TME) regulators, exhibiting remarkable plasticity that enables them to either suppress or promote cancer progression. Emerging evidence highlights the critical role of macrophage-derived long non-coding RNAs (lncRNAs) in shaping tumor immunity, influencing macrophage polarization, immune evasion, angiogenesis, metastasis, and therapy resistance. This review comprehensively elucidates the functional roles of M1- and M2-associated lncRNAs, detailing their molecular mechanisms and impact on cancer pathogenesis. In summary, elucidating the roles of lncRNAs derived from macrophages in cancer progression offers new avenues for therapeutic strategies, significantly improving patient outcomes in the fight against the disease. Further research into the functional significance of these lncRNAs and the development of targeted therapies is essential to harness their potential fully in clinical applications. We further explore their potential as biomarkers for cancer prognosis and therapeutic targets for modulating macrophage activity to enhance anti-cancer immunity. Targeting macrophage-derived lncRNAs represents a promising avenue for precision oncology, offering novel strategies to reshape the TME and improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Raed Obaid Saleh
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al Maarif, Anbar, Iraq.
| | - Muktesh Chandra
- Marwadi University Research Center, Department of Bioinformatics, Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - Majid Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, 51001, Hillah, Babylon, Iraq
| | - Thikra F Hasan
- College of Health & Medical Technology, Uruk University, Baghdad, Iraq
| | - Mithilesh Singh
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Manmeet Singh
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
9
|
Su Y, Ouchi R, Daroonpan P, Hamagaki M, Ikeda T, Rika N, Nishii N, Tsushima F, Kano Y, Asakage T, Noguchi M, Harada H, Azuma M. Stratification of the immunotypes of tongue squamous cell carcinoma to improve prognosis and the response to immune checkpoint inhibitors. Cancer Immunol Immunother 2025; 74:130. [PMID: 40025278 PMCID: PMC11872838 DOI: 10.1007/s00262-025-03982-9] [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: 01/23/2025] [Accepted: 02/11/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVES An understanding of the tumor immune microenvironment is required to improve treatment, especially the selection of immune checkpoint inhibitors (ICIs). In this study, we stratified the immunotypes of tongue squamous cell carcinoma (TSCC) based on the results of comprehensive immune profiling. METHODS We enrolled 87 therapy-naïve TSCC and 17 ICI-treated TSCC patients who underwent glossectomy without any other prior therapy. Comprehensive immune profile analyses employed multiplex immunofluorescence and tissue imaging. RESULTS Based on the hierarchies of 58 immune parameters and the spatial distances between cytotoxic T lymphocytes (CTL) and tumor cells, we stratified five immunotypes: Immunoactive type I, border type II, immunosuppressed type III, immunoisolating type IV, and immunodesert type V. The type I frequency was only 16%. Most TSCCs (~ 70%) were of types III-V. The CTL density (CTL-D) was closely correlated with the PD-L1+ pan-macrophages (panM)-D, and the panM-D closely correlated with the PD-1+ CTL-D. This indicated that PD-1 and PD-L1 expression required macrophages and CTL recruitment in the tumor microenvironment. No ICI-treated TSCC patients, all of whom were recurrent/metastatic cases, were of the type I immunotype, and almost half (47.0%) were of the immunodesert type V. Most cases exhibited an imbalance between T-cell PD-1 and macrophage PD-L1 expression. CONCLUSION We defined five TSCC-specific immunotypes based on the results of comprehensive immune profiling analyses. Immunoactive type, which would be sensitive to ICI monotherapy, was rare, and most TSCC cases exhibited immune-regulated immunotypes. Immunotype-based personalized treatments are required to improve clinical outcomes.
Collapse
Affiliation(s)
- Yuya Su
- Department of Oral and Maxillofacial Surgical Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (SCIENCE TOKYO), 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
- Oral Science Center (OSC), Institute of Science Tokyo (SCIENCE TOKYO), Tokyo, 113-8519, Japan
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8549, Japan
| | - Ryo Ouchi
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8549, Japan
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama, 930-0194, Japan
| | - Pissacha Daroonpan
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8549, Japan
- Department of Oral Diagnosis, Naresuan University, Phitsanulok, 65000, Thailand
| | - Miwako Hamagaki
- Division of Surgical Pathology, Institute of Science Tokyo Hospital, Tokyo, 113-8519, Japan
| | - Tohru Ikeda
- Department of Oral Pathology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (SCIENCE TOKYO), Tokyo, 113-8519, Japan
| | - Noji Rika
- Department of Oral and Maxillofacial Surgical Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (SCIENCE TOKYO), 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Naoto Nishii
- Department of Oral and Maxillofacial Surgical Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (SCIENCE TOKYO), 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Fumihiko Tsushima
- Department of Oral and Maxillofacial Surgical Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (SCIENCE TOKYO), 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Yoshihito Kano
- Department of Medical Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (SCIENCE TOKYO), Tokyo, 113-8519, Japan
| | - Takahiro Asakage
- Department of Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (SCIENCE TOKYO), Tokyo, 113-8519, Japan
| | - Makoto Noguchi
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama, 930-0194, Japan
| | - Hiroyuki Harada
- Department of Oral and Maxillofacial Surgical Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (SCIENCE TOKYO), 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Miyuki Azuma
- Department of Oral and Maxillofacial Surgical Oncology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (SCIENCE TOKYO), 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan.
- Oral Science Center (OSC), Institute of Science Tokyo (SCIENCE TOKYO), Tokyo, 113-8519, Japan.
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8549, Japan.
| |
Collapse
|
10
|
Gambardella V, Ong M, Rodriguez-Ruiz ME, Machiels JP, Sanmamed MF, Galvao V, Spreafico A, Renouf DJ, Luen SJ, Galot R, Doger de Spéville B, Calvo E, Naing A, Curdt S, Kolben TM, Rossmann E, Tanos T, Smart K, Amann M, Xie Y, Xu L, Gomez Alcaide E, Städler N, Justies N, Boetsch C, Karanikas V, Schnetzler G, Rohrberg KS. Safety and Antitumor Activity of a Novel aCD25 Treg Depleter RG6292 as a Single Agent and in Combination with Atezolizumab in Patients with Solid Tumors. CANCER RESEARCH COMMUNICATIONS 2025; 5:422-432. [PMID: 39983024 PMCID: PMC11891644 DOI: 10.1158/2767-9764.crc-24-0638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 02/23/2025]
Abstract
PURPOSE Therapeutic depletion of immunosuppressive regulatory T cells (Treg) may overcome resistance to cancer immunotherapies. RG6292 is an anti-CD25 antibody that preferentially depletes Tregs while preserving effector T-cell functions in preclinical models. The safety, pharmacokinetics, pharmacodynamics, and antitumor efficacy of selective Treg depletion by RG6292 administered as monotherapy or in combination with atezolizumab were evaluated in two phase I studies. PATIENTS AND METHODS Adult patients with advanced solid tumors were administered intravenous RG6292, given every 3 weeks alone (study 1: NCT04158583, n = 76) or with 1,200 mg atezolizumab every 3 weeks (study 2: NCT04642365, n = 49). Both studies included dose escalation and expansion parts to determine the maximum tolerated dose and recommended phase II dose. RESULTS RG6292 was well tolerated. Pruritus and rash were the most frequent adverse events and were manageable with supportive treatment. Serum RG6292 levels increased dose proportionally, independent of the atezolizumab combination. RG6292 induced a sustained dose-dependent depletion of peripheral Tregs with no apparent effect on other immune cells. Evidence of intratumoral Treg reduction (≥50% vs. baseline) was observed at RG6292 doses of 35 to 100 mg. The maximum tolerated dose was 165 mg every 3 weeks, and the recommended phase II dose was proposed as 70 mg every 3 weeks. Objective responses were limited to three partial responses in patients receiving RG6292 combined with atezolizumab. CONCLUSIONS RG6292 induced a dose-dependent peripheral blood and measurable intratumoral Treg depletion in concordance with the proposed mode of action; however, clinical efficacy as a single agent or combined with atezolizumab was insufficient to warrant further exploration in this population. SIGNIFICANCE RG6292 (vopikitug) targets CD25 (IL-2Rα) and mediates regulatory T-cell depletion while not interfering with IL-2 signaling. Peripheral and intratumoral Treg depletion was shown in two phase I studies. However, RG6292 alone or in combination with atezolizumab was insufficient to reverse and rescue from established resistance mechanisms in solid tumors.
Collapse
Affiliation(s)
| | - Michael Ong
- The Ottawa Hospital Cancer Centre, Ottawa, Canada
| | | | - Jean-Pascal Machiels
- Department of Medical Oncology, Institut Roi Albert II, Cliniques universitaires Saint-Luc, and Institut de Recherche Clinique et Expérimentale, UCLouvain, Brussels, Belgium
| | - Miguel F. Sanmamed
- Department of Medical Oncology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Vladimir Galvao
- Vall d/Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | | | - Stephen J. Luen
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Rachel Galot
- Department of Medical Oncology, Institut Roi Albert II, Cliniques universitaires Saint-Luc, and Institut de Recherche Clinique et Expérimentale, UCLouvain, Brussels, Belgium
| | | | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Aung Naing
- MD Anderson Cancer Center, Houston, Texas
| | - Samira Curdt
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Development, Penzberg, Germany
| | - Theresa Maria Kolben
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Development, Penzberg, Germany
| | - Eva Rossmann
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Tamara Tanos
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Kevin Smart
- Roche Innovation Centre Welwyn, Roche Pharmaceutical Research and Early Development, Welwyn, United Kingdom
| | - Maria Amann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, Schlieren, Switzerland
| | - Yuying Xie
- F. Hoffmann-La Roche Ltd., Mississauga, Canada
| | - Linxinyu Xu
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Enrique Gomez Alcaide
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Nicolas Städler
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Nicole Justies
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Christophe Boetsch
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Vaios Karanikas
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, Schlieren, Switzerland
| | - Gabriel Schnetzler
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Kristoffer S. Rohrberg
- Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Assi T, Moussa T, Ngo C, Faron M, Verret B, Lévy A, Honoré C, Hénon C, Péchoux CL, Bahleda R, Vibert J, Cesne AL. Therapeutic advances in Tenosynovial giant cell Tumor: Targeting the CSF1/CSF1R axis. Cancer Treat Rev 2025; 134:102904. [PMID: 40020639 DOI: 10.1016/j.ctrv.2025.102904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025]
Abstract
Tenosynovial giant cell tumor is a non-malignant primary locally aggressive articular disease that affects the synovium of joints, tendon sheaths, and bursae. It is characterized by a translocation t (1;2), leading to the overexpression of CSF1 in the tumor microenvironment. CSF1 induces the recruitment of non-malignant cells, mainly macrophages, followed by the differentiation and polarization of these cells into the M2 phenotype. Surgery, particularly total synovectomy, remains the cornerstone of TGCT management. However, recurrence rates vary, reaching 40 to 60% in diffuse disease, often resulting in progressive joint dysfunction, pain, and potential need for joint replacement or limb amputation. Systemic therapy is recommended in recurrent TGCT in patients not amenable to additional surgery. Targeting the CSF1/CSF1R axis has successfully improved tumor responses and enhanced symptomatic function. In this review, we aim to explore contemporary paradigms in inoperable TGCT patients, with a focus on the physiopathology, clinical efficacy, and safety of CSF1 or CSF1R inhibitors.
