1
|
Xu T, Zhang H, Yang BB, Qadir J, Yuan H, Ye T. Tumor-infiltrating immune cells state-implications for various breast cancer subtypes. Front Immunol 2025; 16:1550003. [PMID: 40438111 PMCID: PMC12116345 DOI: 10.3389/fimmu.2025.1550003] [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: 12/22/2024] [Accepted: 04/24/2025] [Indexed: 06/01/2025] Open
Abstract
Breast cancer presents a variety of subtypes due to its cellular and molecular heterogeneity. The capacity of cancer cells to proliferate, invade, and metastasize depends not only on their intrinsic characters but also on their dynamic interaction with the host tumor microenvironment (TME), which includes immune cells. Meanwhile, the infiltration of immune cells in the TME severely affects the occurrence, development, treatment, and prognosis of breast cancer. Therefore, this review aims to explore the immune invasive tumor microenvironment in different intrinsic subtypes of breast cancer. Additionally, it highlights the mechanistic influence of the infiltrating immune cells on stage-wise dynamics of breast tumorigenesis. Moreover, the present review also attempts to discern the regulatory relationship between tumor infiltrating immune cells and immune microenvironment in different molecular subtypes of breast cancer, thus, spotlighting its clinical significance.
Collapse
Affiliation(s)
- Tianshuang Xu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Hongjun Zhang
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Burton B. Yang
- Sunnybrook Research Institute and the Department of Laboratory Medicine and Pathobiology at the University of Toronto, Toronto, ON, Canada
| | - Javeria Qadir
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Hui Yuan
- School of Stomatology and School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Ting Ye
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Zhao W, Zhang Z, Xie M, Ding F, Zheng X, Sun S, Du J. Exploring tumor-associated macrophages in glioblastoma: from diversity to therapy. NPJ Precis Oncol 2025; 9:126. [PMID: 40316746 PMCID: PMC12048723 DOI: 10.1038/s41698-025-00920-x] [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: 11/21/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Glioblastoma is the most aggressive and lethal cancer of the central nervous system, presenting substantial treatment challenges. The current standard treatment, which includes surgical resection followed by temozolomide and radiation, offers limited success. While immunotherapies, such as immune checkpoint inhibitors, have proven effective in other cancers, they have not demonstrated significant efficacy in GBM. Emerging research highlights the pivotal role of tumor-associated macrophages (TAMs) in supporting tumor growth, fostering treatment resistance, and shaping an immunosuppressive microenvironment. Preclinical studies show promising results for therapies targeting TAMs, suggesting potential in overcoming these barriers. TAMs consist of brain-resident microglia and bone marrow-derived macrophages, both exhibiting diverse phenotypes and functions within the tumor microenvironment. This review delves into the origin, heterogeneity, and functional roles of TAMs in GBM, underscoring their dual roles in tumor promotion and suppression. It also summarizes recent progress in TAM-targeted therapies, which may, in combination with other treatments like immunotherapy, pave the way for more effective and personalized strategies against this aggressive malignancy.
Collapse
Affiliation(s)
- Wenwen Zhao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhi Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mingyuan Xie
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Ding
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangrong Zheng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shicheng Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianyang Du
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
3
|
Chen J, Wang S, Ding Y, Xu D, Zheng S. Radiotherapy-induced alterations in tumor microenvironment: metabolism and immunity. Front Cell Dev Biol 2025; 13:1568634. [PMID: 40356601 PMCID: PMC12066526 DOI: 10.3389/fcell.2025.1568634] [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: 02/18/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Tumor metabolism plays a pivotal role in shaping immune responses within the tumor microenvironment influencing tumor progression, immune evasion, and the efficacy of cancer therapies. Radiotherapy has been shown to impact both tumor metabolism and immune modulation, often inducing immune activation through damage-associated molecular patterns and the STING pathway. In this study, we analyse the particular characteristics of the tumour metabolic microenvironment and its effect on the immune microenvironment. We also review the changes in the metabolic and immune microenvironment that are induced by radiotherapy, with a focus on metabolic sensitisation to the effects of radiotherapy. Our aim is to contribute to the development of research ideas in the field of radiotherapy metabolic-immunological studies.
Collapse
Affiliation(s)
- Jinpeng Chen
- Department of General Surgery, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
- Southeast University Medical School, Nanjing, Jiangsu, China
| | - Sheng Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, China
| | - Yue Ding
- Department of General Surgery, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
- Southeast University Medical School, Nanjing, Jiangsu, China
| | - Duo Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shiya Zheng
- Southeast University Medical School, Nanjing, Jiangsu, China
- Department of Oncology, Southeast University, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Arnhold J. Oxidant-Based Cytotoxic Agents During Aging: From Disturbed Energy Metabolism to Chronic Inflammation and Disease Progression. Biomolecules 2025; 15:547. [PMID: 40305309 PMCID: PMC12025200 DOI: 10.3390/biom15040547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 05/02/2025] Open
Abstract
In humans, aging is an inevitable consequence of diminished growth processes after reaching maturity. The high order of biomolecules in cells and tissues is continuously disturbed by numerous physical and chemical destructive impacts. Host-derived oxidant-based cytotoxic agents (reactive species, transition free metal ions, and free heme) contribute considerably to this damage. These agents are under the control of immediately acting antagonizing principles, which are important to ensure cell and tissue homeostasis. In this review, I apply the concept of host-derived cytotoxic agents and their interplay with antagonizing principles to the aging process. During aging, energy metabolism and the supply of tissues with dioxygen and nutrients are increasingly disturbed. In addition, a chronic inflammatory state develops, a condition known as inflammaging. The balance between oxidant-based cytotoxic agents and protective mechanisms is analyzed depending on age-based physiological alterations in ATP production. Disturbances in this balance are associated with the development of age-related diseases and comorbidities. An enhanced production of reactive species from dysfunctional mitochondria, alterations in cellular redox homeostasis, and adaptations to hypoxia are highlighted. Examples of how disturbances between oxidant-based cytotoxic agents and antagonizing principles contribute to the pathogenesis of diseases in persons of advanced age are given.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
5
|
Li X, Guan R, Zhang S. Factors Contributing to the High Malignancy Level of Cholangiocarcinoma and Its Epidemiology: Literature Review and Data. BIOLOGY 2025; 14:351. [PMID: 40282217 PMCID: PMC12025278 DOI: 10.3390/biology14040351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
CCA is a highly desmoplastic malignant cancer and is the second most common primary liver malignancy after hepatocellular carcinoma (HCC), accounting for approximately 15% of all primary liver tumors. CCA has a poor prognosis, with an average five-year survival rate of 9%, which is lower than that of pancreatic cancer. Although considerable efforts have been invested into the genomics, epigenetics, and risk factors, very little is known about what might have been the key causes for the high malignancy level of CCA. In this review, we analyze the incidence and mortality of CCA in different regions based on data from 1994 to 2022 obtained from the International Agency for Research on Cancer (IARC), discuss the current status of treatment of the disease, and focus on what might be the main factors contributing to the high malignancy level of CCA: alkalosis caused by the Fenton reaction, hypoxia, and the TIME. The review includes studies published from 1979 to 2024, aiming to provide an updated synthesis of basic early classical theoretical knowledge and current knowledge about CCA. By revealing the epidemiological characteristics of CCA, the potential mechanisms of high malignancy, and the current challenges of treatment, this review aims to provide new directions for future cancer research, promote the development of personalized treatment strategies, and facilitate a deeper understanding and the more effective management of CCA worldwide.