Collapse
Affiliation(s)
- Tarek Assi
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France; Radiology Department, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Tania Moussa
- Radiology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Carine Ngo
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Matthieu Faron
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Benjamin Verret
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Antonin Lévy
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Charles Honoré
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Clémence Hénon
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | | | | | - Julien Vibert
- Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| | - Axel Le Cesne
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France; Sarcoma Unit, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
12
|
Khizar H, Ali K, Wang J. From silent partners to potential therapeutic targets: macrophages in colorectal cancer. Cancer Immunol Immunother 2025; 74:121. [PMID: 39998578 PMCID: PMC11861851 DOI: 10.1007/s00262-025-03965-w] [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: 11/15/2024] [Accepted: 01/30/2025] [Indexed: 02/27/2025]
Abstract
Cancer cells grow and survive in the tumor microenvironment, which is a complicated process. As a key part of how colorectal cancer (CRC) progresses, tumor-associated macrophages (TAMs) exhibit a double role. Through angiogenesis, this TAM can promote the growth of cancers. Although being able to modify and adjust immune cells is a great advantage, these cells can also exhibit anti-cancer properties including direct killing of cancer cells, presenting antigens, and aiding T cell-mediated responses. The delicate regulatory mechanisms between the immune system and tumors are composed of a complex network of pathways regulated by several factors including hypoxia, metabolic reprogramming, cytokine/chemokine signaling, and cell interactions. Decoding and figuring out these complex systems become significant in building targeted treatment programs. Targeting TAMs in CRC involves disrupting chemokine signaling or adhesion molecules, reprogramming them to an anti-tumor phenotype using TLR agonists, CD40 agonists, or metabolic modulation, and selectively removing TAM subsets that promote tumor growth. Multi-drug resistance, the absence of an accurate biomarker, and drug non-specificity are also major problems. Combining macrophage-targeted therapies with chemotherapy and immunotherapy may revolutionize treatment. Macrophage studies will advance with new technology and multi-omics methodologies to help us understand CRC and build specific and efficient treatments.
Collapse
Affiliation(s)
- Hayat Khizar
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Kamran Ali
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310009, China.
| |
Collapse
|
13
|
Dong S, Li X, Huang Q, Li Y, Li J, Zhu X, Xue C, Chen R, Zeng Y, Wu J, Zhong Y, Hu S. Resistance to immunotherapy in non-small cell lung cancer: Unraveling causes, developing effective strategies, and exploring potential breakthroughs. Drug Resist Updat 2025; 81:101215. [PMID: 40081220 DOI: 10.1016/j.drup.2025.101215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Over the last two decades, advancements in deciphering the intricate interactions between oncology and immunity have fueled a meteoric rise in immunotherapy for non-small cell lung cancer, typified by an explosive growth of immune checkpoint inhibitors. However, resistance to immunotherapy remains inevitable. Herein we unravel the labyrinthine mechanisms of resistance to immunotherapy, characterized by their involvement of nearly all types of cells within the body, beyond the extrinsic cancer cells, and importantly, such cells are not only (inhibitory or excitatory, or both) signal recipients but also producers, acting in a context-dependent manner. At the molecular level, these mechanisms underlie genetic and epigenetic aberrations, which are regulated by or regulate various protein kinases, growth factors, and cytokines with inherently dynamic and spatially heterogeneous properties. Additionally, macroscopic factors such as nutrition, comorbidities, and the microbiome within and around organs or tumor cells are involved. Therefore, developing therapeutic strategies combined with distinct action informed by preclinical, clinical, and real-world evidence, such as radiotherapy, chemotherapy, targeted therapy, antibody-drug conjugates, oncolytic viruses, and cell-based therapies, may stand as a judicious reality, although the ideality is to overcome resistance point-by-point through a novel drug. Notably, we highlight a realignment of treatment aims, moving the primary focus from eliminating cancer cells -- such as through chemotherapy and radiotherapy -- to promoting immune modulation and underscore the value of regulating various components within the host macro- or micro-environment, as their effects, even if seemingly minimal, can cumulatively contribute to visible clinical benefit when applied in combination with ICIs. Lastly, this review also emphasizes the current hurdles scattered throughout preclinical and clinical studies, and explores evolving directions in the landscape of immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Shuang Dong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Xiaoyu Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Qing Huang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuanxiang Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | | | - Xianmin Zhu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Chang Xue
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Runzhi Chen
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuan Zeng
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Jingyi Wu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yi Zhong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| |
Collapse
|
14
|
Naei VY, Tubelleza R, Monkman J, Sadeghirad H, Donovan ML, Blick T, Wicher A, Bodbin S, Viratham A, Stad R, Basu S, Cooper C, Barnett C, O'Byrne K, Ladwa R, Warkiani ME, Hughes BGM, Kulasinghe A. Spatial interaction mapping of PD-1/PD-L1 in head and neck cancer reveals the role of macrophage-tumour barriers associated with immunotherapy response. J Transl Med 2025; 23:177. [PMID: 39939997 PMCID: PMC11818323 DOI: 10.1186/s12967-025-06186-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Mucosal head and neck squamous cell carcinoma (HNSCC) is often diagnosed at an advanced stage, where the prognosis is poor due to the high rates of recurrence and metastasis. With approximately one million new cases projected in 2024, worldwide mortality of HNSCC is estimated to reach 50% of detected cases the same year. Patients with early-stage tumours showed a 50-60% five-year survival rate in the US. Immune checkpoint inhibitors (ICIs) have shown promising results in prolonging survival in a subset of patients with recurrent or metastatic disease. However, challenges remain, particularly the limited efficacy of PD-1/PD-L1 blockade therapies. PD-L1 protein expression has been shown to be limited in its predictive power for ICI therapies. Emerging evidence shows that intricate characterisation of the tumour microenvironment (TME) is fundamental to understand interacting cells. This study aims to bridge the gap in understanding the tumor microenvironment by identifying distinct spatial patterns of PD-1/PD-L1 interactions and their association with immunotherapy responses in head and neck squamous cell carcinoma (HNSCC). METHODS In this study, we sought to apply a more nuanced approach to understanding cellular interactions by mapping PD-1/PD-L1 interactions across whole-slide HNSCC tissue samples collected prior to ICI therapy. We used a combination of spatial proteomics (Akoya Biosciences) and an in situ proximity ligation assay (isPLA, Navinci Diagnostics) to visualise PD-1/PD-L1 interactions across cell types and cellular neighbourhoods within the tumour TME. RESULTS Our findings indicate the existence of isPLA+ PD-1/PD-L1 interactions between macrophages/CD3 T cell-enriched neighbourhoods and tumour cells at the tumour-stroma boundaries in ICI-resistant tumours. The presence of these dense macrophage-tumour layers, which are either absent or dispersed in responders, indicates a barrier that may restrict immune cell infiltration and promote immune escape mechanisms. In contrast, responders had abundant B and T cell aggregates, predominantly around the tumour edges linked to enhanced immune responses to ICI therapy and better clinical outcomes. CONCLUSION This study highlights the utility of isPLA in detecting distinct tumour-immune interactions within the TME, offering new cellular interaction metrics for stratifying and optimising immunotherapy strategies.
Collapse
Affiliation(s)
- Vahid Yaghoubi Naei
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Rafael Tubelleza
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Brisbane, Australia
| | - James Monkman
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Brisbane, Australia
| | - Habib Sadeghirad
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Meg L Donovan
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Brisbane, Australia
| | - Tony Blick
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | | | | | | | | | | | | | | | - Ken O'Byrne
- The Princess Alexandra Hospital, Brisbane, Australia
| | - Rahul Ladwa
- The Princess Alexandra Hospital, Brisbane, Australia
| | | | - Brett G M Hughes
- The Royal Brisbane and Women's Hospital, Brisbane, Australia
- School of Medicine, University of Queensland, Brisbane, Australia
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Brisbane, Australia.
| |
Collapse
|
15
|
Shang Q, Zhang P, Lei X, Du L, Qu B. Insights into CSF-1/CSF-1R signaling: the role of macrophage in radiotherapy. Front Immunol 2025; 16:1530890. [PMID: 40007537 PMCID: PMC11851012 DOI: 10.3389/fimmu.2025.1530890] [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: 11/19/2024] [Accepted: 01/13/2025] [Indexed: 02/27/2025] Open
Abstract
Macrophage plays an important role in homeostasis and immunity, and dysfunctional macrophage polarization is believed to be associated with the pathogenesis of tissue fibrosis and tumor progression. Colony stimulating factor-1 (CSF-1), a polypeptide chain cytokine, through its receptor (CSF-1R) regulates the differentiation of macrophages. Recently, the promising therapeutic potential of CSF-1/CSF-1R signaling pathway inhibition in cancer treatment is widely used. Furthermore, inhibition of CSF-1/CSF-1R signaling combined with radiotherapy has been extensively studied to reduce immunosuppression and promote abscopal effect. In addition, cumulative evidence demonstrated that M2 phenotype macrophage is dominant in tissue fibrosis and the inhibition of CSF-1/CSF-1R signaling pathway ameliorated pulmonary fibrosis, including radiation-induced lung fibrosis. Herein, we provide a comprehensive review of the CSF-1/CSF-1R signaling pathway in radiotherapy, with a focus on advances in macrophage-targeted strategies in the treatment of cancer and pulmonary fibrosis.