Collapse
Affiliation(s)
- Xuan Li
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China;
| | - Renchu Guan
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China;
| | - Shuangquan Zhang
- School of Cyber Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| |
Collapse
|
6
|
Bayat M, Nahand JS. Battlegrounds of treatment resistance: decoding the tumor microenvironment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04055-5. [PMID: 40131387 DOI: 10.1007/s00210-025-04055-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
The tumor microenvironment (TME) emerges as a formidable actor in the cancer treatment landscape, wielding the power to thwart therapeutic efficacy across various modalities, including chemotherapy, radiotherapy, immunotherapy, targeted therapy, and hormonal therapy. This intricate ecosystem comprising diverse cellular constituents, signaling molecules, and the extracellular matrix fosters a dynamic interplay that profoundly influences tumor behavior and treatment outcomes. This review explores the mechanisms through which the TME drives resistance to standard therapies, emphasizing key factors such as hypoxia, immune evasion, and metabolic reprogramming. Furthermore, we illuminate innovative strategies aimed at reprogramming this hostile environment, including the application of therapeutic vaccines, CAR T cell therapy, and combination immunotherapies designed to enhance anti-tumor responses. By advocating for multidimensional approaches that dismantle the TME's barriers to effective treatment, this review calls for a transformative shift in cancer treatment paradigms. By bridging the gap between the TME's complexities and targeted therapeutic strategies, we pave the way for targeted interventions that promise to enhance clinical outcomes and improve patient prognosis in the relentless battle against cancer.
Collapse
Affiliation(s)
- Mobina Bayat
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadri Nahand
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Das G, Ptacek J, Campbell J, Li X, Havlinova B, Noonepalle SK, Villagra A, Barinka C, Novakova Z. Targeting prostate cancer by new bispecific monocyte engager directed to prostate-specific membrane antigen. PLoS One 2025; 20:e0307353. [PMID: 40096254 PMCID: PMC11913275 DOI: 10.1371/journal.pone.0307353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/28/2025] [Indexed: 03/19/2025] Open
Abstract
Prostate cancer (PCa) ranks as the second leading cause of cancer-related deaths among men in the United States. Prostate-specific membrane antigen (PSMA) represents a well-established biomarker of PCa, and its levels correlate positively with the disease progression, culminating at the stage of metastatic castration-resistant prostate cancer. Due to its tissue-specific expression and cell surface localization, PSMA shows superior potential for precise imaging and therapy of PCa. Antibody-based immunotherapy targeting PSMA offers the promise of selectively engaging the host immune system with minimal off-target effects. Here we report on the design, expression, purification, and characterization of a bispecific engager, termed 5D3-CP33, that efficiently recruits macrophages to the vicinity of PSMA-positive cancer cells mediating PCa death. The engager was engineered by fusing the anti-PSMA 5D3 antibody fragment to a cyclic peptide 33 (CP33), selectively binding the Fc gamma receptor I (FcγRI/CD64) on the surface of phagocytes. Functional parts of the 5D3-CP33 engager revealed a nanomolar affinity for PSMA and FcγRI/CD64 with dissociation constants of KD = 3 nM and KD = 140 nM, respectively. At a concentration as low as 0.3 nM, the engager was found to trigger the production of reactive oxygen species by U937 monocytic cells in the presence of PSMA-positive cells. Moreover, flow cytometry analysis demonstrated antibody-dependent cell-mediated phagocytosis of PSMA-positive cancer cells by U937 monocytes when exposed to 0.15 nM 5D3-CP33. Our findings illustrate that 5D3-CP33 effectively and specifically activates monocytes upon PSMA-positive target engagement, resulting in the elimination of tumor cells. The 5D3-CP33 engager can thus serve as a promising lead for developing new immunotherapy tools for the efficient treatment of PCa.
Collapse
Affiliation(s)
- Gargi Das
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jakub Ptacek
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Jana Campbell
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Xintang Li
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C., United States of America
| | - Barbora Havlinova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Satish kumar Noonepalle
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C., United States of America
| | - Alejandro Villagra
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C., United States of America
| | - Cyril Barinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Zora Novakova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| |
Collapse
|
8
|
He Y, Hong Q, Chen S, Zhou J, Qiu S. Reprogramming tumor-associated macrophages in gastric cancer: a pathway to enhanced immunotherapy. Front Immunol 2025; 16:1558091. [PMID: 40098971 PMCID: PMC11911521 DOI: 10.3389/fimmu.2025.1558091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Gastric cancer (GC) remains a significant global health concern due to its poor prognosis and limited therapeutic options, particularly in advanced stages. Tumor microenvironment (TME), particularly tumor-associated macrophages (TAMs), plays a key role in tumor progression, immune evasion, and therapy resistance. TAMs exhibit plasticity, shifting between pro-inflammatory M1 and immunosuppressive M2 phenotypes, with the latter predominating in GC and contributing to poor outcomes. Recent therapeutic advancements focus on targeting TAMs, including inhibiting M2 polarization, reprogramming TAMs to M1 phenotypes, and combining TAM-targeted approaches with immune checkpoint inhibitors. Innovations in nanotechnology, metabolic reprogramming, and targeting key pathways such as interleukin-6 and C-C motif ligand 2/C-C motif chemokine receptor 2 further enhance these strategies. However, challenges remain, including the spatial and functional heterogeneity of TAMs within the TME and the need for selective targeting to avoid disrupting immune homeostasis. Ongoing research on TAM origins, functions, and interactions within the TME is crucial for developing precise and effective therapies. These advances hold promise not only for improving outcomes in GC but also for addressing other cancers with similarly complex microenvironments.