Collapse
Affiliation(s)
- Qingchao Shang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Radiation Oncology, General Hospital of Southern Theatre Command of PLA, Guang Zhou, China
| | - Pei Zhang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao Lei
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lehui Du
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Baolin Qu
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
16
|
Shah M, Hussain M, Woo HG. Structural insights into antibody-based immunotherapy for hepatocellular carcinoma. Genomics Inform 2025; 23:1. [PMID: 39833954 PMCID: PMC11744992 DOI: 10.1186/s44342-024-00033-0] [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: 10/24/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of primary liver cancer and remains a leading cause of cancer-related deaths worldwide. While traditional approaches like surgical resection and tyrosine kinase inhibitors struggle against the tumor's immune evasion, monoclonal antibody (mAb)-based immunotherapies have emerged as promising alternatives. Several therapeutic antibodies that counter the immunosuppressive tumor microenvironment have demonstrated efficacy in clinical trials, leading to FDA approvals for advanced HCC treatment. A crucial aspect of advancing these therapies lies in understanding the structural interactions between antibodies and their targets. Recent findings indicate that mAbs and bispecific antibodies (bsAbs) can target different, non-overlapping epitopes on immune checkpoints such as PD-1 and CTLA-4. This review delves into the epitope-paratope interactions of structurally unresolved mAbs and bsAbs, and discusses the potential for combination therapies based on their non-overlapping epitopes. By leveraging this unique feature, combination therapies could enhance immune activation, reduce resistance, and improve overall efficacy, marking a new direction for antibody-based immunotherapy in HCC.
Collapse
Affiliation(s)
- Masaud Shah
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Muhammad Hussain
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
- Department of Biomedical Science, Graduate School, Ajou University, Suwon, 16499, Republic of Korea
| | - Hyun Goo Woo
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea.
- Department of Biomedical Science, Graduate School, Ajou University, Suwon, 16499, Republic of Korea.
- Ajou Translational Omics Center (ATOC), Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon, Republic of Korea.
| |
Collapse
|
17
|
Sui XY, Cao SW, Song XQ, Liu XY, Chen C, Yan Q, Wang ZQ, Zhang WJ, Ma LX, Jin X, Ma D, Xiao Y, Wu SY, Xu Y, Shao ZM, Fan L. MORF4L2 induces immunosuppressive microenvironment and immunotherapy resistance through GRHL2/MORF4L2/H4K12Ac/CSF1 axis in triple-negative breast cancer. Biomark Res 2025; 13:6. [PMID: 39780291 PMCID: PMC11715975 DOI: 10.1186/s40364-024-00719-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Although immunotherapy has achieved great progress in advanced triple-negative breast cancer (TNBC), there are still numerous patients who do not benefit from immunotherapy. Therefore, identification of the key molecule that induces immune escape and clarification of its specific mechanism in TNBC are urgently needed. METHODS In this research, single cell sequencing and bulk sequencing were conducted for biomarker screening. Immunohistochemistry, multiplex immunofluorescence, and orthotopic TNBC tumor model were applied in identifying the key molecule driving immune escape. At the mechanical level, RNA sequencing, in vitro co-culturing system, flow cytometry, Western blotting, ELISA, and real-time qPCR were carried out. RESULTS Mortality factor 4 like 2 (MORF4L2) expression is significantly up-regulated among patients who developed anti-PD1 resistance. MORF4L2 enhances anti-PD1 resistance by inducing the chemotaxis of macrophage infiltration and promoting their polarization towards the alternative activation phenotype (M2), thus creating an immunosuppressive microenvironment. Mechanistically, MORF4L2 actes as part of NuA4 histone acetyltransferase (HAT) complex, contributes to to histone 4 lysine 12 acetylation (H4K12Ac) and activates the downstream transcription of macrophage colony-stimulating factor (CSF1). CSF1 is secreted by tumor cells and binds to the macrophage-surface CSF1 receptor (CSF1R), which chemotactically converted and polarized macrophages to the M2 phenotype. Furthermore, we revealed that grainyhead like transcription factor 2 (GRHL2) could promote MORF4L2 transcription by binding to the MORF4L2 enhancer region. Notably, BLZ549, an inhibitor of CSF1R, restored the anti-PD1 sensitivity by blocking the GRHL2/MORF4L2/H4K12Ac/CSF1 axis. CONCLUSIONS GRHL2/MORF4L2/H4K12Ac/CSF1 axis plays an important role in anti-PD1 resistance. CSF1R inhibitors can reverse GRHL2/MORF4L2-mediated anti-PD1 resistance.
Collapse
Affiliation(s)
- Xin-Yi Sui
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuo-Wen Cao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Qing Song
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi-Yu Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chao Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qingya Yan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zhi-Qing Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Juan Zhang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin-Xiaoxi Ma
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi Jin
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding Ma
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Xiao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Song-Yang Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Xu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Lei Fan
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Kong R, Huang J, Wu Y, Yan N, Chen X, Cheng H. Tricomponent immunoactivating nanomedicine to downregulate PD-L1 and polarize macrophage for photodynamic immunotherapy of colorectal cancer. Int J Pharm 2025; 668:124968. [PMID: 39561904 DOI: 10.1016/j.ijpharm.2024.124968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/05/2024] [Accepted: 11/16/2024] [Indexed: 11/21/2024]
Abstract
The unsatisfactory immunotherapeutic responses are primarily attributed to the insufficient immune recognition and the presence of an immunosuppressive tumor microenvironment (ITM). This study focuses on the development of a tricomponent immunoactivating nanomedicine called TIN that combines a photosensitizer, an inhibitor of epidermal growth factor receptor (EGFR) and a CSF-1R inhibitor to enable photodynamic immunotherapy by downregulating PD-L1 expression and repolarizing tumor-associated macrophages (TAMs). TIN is designed to facilitate the drug delivery and target specific pathways involved in tumor progression. By inhibiting the activity of EGFR and CSF-1R, TIN reduces PD-L1 expression on tumor cells and induces the TAMs polarization to M1 phenotype, restoring the immune recognition of T cells and the phagocytosis of macrophage to reshape the immunosuppressive microenvironment. Additionally, the photodynamic therapy (PDT) of TIN can greatly destroy the primary tumor and trigger immunogenic cell death (ICD). Importantly, the immune checkpoint blockade effect of TIN can enhance the immune response of PDT-induced ICD for metastatic tumor treatment. This study presents a self-assembling strategy for the development of an all-in-one nanomedicine, effectively integrating multiple therapeutic modalities to provide a comprehensive and systemic approach for tumor suppression.
Collapse
Affiliation(s)
- Renjiang Kong
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Jiaqi Huang
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Yeyang Wu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Ni Yan
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.
| | - Hong Cheng
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
19
|
Singer M, Zhang Z, Dayyani F, Zhang Z, Yaghmai V, Choi A, Valerin J, Imagawa D, Abi-Jaoudeh N. Modulation of Tumor-Associated Macrophages to Overcome Immune Suppression in the Hepatocellular Carcinoma Microenvironment. Cancers (Basel) 2024; 17:66. [PMID: 39796695 PMCID: PMC11718901 DOI: 10.3390/cancers17010066] [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: 12/16/2024] [Revised: 12/21/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a major global health issue characterized by poor prognosis and complex tumor biology. One of the critical components of the HCC tumor microenvironment (TME) is tumor-associated macrophages (TAMs), which play a pivotal role in modulating tumor growth, immune evasion, and metastasis. Macrophages are divided into two major subtypes: pro-inflammatory M1 and anti-inflammatory M2, both of which may exist in TME with altered function and proportion. The anti-inflammatory M2 macrophages are further subdivided into four distinct immune suppressive subsets. TAMs are generally counted as M2-like macrophages with altered immune suppressive functions that exert a significant influence on both cancer progression and the ability of tumors to escape immune surveillance. Their involvement in modulating immune responses via different mechanisms at the local and systemic levels has made them a key target for therapeutic interventions seeking to enhance treatment outcomes. How TAMs' depletion influences immune responses in cancer is the primary interest in cancer immunotherapies. The purpose of this review is to delve into the recent progress made in TAM-targeting therapies. We will explore the current theories, benefits, and challenges associated with TAMs' depletion or inhibition. The manuscript concludes with future directions and potential implications for clinical practice.
Collapse
Affiliation(s)
- Mahmoud Singer
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| | - Zhuoli Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| | - Farshid Dayyani
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92867, USA; (F.D.); (A.C.); (J.V.)
| | - Zigeng Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| | - Vahid Yaghmai
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| | - April Choi
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92867, USA; (F.D.); (A.C.); (J.V.)
| | - Jennifer Valerin
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92867, USA; (F.D.); (A.C.); (J.V.)
| | - David Imagawa
- Department of Surgery, University of California Irvine, Orange, CA 92697, USA;
| | - Nadine Abi-Jaoudeh
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA; (Z.Z.); (Z.Z.); (V.Y.)