Collapse
Affiliation(s)
| | | | | | | | - Shengliang Qiu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang
Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Xu N, Zhang T, Sun W, Ye C, Zhou H. Identification of an extracellular matrix signature for predicting prognosis and sensitivity to therapy of patients with gastric cancer. Sci Rep 2025; 15:7464. [PMID: 40032943 PMCID: PMC11876314 DOI: 10.1038/s41598-025-88376-8] [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: 09/01/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Extracellular matrix (ECM) is a vital component of the tumor microenvironment and plays a crucial role in the development and progression of gastric cancer (GC). Co-expression networks were established by means of the "WGCNA" package, the optimal model for extracellular matrix scores (ECMs) was developed and validated, with its accuracy in predicting the prognosis and treatment sensitivity of GC patients assessed. We performed univariate cox regression analysis [HR = 6.8 ( 3.3-14 ), p < 0.001] which demonstrated that ECMs was an independent risk character and perceptibly superior to other factors with further analysis of multivariate Cox regression [HR = 8.68 ( 4.16-18.08 ), p < 0.001]. The nomogram, presenting the clinical prognosis model for GC patients, demonstrated accuracy through KM analysis [HR = 3.97 (2.56-6.16), p < 0.001] and ROC curves with AUC values of 0.70, 0.72, and 0.72 at 1, 3, and 5 years, respectively. Using the ECMs model, we stratified GC patients into high- and low-risk groups, enabling precise predictions of prognosis and drug sensitivity. This stratification provides a new strategic direction for the personalized treatment of GC.
Collapse
Affiliation(s)
- Nan Xu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Taojing Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Weiwei Sun
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chenxiao Ye
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Huamiao Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China.
| |
Collapse
|
10
|
Zhang J, Wang F, Sun Z, Ye J, Chu H. Multidimensional applications of prussian blue-based nanoparticles in cancer immunotherapy. J Nanobiotechnology 2025; 23:161. [PMID: 40033359 PMCID: PMC11874808 DOI: 10.1186/s12951-025-03236-x] [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: 10/25/2024] [Accepted: 02/16/2025] [Indexed: 03/05/2025] Open
Abstract
Immunotherapy holds notable progress in the treatment of cancer. However, the clinical therapeutic effect remains a significant challenge due to immune-related side effects, poor immunogenicity, and immunosuppressive microenvironment. Nanoparticles have emerged as a revolutionary tool to surmount these obstacles and amplify the potency of immunotherapeutic agents. Prussian blue nanoparticles (PBNPs) exhibit multi-dimensional immune function in cancer immunotherapy, including acting as a nanocarrier to deliver immunotherapeutic agents, as a photothermal agent to improve the efficacy of immunotherapy through photothermal therapy, as a nanozyme to regulate tumor microenvironment, and as an iron donor to induce immune events related to ferroptosis and tumor-associated macrophages polarization. This review focuses on the advances and applications of PBNPs in cancer immunotherapy. First, the biomedical functions of PBNPs are introduced. Then, based on the immune function of PBNPs, we systematically reviewed the multidimensional application of PBNPs in cancer immunotherapy. Finally, the challenges and future developments of PBNPs-based cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
11
|
Dai Q, Peng Y, He P, Wu X. Interactions and communications in the prostate tumour microenvironment: evolving towards effective cancer therapy. J Drug Target 2025; 33:295-315. [PMID: 39445641 DOI: 10.1080/1061186x.2024.2418344] [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: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Prostate cancer is one of the most common malignancies in men. The tumour microenvironment (TME) has a critical role in the initiation, progression, and metastasis of prostate cancer. TME contains various cell types, including cancer-associated fibroblasts (CAFs), endothelial cells, immune cells such as macrophages, lymphocytes B and T, natural killer (NK) cells, and other proteins such as extracellular matrix (ECM) components. The interactions and communications between these cells within the TME are crucial for the growth and response of various solid tumours, such as prostate cancer to different anticancer modalities. In this review article, we exemplify the various mechanisms by which the TME influences prostate cancer progression. The roles of different cells, cytokines, chemokines, and growth factors in modulating the immune response and prostate tumour growth will be discussed. The impact of these cells and factors and other ECM components on tumour cell invasion and metastasis will also be discussed. We explain how these interactions in TME can affect the response of prostate cancer to therapy. We also highlight the importance of understanding these interactions to develop novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Peng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
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
|
Branco H, Xavier CPR, Riganti C, Vasconcelos MH. Hypoxia as a critical player in extracellular vesicles-mediated intercellular communication between tumor cells and their surrounding microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189244. [PMID: 39672279 DOI: 10.1016/j.bbcan.2024.189244] [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: 06/18/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
In the past years, increasing attention has been paid to the role of extracellular vesicles (EVs) as mediators of intercellular communication in cancer. These small size particles mediate the intercellular transfer of important bioactive molecules involved in malignant initiation and progression. Hypoxia, or low partial pressure of oxygen, is recognized as a remarkable feature of solid tumors and has been demonstrated to exert a profound impact on tumor prognosis and therapeutic efficacy. Indeed, the high-pitched growth rate and chaotic neovascular architecture that embodies solid tumors results in a profound reduction in oxygen pressure within the tumor microenvironment (TME). In response to oxygen-deprived conditions, tumor cells and their surrounding milieu develop homeostatic adaptation mechanisms that contribute to the establishment of a pro-tumoral phenotype. Latest evidence suggests that the hypoxic microenvironment that surrounds the tumor bulk may be a clincher for the observed elevated levels of circulating EVs in cancer patients. Thus, it is proposed that EVs may play a role in mediating intercellular communication in response to hypoxic conditions. This review focuses on the EVs-mediated crosstalk that is established between tumor cells and their surrounding immune, endothelial, and stromal cell populations, within the hypoxic TME.