| |
Collapse
|
20
|
Zheng H, Yang X, Huang N, Yuan S, Li J, Liu X, Jiang Q, Wu S, Ju Y, Kleeff J, Yin X, Liao Q, Liu Q, Zhao Y. Chimeric antigen receptor macrophages targeting c-MET(CAR-M-c-MET) inhibit pancreatic cancer progression and improve cytotoxic chemotherapeutic efficacy. Mol Cancer 2024; 23:270. [PMID: 39643883 PMCID: PMC11622543 DOI: 10.1186/s12943-024-02184-8] [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: 05/02/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant tumors. Macrophages are abundant in the tumor microenvironment, making them an attractive target for therapeutic intervention. While current immunotherapies, including immune checkpoint inhibition (ICI) and chimeric antigen receptor T (CAR-T) cells, have shown limited efficacy in pancreatic cancer, a novel approach involving chimeric antigen receptor macrophages (CAR-M) has, although promising, not been explored in pancreatic cancer. In this study, we first investigated the role of CAR-M cells targeting c-MET in pancreatic cancer. METHODS The effectiveness and rationality of c-MET as a target for CAR-M in pancreatic cancer were validated through bioinformatic analyses and immunohistochemical staining of samples from pancreatic cancer patients. We utilized flow cytometry and bioluminescence detection methods to demonstrate the specific binding and phagocytic killing effect of CAR-M on pancreatic cancer cells. Additionally, we observed the process of CAR-M engulfing pancreatic cancer cells using confocal microscopy and a long-term fluorescence live cell imaging system. In an in situ tumor model transplanted into NOD/SCID mice, we administered intraperitoneal injections of CAR-M to confirm its inhibitory function on pancreatic cancer. Furthermore, we validated these findings in human monocyte-derived macrophages (hMDM). RESULTS Bioinformatics and tumor tissue microarray analyses revealed significantly higher expression levels of c-MET in tumor tissues, compared to the paired peritumoral tissues, and higher c-MET expression correlated with worse patient survival. CAR-M cells were engineered using human monocytic THP-1 cell line and hMDM targeting c-MET (CAR-M-c-MET). The CAR-M-c-MET cells demonstrated highly specific binding to pancreatic cancer cells and exhibited more phagocytosis and killing abilities than the pro-inflammatory polarized control macrophages. In addition, CAR-M-c-MET cells synergized with various cytotoxic chemotherapeutic drugs. In a NOD/SCID murine model, intraperitoneally injected CAR-M-c-MET cells rapidly migrated to tumor tissue and substantially inhibited tumor growth, which did not lead to obvious side effects. Cytokine arrays and mRNA sequencing showed that CAR-M-c-MET produced higher levels of immune activators than control macrophages. CONCLUSIONS This study provides compelling evidence for the safety and efficacy of CAR-M therapy in treating pancreatic cancer. The results demonstrate that CAR-M-c-MET significantly suppresses pancreatic cancer progression and enhances the effectiveness of cytotoxic chemotherapy. Remarkably, no discernible side effects occur. Further clinical trials are warranted in human pancreatic cancer patients.
Collapse
Affiliation(s)
- Huaijin Zheng
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Xinzhe Yang
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Nan Huang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Shangqin Yuan
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Jiayi Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Xudong Liu
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Qing Jiang
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Shanshan Wu
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Yue Ju
- Roc Rock Biotechnology (Shenzhen), Shenzhen, 518118, China
| | - Jorg Kleeff
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - Xiushan Yin
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China.
- Roc Rock Biotechnology (Shenzhen), Shenzhen, 518118, China.
| | - Quan Liao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China.
| | - Qiaofei Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China.
| |
Collapse
|
21
|
Ma Y, Sun Y, Guo H, Yang R. Tumor-associated macrophages in bladder cancer: roles and targeted therapeutic strategies. Front Immunol 2024; 15:1418131. [PMID: 39606239 PMCID: PMC11599180 DOI: 10.3389/fimmu.2024.1418131] [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: 04/16/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Bladder cancer (BC) is the ninth most common and "expensive" cancer in the world. Despite the availability of various treatment modalities such as chemotherapy, immunotherapy and surgery, the overall survival rate of patients with advanced bladder cancer remains low. As one of the most abundant infiltrating immune cells in bladder cancer, tumor-associated macrophages (TAMs) play an important role in the development of BC and in the standard regimen of intravesical BCG therapy. Targeting TAMs have achieved excellent results in clinical trials for a variety of other cancers, but few studies have been conducted for bladder cancer. Further exploration is still needed to develop TAM-related therapeutic strategies for BC treatment, which are expected to improve the therapeutic efficacy and life quality of patients. This review summarizes the relationship between TAMs in bladder cancer and disease staging, evolution, patient prognosis, and treatment outcome. Several potential TAM targets in BC are also pointed, which may help to inhibit tumor-promoting TAMs and provide new therapeutic approaches for advanced BC.
Collapse
Affiliation(s)
- Yuanchun Ma
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ying Sun
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Urology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University Advanced Institute of Life Sciences (NAILS), Nanjing, China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
22
|
Horino T, Horiguchi H, Yumoto S, Kadomatsu T, Hara Y, Yagi T, Baba Y, Miyamoto Y, Baba H, Oike Y. Angiopoietin-Like Protein 2 Expression in Tumor Cells Supports Tumor-Associated Macrophage-Induced Tumor Progression in Esophageal Cancer. Ann Surg Oncol 2024; 31:7693-7704. [PMID: 38981990 DOI: 10.1245/s10434-024-15557-6] [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: 02/13/2024] [Accepted: 05/21/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Tumor-associated macrophages (TAM), a major component of the tumor microenvironment, play key roles in tumor formation and progression; however, mechanisms underlying TAM-induced tumor progression are complex and not well known. We previously reported that tumor cell-derived angiopoietin-like protein 2 (ANGPTL2) functions as a tumor promoter in some cancer contexts. METHODS We examined ANGPTL2 expression in paraffin-embedded tumor samples from resected specimens of 221 patients with esophageal cancer. Patients were subdivided into four groups based on immunohistochemistry scores described above: ANGPTL2-low/TAM-low, ANGPTL2-low/TAM-high, ANGPTL2-high/TAM-low, and ANGPTL2-high/TAM-high groups. Gene expression datasets of esophageal cancer cell lines were obtained from the cancer cell line encyclopedia public database. RESULTS In this study, we demonstrate that TAM infiltration is associated with poor prognosis in patients with esophageal cancer whose tumor cells show relatively higher ANGPTL2 expression levels; however, TAM infiltration did not affect prognosis in patients with ANGPTL2-low-expressing esophageal cancer, suggesting that ANGPTL2 expression in esophageal cancer cells is required for TAM-induced tumor progression. Our analysis of public datasets indicates a potential positive correlation of ANGPTL2 expression levels with that of transforming growth factor (TGF)-β, a TAM-activating factor, in esophageal cancer cell lines. CONCLUSION We conclude that ANGPTL2 signaling in tumor cells supports TAM-induced tumor progression and contributes to poor prognosis in patients with esophageal cancer. These findings overall provide novel insight into pro-tumor ANGPTL2 functions and illustrate the essential role of cancer cell/TAM crosstalk in cancer progression.
Collapse
Affiliation(s)
- Taichi Horino
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan.
- Department of Aging and Geriatric Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Shinsei Yumoto
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Hara
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taisuke Yagi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Surgery, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Division of Translational Research and Advanced Treatment Against Gastrointestinal Cancer, Kumamoto University Hospital, Kumamoto, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan.
- Department of Aging and Geriatric Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
23
|
Wang L, Zhang L, Zhang Z, Wu P, Zhang Y, Chen X. Advances in targeting tumor microenvironment for immunotherapy. Front Immunol 2024; 15:1472772. [PMID: 39421736 PMCID: PMC11484021 DOI: 10.3389/fimmu.2024.1472772] [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: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The tumor microenvironment (TME) provides essential conditions for the occurrence, invasion, and spread of cancer cells. Initial research has uncovered immunosuppressive properties of the TME, which include low oxygen levels (hypoxia), acidic conditions (low pH), increased interstitial pressure, heightened permeability of tumor vasculature, and an inflammatory microenvironment. The presence of various immunosuppressive components leads to immune evasion and affects immunotherapy efficacy. This indicates the potential value of targeting the TME in cancer immunotherapy. Therefore, TME remodeling has become an effective method for enhancing host immune responses against tumors. In this study, we elaborate on the characteristics and composition of the TME and how it weakens immune surveillance and summarize targeted therapeutic strategies for regulating the TME.
Collapse
Affiliation(s)
- Lugang Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liubo Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Wu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
24
|
Wang Q, Wang J, Xu K, Luo Z. Targeting the CSF1/CSF1R signaling pathway: an innovative strategy for ultrasound combined with macrophage exhaustion in pancreatic cancer therapy. Front Immunol 2024; 15:1481247. [PMID: 39416792 PMCID: PMC11479911 DOI: 10.3389/fimmu.2024.1481247] [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: 08/15/2024] [Accepted: 09/11/2024] [Indexed: 10/19/2024] Open
Abstract
Pancreatic cancer (PC) is a highly aggressive and lethal malignancy characterized by a complex tumor microenvironment (TME) and immunosuppressive features that limit the efficacy of existing treatments. This paper reviews the potential of combining ultrasound with macrophage exhaustion in the treatment of pancreatic cancer. Macrophages, particularly tumor-associated macrophages (TAMs), are crucial in pancreatic cancer progression and immune escape. Prolonged exposure to the immunosuppressive TME leads to macrophage exhaustion, reducing their anti-tumor ability and instead promoting tumor growth. The CSF1/CSF1R signaling pathway is key in macrophage recruitment and functional regulation, making it an effective target for combating macrophage exhaustion. Ultrasound technology not only plays a significant role in diagnosis and staging but also enhances therapeutic efficacy by guiding radiofrequency ablation (RFA) and percutaneous alcohol injection (PEI) in combination with immunomodulators. Additionally, ultrasound imaging can monitor the number and functional status of TAMs in real-time, providing a basis for optimizing treatment strategies. Future studies should further investigate the combined use of ultrasound and immunomodulators to refine treatment regimens, address challenges such as individual variability and long-term effects, and offer new hope for pancreatic cancer patients.
Collapse
Affiliation(s)
- Qian Wang
- Department of Ultrasound, Xichong People’s Hospital, Nanchong, China
| | - Jianhong Wang
- Department of Internal Medicine, Guang’an Vocational & Technical College, Guang’an, China
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Zhibin Luo
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| |
Collapse
|
25
|
Jiménez-Andrade Y, Flesher JL, Park JM. Cancer Therapy-induced Dermatotoxicity as a Window to Understanding Skin Immunity. Hematol Oncol Clin North Am 2024; 38:1011-1025. [PMID: 38866636 PMCID: PMC11368641 DOI: 10.1016/j.hoc.2024.05.002] [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] [Indexed: 06/14/2024]
Abstract
Pruritus, rash, and various other forms of dermatotoxicity are the most frequent adverse events among patients with cancer receiving targeted molecular therapy and immunotherapy. Immune checkpoint inhibitors, macrophage-targeting agents, and epidermal growth factor receptor/MEK inhibitors not only exert antitumor effects but also interfere with molecular pathways essential for skin immune homeostasis. Studying cancer therapy-induced dermatotoxicity helps us identify molecular mechanisms governing skin immunity and deepen our understanding of human biology. This review summarizes new mechanistic insights emerging from the analysis of cutaneous adverse events and discusses knowledge gaps that remain to be closed by future research.