Collapse
Affiliation(s)
- Helena Branco
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, 4585-116 Gandra, Portugal.
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126 Torino, Italy; Interdepartmental Research Center for Molecular Biotechnology "G. Tarone", University of Torino, 10126 Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
14
|
Mahmoudian M, Alizadeh S, Lotfi D, Khazaei Monfared Y, Mahdipour M, Trotta F, Zakeri-Milani P, Islambulchilar Z. Modulating exosomal communication between macrophages and melanoma cancer cells via cyclodextrin-based nanosponges loaded with doxorubicin. Nanotoxicology 2025; 19:17-27. [PMID: 39727335 DOI: 10.1080/17435390.2024.2446553] [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: 09/16/2024] [Revised: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
The cellular components of the tumor microenvironment (TME) comprise cancer cells and nonmalignant cells including stromal and immune cells. Exosomes are extracellular vesicles secreted by various types of cells that play a crucial role in intercellular communications within TME. The main goal of this study was to elucidate how exosomes of macrophage cells treated with doxorubicin (DOX) and DOX-loaded cyclodextrin-based nanosponges (DOX-CDNSs), affect melanoma cancer cell proliferation. For this aim, the exosomes of the murine macrophage cell line (RAW 264.7) were isolated and characterized after treating the cells with DOX and DOX-CDNSs. The results demonstrated that DOX-CDNSs at a treatment concentration of 1 µg/mL, were nontoxic for macrophages and remarkably toxic against cancer cells. However, DOX was nontoxic for both cell types at the same treatment concentration. DOX and DOX-CDNSs remarkably declined the viability of both cell types at higher concentrations (25 and 50 µg/mL). Intriguingly, the exosomes of DOX-CD-NSs treated macrophages promoted the viability of cancer cells at the treatment concentrations of 1, 20, and 40 µg/mL. While the exosomes of DOX-treated macrophages increased cell viability of cancer cells only at the lowest concentration. In conclusion, this study suggests that utilization of CD-NSs may augment the toxicity of DOX against cancer cells, while it could direct macrophages toward secreting exosomes that favor the growth of cancer cells.
Collapse
Affiliation(s)
- Mohammad Mahmoudian
- Candiolo Cancer Institute, FPO-IRCCS, University of Turin, Candiolo, Italy
- Deartment of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shokoufeh Alizadeh
- Deartment of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Darya Lotfi
- Deartment of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Khazaei Monfared
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, University of Turin, Torino, Italy
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Centre, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ziba Islambulchilar
- Deartment of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Freeman MT, Parvaresh-Rizi A, Meenach SA. Enhanced Macrophage Uptake of Spray-Dried Phosphatidylserine-Loaded Microparticles for Pulmonary Drug Delivery Applications. J Drug Deliv Sci Technol 2025; 104:106535. [PMID: 39830679 PMCID: PMC11737435 DOI: 10.1016/j.jddst.2024.106535] [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] [Indexed: 01/22/2025]
Abstract
Macrophages are an integral part of the innate immune system and act as a first line of defense to pathogens; however, macrophages can be reservoirs for pathogens to hide and replicate. Tuberculosis, influenza virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are common diseases whose pathogens are uptaken into macrophages. Current treatments for diseases such as these are limited by the therapeutic delivery method, which typically involves systemic delivery in large, frequent doses. This study aims to overcome this limitation via the development of an inhalable dry powder microparticle (MP) formulation capable of targeted drug delivery to alveolar macrophages in addition to controlled release of a therapeutic. A simple one-step spray drying method was used to synthesize acetalated dextran (Ac-Dex) MP loaded with the model therapeutic, curcumin, and 1,2-dipalmitoyl-sn-glycero-3-phospho-L-serine (DPPS), which is a phospholipid that induces ligand-receptor mediated macrophage phagocytosis. The resulting MP exhibited significantly more uptake by RAW 264.7 macrophages in comparison to MP without DPPS, and it was shown that DPPS-mediated uptake was macrophage specific. The particles exhibited pH-responsive release and in vitro aerosol dispersion analysis confirmed the MP can be effectively aerosolized for pulmonary delivery. Overall, the described MP has the potential to improve treatment efficacy for macrophage-associated diseases.
Collapse
Affiliation(s)
- Matthew T. Freeman
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881 USA
| | - Arianne Parvaresh-Rizi
- Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, RI 02881 USA
| | - Samantha A. Meenach
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881 USA
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881 USA
| |
Collapse
|
16
|
Pierini S, Gabbasov R, Oliveira-Nunes MC, Qureshi R, Worth A, Huang S, Nagar K, Griffin C, Lian L, Yashiro-Ohtani Y, Ross K, Sloas C, Ball M, Schott B, Sonawane P, Cornell L, Blumenthal D, Chhum S, Minutolo N, Ciccaglione K, Shaw L, Zentner I, Levitsky H, Shestova O, Gill S, Varghese B, Cushing D, Ceeraz DeLong S, Abramson S, Condamine T, Klichinsky M. Chimeric antigen receptor macrophages (CAR-M) sensitize HER2+ solid tumors to PD1 blockade in pre-clinical models. Nat Commun 2025; 16:706. [PMID: 39814734 PMCID: PMC11735936 DOI: 10.1038/s41467-024-55770-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
We previously developed human CAR macrophages (CAR-M) and demonstrated redirection of macrophage anti-tumor function leading to tumor control in immunodeficient xenograft models. Here, we develop clinically relevant fully immunocompetent syngeneic models to evaluate the potential for CAR-M to remodel the tumor microenvironment (TME), induce T cell anti-tumor immunity, and sensitize solid tumors to PD1/PDL1 checkpoint inhibition. In vivo, anti-HER2 CAR-M significantly reduce tumor burden, prolong survival, remodel the TME, increase intratumoral T cell and natural killer (NK) cell infiltration, and induce antigen spreading. CAR-M therapy protects against antigen-negative relapses in a T cell dependent fashion, confirming long-term anti-tumor immunity. In HER2+ solid tumors with limited sensitivity to anti-PD1 (aPD1) monotherapy, the combination of CAR-M and aPD1 significantly improves tumor growth control, survival, and remodeling of the TME in pre-clinical models. These results demonstrate synergy between CAR-M and T cell checkpoint blockade and provide a strategy to potentially enhance response to aPD1 therapy for patients with non-responsive tumors.