Collapse
Affiliation(s)
- Yanek Jiménez-Andrade
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA
| | - Jessica L Flesher
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA
| | - Jin Mo Park
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Charlestown, MA 02129, USA.
| |
Collapse
|
26
|
Chen F, Sheng J, Li X, Gao Z, Hu L, Chen M, Fei J, Song Z. Tumor-associated macrophages: orchestrators of cholangiocarcinoma progression. Front Immunol 2024; 15:1451474. [PMID: 39290697 PMCID: PMC11405194 DOI: 10.3389/fimmu.2024.1451474] [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: 06/19/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a rare but highly invasive cancer, with its incidence rising in recent years. Currently, surgery remains the most definitive therapeutic option for CCA. However, similar to other malignancies, most CCA patients are not eligible for surgical intervention at the time of diagnosis. The chemotherapeutic regimen of gemcitabine combined with cisplatin is the standard treatment for advanced CCA, but its effectiveness is often hampered by therapeutic resistance. Recent research highlights the remarkable plasticity of tumor-associated macrophages (TAMs) within the tumor microenvironment (TME). TAMs play a crucial dual role in either promoting or suppressing tumor development, depending on the factors that polarize them toward pro-tumorigenic or anti-tumorigenic phenotypes, as well as their interactions with cancer cells and other stromal components. In this review, we critically examine recent studies on TAMs in CCA, detailing the expression patterns and prognostic significance of different TAM subtypes in CCA, the mechanisms by which TAMs influence CCA progression and immune evasion, and the potential for reprogramming TAMs to enhance anticancer therapies. This review aims to provide a framework for deeper future research.
Collapse
Affiliation(s)
- Fei Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jian Sheng
- Department of Research and Teaching, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaoping Li
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zhaofeng Gao
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lingyu Hu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Minjie Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jianguo Fei
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zhengwei Song
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
27
|
Zhang Y, Ding X, Zhang X, Li Y, Xu R, Li HJ, Zuo D, Chen G. Unveiling the contribution of tumor-associated macrophages in driving epithelial-mesenchymal transition: a review of mechanisms and therapeutic Strategies. Front Pharmacol 2024; 15:1404687. [PMID: 39286635 PMCID: PMC11402718 DOI: 10.3389/fphar.2024.1404687] [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: 03/21/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs), fundamental constituents of the tumor microenvironment (TME), significantly influence cancer development, primarily by promoting epithelial-mesenchymal transition (EMT). EMT endows cancer cells with increased motility, invasiveness, and resistance to therapies, marking a pivotal juncture in cancer progression. The review begins with a detailed exposition on the origins of TAMs and their functional heterogeneity, providing a foundational understanding of TAM characteristics. Next, it delves into the specific molecular mechanisms through which TAMs induce EMT, including cytokines, chemokines and stromal cross-talking. Following this, the review explores TAM-induced EMT features in select cancer types with notable EMT characteristics, highlighting recent insights and the impact of TAMs on cancer progression. Finally, the review concludes with a discussion of potential therapeutic targets and strategies aimed at mitigating TAM infiltration and disrupting the EMT signaling network, thereby underscoring the potential of emerging treatments to combat TAM-mediated EMT in cancer. This comprehensive analysis reaffirms the necessity for continued exploration into TAMs' regulatory roles within cancer biology to refine therapeutic approaches and improve patient outcomes.
Collapse
Affiliation(s)
- Yijia Zhang
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaofei Ding
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| | - Xue Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ye Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Rui Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Hai-Jun Li
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Guang Chen
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
28
|
Wu X, Stabile LP, Burns TF. The Emerging Role of Immune Checkpoint Blockade for the Treatment of Lung Cancer Brain Metastases. Clin Lung Cancer 2024; 25:483-501. [PMID: 38991863 DOI: 10.1016/j.cllc.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/15/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024]
Abstract
Lung cancer has the highest incidence of brain metastases (BM) among solid organ cancers. Traditionally whole brain radiation therapy has been utilized for non-small-cell lung cancer (NSCLC) BM treatment, although stereotactic radiosurgery has emerged as the superior treatment modality for most patients. Highly penetrant central nervous system (CNS) tyrosine kinase inhibitors have also shown significant CNS activity in patients harboring select oncogenic drivers. There is emerging evidence that patients without oncogene-driven tumors derive benefit from the use of immune checkpoint inhibitors (ICIs). The CNS activity of ICIs have not been well studied given exclusion of patients with active BM from landmark trials, due to concerns of inadequate CNS penetration and activity. However, studies have challenged the idea of an immune-privileged CNS, given the presence of functional lymphatic drainage within the CNS and destruction of the blood brain barrier by BM. An emerging understanding of the interactions between tumor and CNS immune cells in the BM tumor microenvironment also support a role for immunotherapy in BM treatment. In addition, posthoc analyses of major trials have shown improved intracranial response and survival benefit of regimens with ICIs over chemotherapy (CT) alone for patients with BM. Two prospective phase 2 trials evaluating pembrolizumab monotherapy and atezolizumab plus CT in patients with untreated NSCLC BM also demonstrated significant intracranial responses. This review describes the interplay between CNS immune cells and tumor cells, discusses current evidence for ICI CNS activity from retrospective and prospective studies, and speculates on future directions of investigation.
Collapse
Affiliation(s)
- Xiancheng Wu
- Department of Medicine, Division of Internal Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Laura P Stabile
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA; UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Timothy F Burns
- UPMC Hillman Cancer Center, Pittsburgh, PA; Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh, Pittsburgh, PA.
| |
Collapse
|
29
|
Minaei E, Ranson M, Aghmesheh M, Sluyter R, Vine KL. Enhancing pancreatic cancer immunotherapy: Leveraging localized delivery strategies through the use of implantable devices and scaffolds. J Control Release 2024; 373:145-160. [PMID: 38996923 DOI: 10.1016/j.jconrel.2024.07.023] [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: 04/11/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Pancreatic cancer (PC) remains the predominant type of upper gastrointestinal tract cancer, associated with heightened morbidity and a survival rate below 12%. While immunotherapy has brought about transformative changes in the standards of care for most solid tumors, its application in PC is hindered by the ''cold tumor'' microenvironment, marked by the presence of immunosuppressive cells. Modest response rates in PC are attributed, in part to, the fibrotic stroma that obstructs the delivery of systemic immunotherapy. Furthermore, the occurrence of immune-related adverse events (iRAEs) often necessitates the use of sub-therapeutic doses or treatment discontinuation. In the pursuit of innovative approaches to enhance the effectiveness of immunotherapy for PC, implantable drug delivery devices and scaffolds emerge as promising strategies. These technologies offer the potential for sustained drug delivery directly to the tumor site, overcoming stromal barriers, immunosuppression, T cell exclusion, immunotherapy resistance, optimizing drug dosage, and mitigating systemic toxicity. This review offers a comprehensive exploration of pancreatic ductal adenocarcinoma (PDAC), the most common and aggressive form of PC, accompanied by a critical analysis of the challenges the microenvironment presents to the development of successful combinational immunotherapy approaches. Despite efforts, these approaches have thus far fallen short in enhancing treatment outcomes for PDAC. The review will subsequently delve into the imperative need for refining delivery strategies, providing an examination of past and ongoing studies in the field of localized immunotherapy for PDAC. Addressing these issues will lay the groundwork for the development of effective new therapies, thereby enhancing treatment response, patient survival, and overall quality of life for individuals diagnosed with PDAC.
Collapse
Affiliation(s)
- E Minaei
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia.
| | - M Ranson
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - M Aghmesheh
- Nelune Comprehensive Cancer Centre, Bright Building, Prince of Wales Hospital, Sydney, NSW, Australia; Faculty of Medicine and Health, UNSW, Sydney, NSW, Australia
| | - R Sluyter
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - K L Vine
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia.
| |
Collapse
|
30
|
Zhang Y, Han G, Gu J, Chen Z, Wu J. Role of tumor-associated macrophages in hepatocellular carcinoma: impact, mechanism, and therapy. Front Immunol 2024; 15:1429812. [PMID: 39170620 PMCID: PMC11335564 DOI: 10.3389/fimmu.2024.1429812] [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: 05/08/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly frequent malignancy worldwide. The occurrence and progression of HCC is a complex process closely related to the polarization of tumor-associated macrophages (TAMs) in the tumor microenvironment (TME). The polarization of TAMs is affected by a variety of signaling pathways and surrounding cells. Evidence has shown that TAMs play a crucial role in HCC, through its interaction with other immune cells in the TME. This review summarizes the origin and phenotypic polarization of TAMs, their potential impacts on HCC, and their mechanisms and potential targets for HCC immunotherapy.
Collapse
Affiliation(s)
- Yinqi Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Guoyong Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Jian Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Zhiqiang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Jindao Wu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| |
Collapse
|
31
|
Habib S, Osborn G, Willsmore Z, Chew MW, Jakubow S, Fitzpatrick A, Wu Y, Sinha K, Lloyd-Hughes H, Geh JLC, MacKenzie-Ross AD, Whittaker S, Sanz-Moreno V, Lacy KE, Karagiannis SN, Adams R. Tumor associated macrophages as key contributors and targets in current and future therapies for melanoma. Expert Rev Clin Immunol 2024; 20:895-911. [PMID: 38533720 PMCID: PMC11286214 DOI: 10.1080/1744666x.2024.2326626] [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: 12/22/2023] [Accepted: 02/29/2024] [Indexed: 03/28/2024]
Abstract
INTRODUCTION Despite the success of immunotherapies for melanoma in recent years, there remains a significant proportion of patients who do not yet derive benefit from available treatments. Immunotherapies currently licensed for clinical use target the adaptive immune system, focussing on Tcell interactions and functions. However, the most prevalent immune cells within the tumor microenvironment (TME) of melanoma are macrophages, a diverse immune cell subset displaying high plasticity, to which no current therapies are yet directly targeted. Macrophages have been shown not only to activate the adaptive immune response, and enhance cancer cell killing, but, when influenced by factors within the TME of melanoma, these cells also promote melanoma tumorigenesis and metastasis. AREAS COVERED We present a review of the most up-to-date literatureavailable on PubMed, focussing on studies from within the last 10 years. We also include data from ongoing and recent clinical trials targeting macrophages in melanoma listed on clinicaltrials.gov. EXPERT OPINION Understanding the multifaceted role of macrophages in melanoma, including their interactions with immune and cancer cells, the influence of current therapies on macrophage phenotype and functions and how macrophages could be targeted with novel treatment approaches, are all critical for improving outcomes for patients with melanoma.
Collapse
Affiliation(s)
- Shabana Habib
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
| | - Gabriel Osborn
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
| | - Zena Willsmore
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
| | - Min Waye Chew
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
| | - Sophie Jakubow
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
| | - Amanda Fitzpatrick
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
- Oncology Department, Guy’s and St Thomas’ Hospital, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Innovation Hub, Guy’s Hospital, London, UK
| | - Yin Wu
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
- Oncology Department, Guy’s and St Thomas’ Hospital, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King’s College London, London, UK
| | - Khushboo Sinha
- St John’s Institute of Dermatology, Guy’s, King’s and St. Thomas’ Hospitals NHS Foundation Trust, London, England
| | - Hawys Lloyd-Hughes
- Department of Plastic Surgery, Guy’s, King’s and St. Thomas’ Hospitals, London, England
| | - Jenny L. C. Geh
- St John’s Institute of Dermatology, Guy’s, King’s and St. Thomas’ Hospitals NHS Foundation Trust, London, England
- Department of Plastic Surgery, Guy’s, King’s and St. Thomas’ Hospitals, London, England
| | | | - Sean Whittaker
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
| | - Victoria Sanz-Moreno
- The Breast Cancer Now Toby Robins Research Centre, Division of Breast Cancer Research, The Institute of Cancer Research, London
| | - Katie E. Lacy
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
| | - Sophia N Karagiannis
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Innovation Hub, Guy’s Hospital, London, UK
| | - Rebecca Adams
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, London, UK
| |
Collapse
|
32
|
Paul S, Konig MF, Pardoll DM, Bettegowda C, Papadopoulos N, Wright KM, Gabelli SB, Ho M, van Elsas A, Zhou S. Cancer therapy with antibodies. Nat Rev Cancer 2024; 24:399-426. [PMID: 38740967 PMCID: PMC11180426 DOI: 10.1038/s41568-024-00690-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
The greatest challenge in cancer therapy is to eradicate cancer cells with minimal damage to normal cells. Targeted therapy has been developed to meet that challenge, showing a substantially increased therapeutic index compared with conventional cancer therapies. Antibodies are important members of the family of targeted therapeutic agents because of their extraordinarily high specificity to the target antigens. Therapeutic antibodies use a range of mechanisms that directly or indirectly kill the cancer cells. Early antibodies were developed to directly antagonize targets on cancer cells. This was followed by advancements in linker technologies that allowed the production of antibody-drug conjugates (ADCs) that guide cytotoxic payloads to the cancer cells. Improvement in our understanding of the biology of T cells led to the production of immune checkpoint-inhibiting antibodies that indirectly kill the cancer cells through activation of the T cells. Even more recently, bispecific antibodies were synthetically designed to redirect the T cells of a patient to kill the cancer cells. In this Review, we summarize the different approaches used by therapeutic antibodies to target cancer cells. We discuss their mechanisms of action, the structural basis for target specificity, clinical applications and the ongoing research to improve efficacy and reduce toxicity.
Collapse
Affiliation(s)
- Suman Paul
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Maximilian F Konig
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Katharine M Wright
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA
| | - Sandra B Gabelli
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA.
| | - Mitchell Ho
- Antibody Engineering Program, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | | | - Shibin Zhou
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
33
|
Bhattacharya A, Dasgupta AK. Multifaceted perspectives of detecting and targeting solid tumors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 389:1-66. [PMID: 39396844 DOI: 10.1016/bs.ircmb.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Solid tumors are the most prevalent form of cancer. Considerable technological and medical advancements had been achieved for the diagnosis of the disease. However, detection of the disease in an early stage is of utmost importance, still far from reality. On the contrary, the treatment and therapeutic area to combat solid tumors are still in its infancy. Conventional treatments like chemotherapy and radiation therapy pose challenges due to their indiscriminate impact on healthy and cancerous cells. Contextually, efficient drug targeting is a pivotal approach in solid tumor treatment. This involves the precise delivery of drugs to cancer cells while minimizing harm to healthy cells. Targeted drugs exhibit superior efficacy in eradicating cancer cells while impeding tumor growth and mitigate side effects by optimizing absorption which further diminishes the risk of resistance. Furthermore, tailoring targeted therapies to a patient's tumor-specific molecular profile augments treatment efficacy and reduces the likelihood of relapse. This chapter discuss about the distinctive characteristics of solid tumors, the possibility of early detection of the disease and potential therapeutic angle beyond the conventional approaches. Additionally, the chapter delves into a hitherto unknown attribute of magnetic field effect to target cancer cells which exploit the relatively less susceptibility of normal cells compared to cancer cells to magnetic fields, suggesting a future potential of magnetic nanoparticles for selective cancer cell destruction. Lastly, bioinformatics tools and other unconventional methodologies such as AI-assisted codon bias analysis have a crucial role in comprehending tumor biology, aiding in the identification of futuristic targeted therapies.
Collapse
Affiliation(s)
- Abhishek Bhattacharya
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Anjan Kr Dasgupta
- Department of Biochemistry, University of Calcutta, Kolkata, West Bengal, India.
| |
Collapse
|
34
|
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
|
35
|
Kuznetsova AB, Kolesova EP, Parodi A, Zamyatnin AA, Egorova VS. Reprogramming Tumor-Associated Macrophage Using Nanocarriers: New Perspectives to Halt Cancer Progression. Pharmaceutics 2024; 16:636. [PMID: 38794298 PMCID: PMC11124960 DOI: 10.3390/pharmaceutics16050636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer remains a significant challenge for public healthcare systems worldwide. Within the realm of cancer treatment, considerable attention is focused on understanding the tumor microenvironment (TME)-the complex network of non-cancerous elements surrounding the tumor. Among the cells in TME, tumor-associated macrophages (TAMs) play a central role, traditionally categorized as pro-inflammatory M1 macrophages or anti-inflammatory M2 macrophages. Within the TME, M2-like TAMs can create a protective environment conducive to tumor growth and progression. These TAMs secrete a range of factors and molecules that facilitate tumor angiogenesis, increased vascular permeability, chemoresistance, and metastasis. In response to this challenge, efforts are underway to develop adjuvant therapy options aimed at reprogramming TAMs from the M2 to the anti-tumor M1 phenotype. Such reprogramming holds promise for suppressing tumor growth, alleviating chemoresistance, and impeding metastasis. Nanotechnology has enabled the development of nanoformulations that may soon offer healthcare providers the tools to achieve targeted drug delivery, controlled drug release within the TME for TAM reprogramming and reduce drug-related adverse events. In this review, we have synthesized the latest data on TAM polarization in response to TME factors, highlighted the pathological effects of TAMs, and provided insights into existing nanotechnologies aimed at TAM reprogramming and depletion.
Collapse
Affiliation(s)
- Alyona B. Kuznetsova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Ekaterina P. Kolesova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Andrey A. Zamyatnin
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Vera S. Egorova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| |
Collapse
|
36
|
Yu Z, Zou J, Xu F. Tumor-associated macrophages affect the treatment of lung cancer. Heliyon 2024; 10:e29332. [PMID: 38623256 PMCID: PMC11016713 DOI: 10.1016/j.heliyon.2024.e29332] [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: 12/26/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024] Open
Abstract
As one of the most common malignant tumors in the world, lung cancer has limited benefits for patients despite its diverse treatment methods due to factors such as personalized medicine targeting histological type, immune checkpoint expression, and driver gene mutations. The high mortality rate of lung cancer is partly due to the immune-suppressive which limits the effectiveness of anti-cancer drugs and induces tumor cell resistance. The currently widely recognized TAM phenotypes include the anti-tumor M1 and pro-tumor M2 phenotypes. M2 macrophages promote the formation of an immune-suppressive microenvironment and hinder immune cell infiltration, thereby inhibiting activation of the anti-tumor immune system and aiding tumor cells in resisting treatment. Analyzing the relationship between different treatment methods and macrophages in the TME can help us better understand the impact of TAMs on lung cancer and confirm the feasibility of targeted TAM therapy. Targeting TAMs to reduce the M2/M1 ratio and reverse the immune-suppressive microenvironment can improve the clinical efficacy of conventional treatment methods and potentially open up more efficient combination treatment strategies, maximizing the benefit for lung cancer patients.
Collapse
Affiliation(s)
- Zhuchen Yu
- Clinical Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Juntao Zou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Fei Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| |
Collapse
|
37
|
Liu J, Yuan Q, Guo H, Guan H, Hong Z, Shang D. Deciphering drug resistance in gastric cancer: Potential mechanisms and future perspectives. Biomed Pharmacother 2024; 173:116310. [PMID: 38394851 DOI: 10.1016/j.biopha.2024.116310] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/07/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Gastric cancer (GC) is a malignant tumor that originates from the epithelium of the gastric mucosa. The latest global cancer statistics show that GC ranks fifth in incidence and fourth in mortality among all cancers, posing a serious threat to public health. While early-stage GC is primarily treated through surgery, chemotherapy is the frontline option for advanced cases. Currently, commonly used chemotherapy regimens include FOLFOX (oxaliplatin + leucovorin + 5-fluorouracil) and XELOX (oxaliplatin + capecitabine). However, with the widespread use of chemotherapy, an increasing number of cases of drug resistance have emerged. This article primarily explores the potential mechanisms of chemotherapy resistance in GC patients from five perspectives: cell death, tumor microenvironment, non-coding RNA, epigenetics, and epithelial-mesenchymal transition. Additionally, it proposes feasibility strategies to overcome drug resistance from four angles: cancer stem cells, tumor microenvironment, natural products, and combined therapy. The hope is that this article will provide guidance for researchers in the field and bring hope to more GC patients.