Collapse
Affiliation(s)
| | | | | | | | | | - Shuo Huang
- Carisma Therapeutics Inc, Philadelphia, PA, USA
| | - Karan Nagar
- Carisma Therapeutics Inc, Philadelphia, PA, USA
| | | | - Lurong Lian
- Carisma Therapeutics Inc, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Lauren Shaw
- Carisma Therapeutics Inc, Philadelphia, PA, USA
| | | | | | - Olga Shestova
- Center for Cellular Immunotherapies, Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Saar Gill
- Center for Cellular Immunotherapies, Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Ward MB, Jones AB, Krenciute G. Therapeutic advantage of combinatorial chimeric antigen receptor T cell and chemotherapies. Pharmacol Rev 2025; 77:100011. [PMID: 39952691 DOI: 10.1124/pharmrev.124.001070] [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: 04/23/2024] [Revised: 08/28/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapies have transformed outcomes for many patients with hematological malignancies. However, some patients do not respond to CAR T cell treatment, and adapting CAR T cells for treatment of solid and brain tumors has been met with many challenges, including a hostile tumor microenvironment and poor CAR T cell persistence. Thus, it is unlikely that CAR T cell therapy alone will be sufficient for consistent, complete tumor clearance across patients with cancer. Combinatorial therapies of CAR T cells and chemotherapeutics are a promising approach for overcoming this because chemotherapeutics could augment CAR T cells for improved antitumor activity or work in tandem with CAR T cells to clear tumors. Herein, we review efforts toward achieving successful CAR T cell and chemical drug combination therapies. We focus on combination therapies with approved chemotherapeutics because these will be more easily translated to the clinic but also review nonapproved chemotherapeutics and drug screens designed to reveal promising new CAR T cell and chemical drug combinations. Overall, this review highlights the promise of CAR T cell and chemotherapy combinations with a specific focus on how combinatorial therapy overcomes challenges faced by either monotherapy and supports the potential of this therapeutic strategy to improve outcomes for patients with cancer. SIGNIFICANCE STATEMENT: Improving currently available CAR T cell products via combinatorial therapy with chemotherapeutics has the potential to drastically expand the types of cancers and number of patients that could benefit from these therapies when neither alone has been sufficient to achieve tumor clearance. Herein, we provide a thorough review of the current efforts toward studying CAR T and chemotherapy combinatorial therapies and offer perspectives on optimal ways to identify new and effective combinations moving forward.
Collapse
Affiliation(s)
- Meghan B Ward
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Amber B Jones
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
18
|
Rahimi S, Shirin F, Moassesfar M, Zafari H, Bahmaie N, Baghebani K, Bidmeshki Y, Sajjadi Manesh SM, Rasoulzadeh Darabad K, Bahmaie M, Nouri E, Kilic A, Ansarin M, Özışık P, Simsek E, Ozensoy Guler O. Role of Hypoxia Induced by Medicinal Plants; A Revolutionary Era of Cellular and Molecular Herbal Medicine in Neuroblastoma Treatment. FRONT BIOSCI-LANDMRK 2024; 29:422. [PMID: 39735975 DOI: 10.31083/j.fbl2912422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/13/2024] [Accepted: 07/19/2024] [Indexed: 12/31/2024]
Abstract
As one of the most common solid pediatric cancers, Neuroblastoma (NBL) accounts for 15% of all of the cancer-related mortalities in infants with increasing incidence all around the world. Despite current therapeutic approaches for NBL (radiotherapies, surgeries, and chemotherapies), these approaches could not be beneficial for all of patients with NBL due to their low effectiveness, and some severe side effects. These challenges lead basic medical scientists and clinical specialists toward an optimal medical interventions for clinical management of NBL. Regardingly, taking molecular and cellular immunopathophysiology involved in the hypoxic microenvironment of NBL into account, it can practically be a contributing approach in the development of "molecular medicine" for treatment of NBL. Interestingly, pivotal roles of "herbal medicine" in the hypoxic microenvironment of NBL have been extensively interrogated for treating a NBL, functionally being served as an anti-cancer agent via inducing a wide range of molecular and cellular signaling, like apoptosis, cell cycle arrest, and inhibiting angiogenesis. Hence, in this review study, the authors aim to summarize the anti-tumor effects of some medicinal plants and their phytoconstituents through molecular immunopathophysiological mechanisms involved in the hypoxic microenvironment of NBL. In addition, they try to open promising windows to immune gene-based therapies for NBL "precision medicine" through clinical advantages of herbal and molecular medicine. An interdisciplinary collaboration among translation and molecular medicine specialists, immunobiologists, herbal medicine specialists, and pediatric neuro-oncologists is highly recommended.
Collapse
Affiliation(s)
- Samin Rahimi
- Department of Genetics, Faculty of Natural Sciences, Tabriz University, 5166616471 Tabriz, Iran
| | - Fatemeh Shirin
- Department of Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, 1651153311 Tehran, Iran
| | - Mahdi Moassesfar
- Department of Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, 1651153311 Tehran, Iran
| | - Hossein Zafari
- Department of Chemical Engineering, Faculty of Chemical Engineering, Shahreza Branch, Islamic Azad University, 8648146411 Shahreza, Iran
| | - Nazila Bahmaie
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University (AYBU), 06800 Ankara, Turkey
| | - Kimia Baghebani
- Department of Biology, College of Basic Sciences, Kermanshah Branch, Islamic Azad University, 6718997551 Kermanshah, Iran
- Now with Department of Biotechnology, College of Environmental and Bioresource Sciences, Jeonbuk National University, 54896 Jeonbuk, Republic of Korea
| | - Yasna Bidmeshki
- Department of Biology, College of Basic Sciences, Kermanshah Branch, Islamic Azad University, 6718997551 Kermanshah, Iran
| | - Seyede Masoumeh Sajjadi Manesh
- Department of Biomedical Engineering, College of Basic Sciences, Qom Branch, Islamic Azad University, 3716146611 Qom, Iran
| | | | - Massoud Bahmaie
- Department of Herbal Medicine, University of Poona, 411007 Poona, India
| | - Elham Nouri
- Clinical Diagnosis Laboratory, Shahid Beheshti University-affiliated Hospital, Zanjan University of Medical Sciences (ZUMS), 4513956111 Zanjan, Iran
- Department of Medical Laboratory Science, Faculty of Paramedicine, Zanjan University of Medical Sciences (ZUMS), 4513956111 Zanjan, Iran
| | - Ahmet Kilic
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University (AYBU), 06800 Ankara, Turkey
| | - Melika Ansarin
- Department of Obstetrics and Gynecology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), 1449614535 Tehran, Iran
| | - Pınar Özışık
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Ankara Bilkent City Hospital, 06800 Ankara, Turkey
- Department of Brain and Nerve Surgery, Faculty of Medicine, Ankara Yildirim Beyazit University (AYBU), 06800 Ankara, Turkey
| | - Ender Simsek
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University (AYBU), 06800 Ankara, Turkey
| | - Ozen Ozensoy Guler
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University (AYBU), 06800 Ankara, Turkey
| |
Collapse
|
19
|
Yang L, Zhang Y, Yang L. Adenosine signaling in tumor-associated macrophages and targeting adenosine signaling for cancer therapy. Cancer Biol Med 2024; 21:995-1011. [PMID: 39629670 PMCID: PMC11667779 DOI: 10.20892/j.issn.2095-3941.2024.0228] [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: 06/24/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024] Open
Abstract
This review examined the critical role of adenosine signaling in modulating the behavior of tumor-associated macrophages (TAMs), a key determinant of the tumor microenvironment (TME). Adenosine is an immunosuppressive metabolite that is highly enriched in the TME due to elevated expression of adenosine triphosphatase (ATPase). Adenosine influences polarization of TAMs through A2A and A2B receptors, which drives a phenotype that supports tumor progression and immune evasion. The adenosine-mediated regulation of TAMs significantly suppresses the TME, dampening the efficacy of current immunotherapies. Targeting the adenosine pathway has shown potential in preclinical studies through reversal of the immunosuppressive microenvironment and antitumor immune response enhancement. Clinical trials are currently underway to determine the impact of A2A receptor antagonists, and CD39 and CD73 inhibition, enzymes that are pivotal in adenosine production, in various cancers. The current understanding of the CD39-CD73-adenosine axis in TAM regulation and the emerging strategies targeting adenosine signaling pathway for therapeutic intervention are the subjects of this review. The current clinical trials focusing on adenosine pathway inhibitors in combination with existing therapies to improve clinical outcomes are summarized and the need for continued research to refine these approaches for cancer treatment is emphasized.
Collapse
Affiliation(s)
- Lei Yang
- Biotherapy Center and Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou 450052, China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou 450052, China
| | - Li Yang
- Biotherapy Center and Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou 450052, China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou 450052, China
| |
Collapse
|
20
|
Affonso de Oliveira JF, Moreno-Gonzalez MA, Ma Y, Deng X, Schuphan J, Steinmetz NF. Plant Virus Intratumoral Immunotherapy with CPMV and PVX Elicits Durable Antitumor Immunity in a Mouse Model of Diffuse Large B-Cell Lymphoma. Mol Pharm 2024; 21:6206-6219. [PMID: 39526560 DOI: 10.1021/acs.molpharmaceut.4c00507] [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: 11/16/2024]
Abstract
Plant viruses are naturally occurring nanoparticles and adjuvants that interact with the mammalian immune system. This property can be harnessed in vaccines and immunotherapy. We have previously demonstrated that intratumoral immunotherapy with cowpea mosaic virus (CPMV) stimulates systemic and durable antitumor immunity in mouse tumor models and canine cancer patients. Here we compared the antitumor efficacy of CPMV with potato virus X (PVX) using a mouse model B-cell lymphoma (A20 and BALB/c mice). Despite their diverse morphologies and physiochemical properties, both plant viruses elicited systemic and long-lasting antitumor immune memory, preventing the recurrence of A20 lymphoma in rechallenge experiments. Data indicate differences in the underlying mechanism: CPMV persists longer in the tumor microenvironment (TME) compared to PVX; CPMV is a potent and multivalent toll-like receptor (TLR) agonist (activating TLRs 2, 4 and 7) while PVX may only weakly engage with TLR7. While CPMV and PVX recruit myeloid cells (neutrophils)─CPMV also recruits macrophages. Data further indicate that antiviral T cells may play a role in antitumor efficacy in the case of CPMV immunotherapy, however this may not be the case for PVX. Regardless of the mechanism of action, both CPMV and PVX elicited a durable antitumor response against a B-cell lymphoma tumor model and thus are intratumoral immunotherapy candidates for clinical development.
Collapse
Affiliation(s)
- Jessica Fernanda Affonso de Oliveira
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, California 92093, United States
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, San Diego, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Miguel A Moreno-Gonzalez
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, California 92093, United States
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, San Diego, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Yifeng Ma
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, California 92093, United States
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, San Diego, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Xinyi Deng
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, California 92093, United States
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, San Diego, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Juliane Schuphan
- Institut für Molekulare Biotechnologie, RWTH Aachen University, Worringer Weg 1, 52074 Aachen, Germany
| | - Nicole F Steinmetz
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, California 92093, United States
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, San Diego, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Radiology, University of California San Diego, La Jolla, California 92093, United States
- Institute for Materials Discovery and Design, University of California San Diego, La Jolla, California 92093, United States
- Center for Engineering in Cancer, Institute of Engineering Medicine, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
21
|
Jabbarzadeh Kaboli P, Roozitalab G, Farghadani R, Eskandarian Z, Zerrouqi A. c-MET and the immunological landscape of cancer: novel therapeutic strategies for enhanced anti-tumor immunity. Front Immunol 2024; 15:1498391. [PMID: 39664377 PMCID: PMC11632105 DOI: 10.3389/fimmu.2024.1498391] [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: 10/06/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
Cellular mesenchymal-epithelial transition factor (c-MET), also known as hepatocyte growth factor receptor (HGFR), is a crucial receptor tyrosine kinase implicated in various solid tumors, including lung, breast, and liver cancers. The concomitant expression of c-MET and PD-L1 in tumors, such as hepatocellular carcinoma, highlights their prognostic significance and connection to therapeutic resistance. Cancer-associated fibroblasts and mesenchymal stromal cells produce hepatocyte growth factor (HGF), activating c-MET signaling in tumor cells and myeloid-derived suppressor cells (MDSC). This activation leads to metabolic reprogramming and increased activity of enzymes like glutaminase (GLS), indoleamine 2,3-dioxygenase (IDO), and arginase 1 (ARG1), depleting essential amino acids in the tumor microenvironment that are vital for effector immune cell function. This review highlights the interplay between tumor cells and myeloid-derived suppressor cells (MDSCs) that create an immunosuppressive environment while providing targets for c-MET-focused immunotherapy. It emphasizes the clinical implications of c-MET inhibition on the behavior of immune cells such as neutrophils, macrophages, T cells, and NK cells. It explores the potential of c-MET antagonism combined with immunotherapeutic strategies to enhance cancer treatment paradigms. This review also discusses the innovative cancer immunotherapies targeting c-MET, including chimeric antigen receptor (CAR) therapies, monoclonal antibodies, and antibody-drug conjugates, while encouraging the development of a comprehensive strategy that simultaneously tackles immune evasion and enhances anti-tumor efficacy further to improve the clinical prognoses for patients with c-MET-positive malignancies. Despite the challenges and variability in efficacy across different cancer subtypes, continued research into the molecular mechanisms and the development of innovative therapeutic strategies will be crucial.