Collapse
Affiliation(s)
- Jiahua Liu
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qihang Yuan
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui Guo
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hewen Guan
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Zhijun Hong
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Dong Shang
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
38
|
Zhou L, Zhao T, Zhang R, Chen C, Li J. New insights into the role of macrophages in cancer immunotherapy. Front Immunol 2024; 15:1381225. [PMID: 38605951 PMCID: PMC11007015 DOI: 10.3389/fimmu.2024.1381225] [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: 02/03/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Macrophages are the main component of the tumor microenvironment, which are differentiated from monocytes in the blood and play an important role in cancer development. Tumor-associated macrophages (TAMs) can promote tumor growth, invasion, metastasis, and resistance to anti-programmed death receptor 1 therapy by regulating programmed cell death ligand 1 expression and interacting with other immune cells in the tumor microenvironment. However, when activated properly, macrophages can also play an anti-tumor role by enhancing the phagocytosis and cytotoxicity of tumor cells. TAM is associated with poor prognosis and drug resistance in patients treated with immunotherapy, indicating that macrophages are attractive targets for combined therapy in cancer treatment. Combination of targeting TAMs and immunotherapy overcomes the drug resistance and achieved excellent results in some cancers, which may be a promising strategy for cancer treatment in the future. Herein, we review the recent findings on the role of macrophages in tumor development, metastasis, and immunotherapy. We focus mainly on macrophage≥centered therapy, including strategies to deplete and reprogram TAMs, which represent the potential targets for improving tumor immunotherapy efficacy.
Collapse
Affiliation(s)
- Li Zhou
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, China
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, China
| | - Tiantian Zhao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ruzhe Zhang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chen Chen
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jiwei Li
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Sadeghi M, Dehnavi S, Sharifat M, Amiri AM, Khodadadi A. Innate immune cells: Key players of orchestra in modulating tumor microenvironment (TME). Heliyon 2024; 10:e27480. [PMID: 38463798 PMCID: PMC10923864 DOI: 10.1016/j.heliyon.2024.e27480] [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: 11/06/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/12/2024] Open
Abstract
The tumor microenvironment (TME) with vital role in cancer progression is composed of various cells such as endothelial cells, immune cells, and mesenchymal stem cells. In particular, innate immune cells such as macrophages, dendritic cells, myeloid-derived suppressor cells, neutrophils, innate lymphoid cells, γδT lymphocytes, and natural killer cells can either promote or suppress tumor progression when present in the TME. An increase in research on the cross-talk between the TME and innate immune cells will lead to new approaches for anti-tumoral therapeutic interventions. This review primarily focuses on the biology of innate immune cells and their main functions in the TME. In addition, it summarizes several innate immune-based immunotherapies that are currently tested in clinical trials.
Collapse
Affiliation(s)
- Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Dehnavi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moosa Sharifat
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir Mohammad Amiri
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Khodadadi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cancer, Petroleum and Environmental Pollutants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
40
|
Ouyang P, Wang L, Wu J, Tian Y, Chen C, Li D, Yao Z, Chen R, Xiang G, Gong J, Bao Z. Overcoming cold tumors: a combination strategy of immune checkpoint inhibitors. Front Immunol 2024; 15:1344272. [PMID: 38545114 PMCID: PMC10965539 DOI: 10.3389/fimmu.2024.1344272] [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: 11/25/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024] Open
Abstract
Immune Checkpoint Inhibitors (ICIs) therapy has advanced significantly in treating malignant tumors, though most 'cold' tumors show no response. This resistance mainly arises from the varied immune evasion mechanisms. Hence, understanding the transformation from 'cold' to 'hot' tumors is essential in developing effective cancer treatments. Furthermore, tumor immune profiling is critical, requiring a range of diagnostic techniques and biomarkers for evaluation. The success of immunotherapy relies on T cells' ability to recognize and eliminate tumor cells. In 'cold' tumors, the absence of T cell infiltration leads to the ineffectiveness of ICI therapy. Addressing these challenges, especially the impairment in T cell activation and homing, is crucial to enhance ICI therapy's efficacy. Concurrently, strategies to convert 'cold' tumors into 'hot' ones, including boosting T cell infiltration and adoptive therapies such as T cell-recruiting bispecific antibodies and Chimeric Antigen Receptor (CAR) T cells, are under extensive exploration. Thus, identifying key factors that impact tumor T cell infiltration is vital for creating effective treatments targeting 'cold' tumors.
Collapse
Affiliation(s)
- Peng Ouyang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Lijuan Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jianlong Wu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yao Tian
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Caiyun Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Dengsheng Li
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zengxi Yao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Ruichang Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Guoan Xiang
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Jin Gong
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhen Bao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
41
|
Denu RA, Dann AM, Keung EZ, Nakazawa MS, Nassif Haddad EF. The Future of Targeted Therapy for Leiomyosarcoma. Cancers (Basel) 2024; 16:938. [PMID: 38473300 PMCID: PMC10930698 DOI: 10.3390/cancers16050938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Leiomyosarcoma (LMS) is an aggressive subtype of soft tissue sarcoma that arises from smooth muscle cells, most commonly in the uterus and retroperitoneum. LMS is a heterogeneous disease with diverse clinical and molecular characteristics that have yet to be fully understood. Molecular profiling has uncovered possible targets amenable to treatment, though this has yet to translate into approved targeted therapies in LMS. This review will explore historic and recent findings from molecular profiling, highlight promising avenues of current investigation, and suggest possible future strategies to move toward the goal of molecularly matched treatment of LMS. We focus on targeting the DNA damage response, the macrophage-rich micro-environment, the PI3K/mTOR pathway, epigenetic regulators, and telomere biology.
Collapse
Affiliation(s)
- Ryan A. Denu
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Amanda M. Dann
- Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Michael S. Nakazawa
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elise F. Nassif Haddad
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
42
|
Cao L, Meng X, Zhang Z, Liu Z, He Y. Macrophage heterogeneity and its interactions with stromal cells in tumour microenvironment. Cell Biosci 2024; 14:16. [PMID: 38303024 PMCID: PMC10832170 DOI: 10.1186/s13578-024-01201-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/22/2024] [Indexed: 02/03/2024] Open
Abstract
Macrophages and tumour stroma cells account for the main cellular components in the tumour microenvironment (TME). Current advancements in single-cell analysis have revolutionized our understanding of macrophage diversity and macrophage-stroma interactions. Accordingly, this review describes new insight into tumour-associated macrophage (TAM) heterogeneity in terms of tumour type, phenotype, metabolism, and spatial distribution and presents the association between these factors and TAM functional states. Meanwhile, we focus on the immunomodulatory feature of TAMs and highlight the tumour-promoting effect of macrophage-tumour stroma interactions in the immunosuppressive TME. Finally, we summarize recent studies investigating macrophage-targeted therapy and discuss their therapeutic potential in improving immunotherapy by alleviating immunosuppression.
Collapse
Affiliation(s)
- Liren Cao
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiaoyan Meng
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhiyuan Zhang
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yue He
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
43
|
Lasser SA, Ozbay Kurt FG, Arkhypov I, Utikal J, Umansky V. Myeloid-derived suppressor cells in cancer and cancer therapy. Nat Rev Clin Oncol 2024; 21:147-164. [PMID: 38191922 DOI: 10.1038/s41571-023-00846-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2023] [Indexed: 01/10/2024]
Abstract
Anticancer agents continue to dominate the list of newly approved drugs, approximately half of which are immunotherapies. This trend illustrates the considerable promise of cancer treatments that modulate the immune system. However, the immune system is complex and dynamic, and can have both tumour-suppressive and tumour-promoting effects. Understanding the full range of immune modulation in cancer is crucial to identifying more effective treatment strategies. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of myeloid cells that develop in association with chronic inflammation, which is a hallmark of cancer. Indeed, MDSCs accumulate in the tumour microenvironment, where they strongly inhibit anticancer functions of T cells and natural killer cells and exert a variety of other tumour-promoting effects. Emerging evidence indicates that MDSCs also contribute to resistance to cancer treatments, particularly immunotherapies. Conversely, treatment approaches designed to eliminate cancer cells can have important additional effects on MDSC function, which can be either positive or negative. In this Review, we discuss the interplay between MDSCs and various other cell types found in tumours as well as the mechanisms by which MDSCs promote tumour progression. We also discuss the relevance and implications of MDSCs for cancer therapy.
Collapse
Affiliation(s)
- Samantha A Lasser
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Feyza G Ozbay Kurt
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Ihor Arkhypov
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Jochen Utikal
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Viktor Umansky
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany.
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany.
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| |
Collapse
|
44
|
Cornice J, Verzella D, Arboretto P, Vecchiotti D, Capece D, Zazzeroni F, Franzoso G. NF-κB: Governing Macrophages in Cancer. Genes (Basel) 2024; 15:197. [PMID: 38397187 PMCID: PMC10888451 DOI: 10.3390/genes15020197] [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: 01/12/2024] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are the major component of the tumor microenvironment (TME), where they sustain tumor progression and or-tumor immunity. Due to their plasticity, macrophages can exhibit anti- or pro-tumor functions through the expression of different gene sets leading to distinct macrophage phenotypes: M1-like or pro-inflammatory and M2-like or anti-inflammatory. NF-κB transcription factors are central regulators of TAMs in cancers, where they often drive macrophage polarization toward an M2-like phenotype. Therefore, the NF-κB pathway is an attractive therapeutic target for cancer immunotherapy in a wide range of human tumors. Hence, targeting NF-κB pathway in the myeloid compartment is a potential clinical strategy to overcome microenvironment-induced immunosuppression and increase anti-tumor immunity. In this review, we discuss the role of NF-κB as a key driver of macrophage functions in tumors as well as the principal strategies to overcome tumor immunosuppression by targeting the NF-κB pathway.