Collapse
Affiliation(s)
| | - Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Zoya Eskandarian
- Research Institute Children’s Cancer Center, and Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Abdessamad Zerrouqi
- Department of Biochemistry, Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Kim YJ, Nanda SS, Jiang F, Pyo SY, Han JY, Koh SS, Kang TH. Pancreatic Adenocarcinoma Up-Regulated Factor (PAUF) Transforms Human Monocytes into Alternative M2 Macrophages with Immunosuppressive Action. Int J Mol Sci 2024; 25:11545. [PMID: 39519098 PMCID: PMC11547018 DOI: 10.3390/ijms252111545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) promote immune evasion, cancer cell proliferation, and metastasis. Ongoing research is focused on finding ways to prevent tumor growth by inhibiting TAM polarization, which has shown a correlation with unfavorable prognosis in clinical studies. Pancreatic adenocarcinoma up-regulated factor (PAUF) is a protein secreted from pancreatic cancer (PC) and acts as a TME modulator that affects the TME by acting on not only cancer cells but also stromal cells and immune cells. Tumor cells can evade the immune system by PAUF binding to Toll-like receptor (TLR) in monocytes, as this research shows. In this study, the examination centered around the recruitment of human monocytes by PAUF and the subsequent differentiation into macrophages. In an in vitro chemotaxis assay, PAUF induced chemotactic migration of TLR2-mediated monocytes. In addition, PAUF induced differentiation of monocytes into M2 macrophages, which was verified based on expressing surface markers and cytokines and morphological analysis. The inhibition of T cell proliferation and function was observed in differentiated M2 macrophages. To conclude, these findings indicate that PAUF functions as a promoter of cancer progression by regulating the recruitment and differentiation of macrophages within TMEs, ultimately causing immunosuppression.
Collapse
Affiliation(s)
- Yeon Jeong Kim
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea
| | - Sitansu Sekhar Nanda
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Fen Jiang
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Seung Yeon Pyo
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Jin-Yeong Han
- Department of Laboratory Medicine, College of Medicine, Dong-A University, Busan 49201, Republic of Korea;
| | - Sang Seok Koh
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Tae Heung Kang
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| |
Collapse
|
23
|
Mesaros O, Onciul M, Matei E, Joldes C, Jimbu L, Neaga A, Serban O, Zdrenghea M, Nanut AM. Macrophages as Potential Therapeutic Targets in Acute Myeloid Leukemia. Biomedicines 2024; 12:2306. [PMID: 39457618 PMCID: PMC11505058 DOI: 10.3390/biomedicines12102306] [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: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous malignant hemopathy, and although new drugs have emerged recently, current treatment options still show limited efficacy. Therapy resistance remains a major concern due to its contribution to treatment failure, disease relapse, and increased mortality among patients. The underlying mechanisms of resistance to therapy are not fully understood, and it is crucial to address this challenge to improve therapy. Macrophages are immune cells found within the bone marrow microenvironment (BMME), of critical importance for leukemia development and progression. One defining feature of macrophages is their plasticity, which allows them to adapt to the variations in the microenvironment. While this adaptability is advantageous during wound healing, it can also be exploited in cancer scenarios. Thus, clinical and preclinical investigations that target macrophages as a therapeutic strategy appear promising. Existing research indicates that targeting macrophages could enhance the effectiveness of current AML treatments. This review addresses the importance of macrophages as therapeutic targets including relevant drugs investigated in clinical trials such as pexidartinib, magrolimab or bexmarilimab, but also provides new insights into lesser-known therapies, like macrophage receptor with a collagenous structure (MACRO) inhibitors and Toll-like receptor (TLR) agonists.
Collapse
Affiliation(s)
- Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Madalina Onciul
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
| | - Emilia Matei
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Pathology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Corina Joldes
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, 19-21 Croitorilor Str., 400162 Cluj-Napoca, Romania
| | - Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Alexandra Neaga
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Oana Serban
- Regina Maria” Regional Laboratory in Cluj-Napoca, 109 Observatorului Str., 400363 Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Ana Maria Nanut
- Regina Maria” Regional Laboratory in Cluj-Napoca, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| |
Collapse
|
24
|
Sabit H, Arneth B, Abdel-Ghany S, Madyan EF, Ghaleb AH, Selvaraj P, Shin DM, Bommireddy R, Elhashash A. Beyond Cancer Cells: How the Tumor Microenvironment Drives Cancer Progression. Cells 2024; 13:1666. [PMID: 39404428 PMCID: PMC11475877 DOI: 10.3390/cells13191666] [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: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Liver cancer represents a substantial global health challenge, contributing significantly to worldwide morbidity and mortality. It has long been understood that tumors are not composed solely of cancerous cells, but also include a variety of normal cells within their structure. These tumor-associated normal cells encompass vascular endothelial cells, fibroblasts, and various inflammatory cells, including neutrophils, monocytes, macrophages, mast cells, eosinophils, and lymphocytes. Additionally, tumor cells engage in complex interactions with stromal cells and elements of the extracellular matrix (ECM). Initially, the components of what is now known as the tumor microenvironment (TME) were thought to be passive bystanders in the processes of tumor proliferation and local invasion. However, recent research has significantly advanced our understanding of the TME's active role in tumor growth and metastasis. Tumor progression is now known to be driven by an intricate imbalance of positive and negative regulatory signals, primarily influenced by specific growth factors produced by both inflammatory and neoplastic cells. This review article explores the latest developments and future directions in understanding how the TME modulates liver cancer, with the aim of informing the design of novel therapies that target critical components of the TME.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt; (H.S.); (E.F.M.)