Collapse
Affiliation(s)
- Jessica Cornice
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Paola Arboretto
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Guido Franzoso
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| |
Collapse
|
45
|
Voissière A, Gomez-Roca C, Chabaud S, Rodriguez C, Nkodia A, Berthet J, Montane L, Bidaux AS, Treilleux I, Eberst L, Terret C, Korakis I, Garin G, Pérol D, Delord JP, Caux C, Dubois B, Ménétrier-Caux C, Bendriss-Vermare N, Cassier PA. The CSF-1R inhibitor pexidartinib affects FLT3-dependent DC differentiation and may antagonize durvalumab effect in patients with advanced cancers. Sci Transl Med 2024; 16:eadd1834. [PMID: 38266104 DOI: 10.1126/scitranslmed.add1834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/21/2023] [Indexed: 01/26/2024]
Abstract
Tumor-associated macrophages (TAMs) are a critical determinant of resistance to PD-1/PD-L1 blockade. This phase 1 study (MEDIPLEX, NCT02777710) investigated the safety and efficacy of pexidartinib, a CSF-1R-directed tyrosine kinase inhibitor (TKI), and durvalumab (anti-PD-L1) in patients with advanced colorectal and pancreatic carcinoma with the aim to enhance responses to PD-L1 blockade by eliminating CSF-1-dependent suppressive TAM. Forty-seven patients were enrolled. No unexpected toxicities were observed, one (2%) high microsatellite instability CRC patient had a partial response, and seven (15%) patients experienced stable disease as their best response. Increase of CSF-1 concentrations and decrease of CD14lowCD16high monocytes in peripheral blood mononuclear cells (PBMCs) confirmed CSF-1R engagement. Treatment decreased blood dendritic cell (DC) subsets and impaired IFN-λ/IL-29 production by type 1 conventional DCs in ex vivo TLR3-stimulated PBMCs. Pexidartinib also targets c-KIT and FLT3, both key growth factor receptors of DC development and maturation. In patients, FLT3-L concentrations increased with pexidartinib treatment, and AKT phosphorylation induced by FLT3-L ex vivo stimulation was abrogated by pexidartinib in human blood DC subsets. In addition, pexidartinib impaired the FLT3-L- but not GM-CSF-dependent generation of DC subsets from murine bone marrow (BM) progenitors in vitro and decreased DC frequency in BM and tumor-draining lymph node in vivo. Our results demonstrate that pexidartinib, through the inhibition of FLT3 signaling, has a deleterious effect on DC differentiation, which may explain the limited antitumor clinical activity observed in this study. This work suggests that inhibition of FLT3 should be considered when combining TKIs with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Aurélien Voissière
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
| | - Carlos Gomez-Roca
- Department of Medical Oncology, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Sylvie Chabaud
- Clinical Research Platform (DRCI), Centre Léon Bérard, Lyon, France
| | - Céline Rodriguez
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Axelle Nkodia
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Justine Berthet
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Laure Montane
- Clinical Research Platform (DRCI), Centre Léon Bérard, Lyon, France
| | | | | | - Lauriane Eberst
- Department of Medical Oncology, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Catherine Terret
- Department of Medical Oncology, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Iphigénie Korakis
- Department of Medical Oncology, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Gwenaelle Garin
- Clinical Research Platform (DRCI), Centre Léon Bérard, Lyon, France
| | - David Pérol
- Clinical Research Platform (DRCI), Centre Léon Bérard, Lyon, France
| | - Jean-Pierre Delord
- Department of Medical Oncology, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Christophe Caux
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Bertrand Dubois
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Christine Ménétrier-Caux
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Nathalie Bendriss-Vermare
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Philippe A Cassier
- Department of Medical Oncology, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| |
Collapse
|
46
|
Kang X, Huang Y, Wang H, Jadhav S, Yue Z, Tiwari AK, Babu RJ. Tumor-Associated Macrophage Targeting of Nanomedicines in Cancer Therapy. Pharmaceutics 2023; 16:61. [PMID: 38258072 PMCID: PMC10819517 DOI: 10.3390/pharmaceutics16010061] [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: 11/22/2023] [Revised: 12/24/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
The tumor microenvironment (TME) is pivotal in tumor growth and metastasis, aligning with the "Seed and Soil" theory. Within the TME, tumor-associated macrophages (TAMs) play a central role, profoundly influencing tumor progression. Strategies targeting TAMs have surfaced as potential therapeutic avenues, encompassing interventions to block TAM recruitment, eliminate TAMs, reprogram M2 TAMs, or bolster their phagocytic capabilities via specific pathways. Nanomaterials including inorganic materials, organic materials for small molecules and large molecules stand at the forefront, presenting significant opportunities for precise targeting and modulation of TAMs to enhance therapeutic efficacy in cancer treatment. This review provides an overview of the progress in designing nanoparticles for interacting with and influencing the TAMs as a significant strategy in cancer therapy. This comprehensive review presents the role of TAMs in the TME and various targeting strategies as a promising frontier in the ever-evolving field of cancer therapy. The current trends and challenges associated with TAM-based therapy in cancer are presented.
Collapse
Affiliation(s)
- Xuejia Kang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA;
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Yongzhuo Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangzhou 528400, China;
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Huiyuan Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Sanika Jadhav
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA;
| | - Zongliang Yue
- Department of Health Outcome and Research Policy, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA;
| | - Amit K. Tiwari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas of Medical Sciences, Little Rock, AR 72205, USA;
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA;
| |
Collapse
|
47
|
Aizaz M, Khan A, Khan F, Khan M, Musad Saleh EA, Nisar M, Baran N. The cross-talk between macrophages and tumor cells as a target for cancer treatment. Front Oncol 2023; 13:1259034. [PMID: 38033495 PMCID: PMC10682792 DOI: 10.3389/fonc.2023.1259034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
Macrophages represent an important component of the innate immune system. Under physiological conditions, macrophages, which are essential phagocytes, maintain a proinflammatory response and repair damaged tissue. However, these processes are often impaired upon tumorigenesis, in which tumor-associated macrophages (TAMs) protect and support the growth, proliferation, and invasion of tumor cells and promote suppression of antitumor immunity. TAM abundance is closely associated with poor outcome of cancer, with impediment of chemotherapy effectiveness and ultimately a dismal therapy response and inferior overall survival. Thus, cross-talk between cancer cells and TAMs is an important target for immune checkpoint therapies and metabolic interventions, spurring interest in it as a therapeutic vulnerability for both hematological cancers and solid tumors. Furthermore, targeting of this cross-talk has emerged as a promising strategy for cancer treatment with the antibody against CD47 protein, a critical macrophage checkpoint recognized as the "don't eat me" signal, as well as other metabolism-focused strategies. Therapies targeting CD47 constitute an important milestone in the advancement of anticancer research and have had promising effects on not only phagocytosis activation but also innate and adaptive immune system activation, effectively counteracting tumor cells' evasion of therapy as shown in the context of myeloid cancers. Targeting of CD47 signaling is only one of several possibilities to reverse the immunosuppressive and tumor-protective tumor environment with the aim of enhancing the antitumor response. Several preclinical studies identified signaling pathways that regulate the recruitment, polarization, or metabolism of TAMs. In this review, we summarize the current understanding of the role of macrophages in cancer progression and the mechanisms by which they communicate with tumor cells. Additionally, we dissect various therapeutic strategies developed to target macrophage-tumor cell cross-talk, including modulation of macrophage polarization, blockade of signaling pathways, and disruption of physical interactions between leukemia cells and macrophages. Finally, we highlight the challenges associated with tumor hypoxia and acidosis as barriers to effective cancer therapy and discuss opportunities for future research in this field.
Collapse
Affiliation(s)
- Muhammad Aizaz
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Aakif Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Faisal Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Maria Khan
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar, Pakistan
| | - Ebraheem Abdu Musad Saleh
- Department of Chemistry, College of Arts & Science, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Maryum Nisar
- School of Interdisciplinary Engineering & Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
48
|
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: 11] [Impact Index Per Article: 5.5] [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
|
49
|
Sun Y, Cronin MF, Mendonça MCP, Guo J, O’Driscoll CM. M2pep-Modified Cyclodextrin-siRNA Nanoparticles Modulate the Immunosuppressive Tumor Microenvironment for Prostate Cancer Therapy. Mol Pharm 2023; 20:5921-5936. [PMID: 37874541 PMCID: PMC10630955 DOI: 10.1021/acs.molpharmaceut.3c00769] [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: 08/24/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023]
Abstract
Prostate cancer (PCa) is the most prevalent cause of cancer deaths in men. Conventional strategies, such as surgery, radiation, or chemotherapy, face challenges including poor prognosis and resistance. Therefore, the development of new improved strategies is vital to enhance patient outcomes. Recently, immunotherapy has shown potential in the treatment of a range of cancers, including PCa. Tumor-associated macrophages (TAMs) play an important role in the tumor microenvironment (TME) and reprogramming of TAMs is associated with remodeling the TME. The colony-stimulating factor-1/colony stimulating factor-1 receptor (CSF-1/CSF-1R) signaling pathway is closely related to the polarization of TAMs. The downregulation of CSF-1R, using small interfering RNA (siRNA), has been shown to achieve the reprogramming of TAMs, from the immunosuppressive M2 phenotype to the immunostimulatory M1 one. To maximize specific cellular delivery an M2 macrophage-targeting peptide, M2pep, was formulated with an amphiphilic cationic β-Cyclodextrin (CD) incorporating CSF-1R siRNA. The resulting nanoparticles (NPs) increased M2 macrophage targeting both in vitro and in vivo, promoting the release of M1 factors and simultaneously downregulating the levels of M2 factors through TAM reprogramming. The subsequent remodeling of the TME resulted in a reduction in tumor growth in a subcutaneous PCa mouse model mainly mediated through the recruitment of cytotoxic T cells. In summary, this M2pep-targeted CD-based delivery system demonstrated significant antitumor efficacy, thus presenting an alternative immunotherapeutic strategy for PCa treatment.
Collapse
Affiliation(s)
- Yao Sun
- School
of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | | | | | - Jianfeng Guo
- School
of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | | |
Collapse
|
50
|
Snyder CM, Gill SI. Good CARMA: Turning bad tumor-resident myeloid cells good with chimeric antigen receptor macrophages. Immunol Rev 2023; 320:236-249. [PMID: 37295964 DOI: 10.1111/imr.13231] [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: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023]
Abstract
In religious philosophy, the concept of karma represents the effect of one's past and present actions on one's future. Macrophages are highly plastic cells with myriad roles in health and disease. In the setting of cancer, macrophages are among the most plentiful members of the immune microenvironment where they generally support tumor growth and restrain antitumor immunity. However, macrophages are not necessarily born bad. Macrophages or their immediate progenitors, monocytes, are induced to traffic to the tumor microenvironment (TME) and during this process they are polarized toward a tumor-promoting phenotype. Efforts to deplete or repolarize tumor-associated macrophages (TAM) for therapeutic benefit in cancer have to date disappointed. By contrast, genetic engineering of macrophages followed by their transit into the TME may allow these impressionable cells to mend their ways. In this review, we summarize and discuss recent advances in the genetic engineering of macrophages for the treatment of cancer.
Collapse
Affiliation(s)
- Christopher M Snyder
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|