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr. 12, 35392 Giessen, Germany
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt;
| | - Engy F. Madyan
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt; (H.S.); (E.F.M.)
| | - Ashraf H. Ghaleb
- Department of Surgery, College of Medicine, Misr University for Science and Technology, Giza P.O. Box 77, Egypt;
- Department of Surgery, College of Medicine, Cairo University, Giza 12613, Egypt
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.S.); (R.B.)
| | - Dong M. Shin
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Ramireddy Bommireddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.S.); (R.B.)
| | - Ahmed Elhashash
- Department of Biology, Texas A&M University, 3258 TAMU I, College Station, TX 77843-3258, USA
| |
Collapse
|
25
|
Malik S, Sureka N, Ahuja S, Aden D, Zaheer S, Zaheer S. Tumor-associated macrophages: A sentinel of innate immune system in tumor microenvironment gone haywire. Cell Biol Int 2024; 48:1406-1449. [PMID: 39054741 DOI: 10.1002/cbin.12226] [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: 12/11/2023] [Revised: 06/10/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
The tumor microenvironment (TME) is a critical determinant in the initiation, progression, and treatment outcomes of various cancers. Comprising of cancer-associated fibroblasts (CAF), immune cells, blood vessels, and signaling molecules, the TME is often likened to the soil supporting the seed (tumor). Among its constituents, tumor-associated macrophages (TAMs) play a pivotal role, exhibiting a dual nature as both promoters and inhibitors of tumor growth. This review explores the intricate relationship between TAMs and the TME, emphasizing their diverse functions, from phagocytosis and tissue repair to modulating immune responses. The plasticity of TAMs is highlighted, showcasing their ability to adopt either protumorigenic or anti-tumorigenic phenotypes based on environmental cues. In the context of cancer, TAMs' pro-tumorigenic activities include promoting angiogenesis, inhibiting immune responses, and fostering metastasis. The manuscript delves into therapeutic strategies targeting TAMs, emphasizing the challenges faced in depleting or inhibiting TAMs due to their multifaceted roles. The focus shifts towards reprogramming TAMs to an anti-tumorigenic M1-like phenotype, exploring interventions such as interferons, immune checkpoint inhibitors, and small molecule modulators. Noteworthy advancements include the use of CSF1R inhibitors, CD40 agonists, and CD47 blockade, demonstrating promising results in preclinical and clinical settings. A significant section is dedicated to Chimeric Antigen Receptor (CAR) technology in macrophages (CAR-M cells). While CAR-T cells have shown success in hematological malignancies, their efficacy in solid tumors has been limited. CAR-M cells, engineered to infiltrate solid tumors, are presented as a potential breakthrough, with a focus on their development, challenges, and promising outcomes. The manuscript concludes with the exploration of third-generation CAR-M technology, offering insight into in-vivo reprogramming and nonviral vector approaches. In conclusion, understanding the complex and dynamic role of TAMs in cancer is crucial for developing effective therapeutic strategies. While early-stage TAM-targeted therapies show promise, further extensive research and larger clinical trials are warranted to optimize their targeting and improve overall cancer treatment outcomes.
Collapse
Affiliation(s)
- Shaivy Malik
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, Jamia Hamdard, New Delhi, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| |
Collapse
|
26
|
Peng Y, Liang S, Liu D, Ma K, Yun K, Zhou M, Hai L, Xu M, Chen Y, Wang Z. Multi-Metallic Nanosheets Reshaping Immunosuppressive Tumor Microenvironment through Augmenting cGAS-STING Innate Activation and Adaptive Immune Responses for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403347. [PMID: 39120546 PMCID: PMC11481177 DOI: 10.1002/advs.202403347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/14/2024] [Indexed: 08/10/2024]
Abstract
The highly immunosuppressive tumor microenvironment (TME) restricts the efficient activation of immune responses. To restore the surveillance of the immune system for robust activation, vast efforts are devoted to normalizing the TME. Here, a manganese-doped layered double hydroxide (Mn-LDH) is developed for potent anti-tumor immunity by reversing TME. Mn-LDH is synthesized via a one-step hydrothermal method. In addition to the inherent proton neutralization capacity of LDH, the introduction of manganese oxide endows LDH with an additional ability to produce oxygen. Mn-LDH effectively releases Mn2+ and Mg2+ upon exposure to TME with high levels of H+ and H2O2, which activates synthase-stimulator of interferon genes pathway and maintains the cytotoxicity of CD8+ T cells respectively, achieving a cascade-like role in innate and adaptive immunity. The locally administered Mn-LDH facilitated a "hot" network consisting of mature dendritic cells, M1-phenotype macrophages, as well as cytotoxic and helper T cells, significantly inhibiting the growth of primary and distal tumors. Moreover, the photothermal conversion capacity of Mn-LDH sparks more robust therapeutic effects in large established tumor models with a single administration and irradiation. Overall, this study guides the rational design of TME-modulating immunotherapeutics for robust immune activation, providing a clinical candidate for next-generation cancer immunotherapy.
Collapse
Affiliation(s)
- Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Kaiqing Yun
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Linna Hai
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Mengdi Xu
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Yiyang Chen
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
- Beijing Key Laboratory of Drug Delivery Technology and Novel FormulationInstitute of Materia MedicaChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| |
Collapse
|
27
|
Scimeca M, Giacobbi E, Servadei F, Palumbo V, Palumbo C, Finazzi-Agrò E, Albisinni S, Mauriello A, Albonici L. Prognostic Value of PlGF Upregulation in Prostate Cancer. Biomedicines 2024; 12:2194. [PMID: 39457506 PMCID: PMC11505493 DOI: 10.3390/biomedicines12102194] [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: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the second most commonly diagnosed cancer in men worldwide, with metastasis, particularly to bone, being the primary cause of mortality. Currently, prognostic markers like PSA levels and Gleason classification are limited in predicting metastasis, emphasizing the need for novel clinical biomarkers. New molecules predicting tumor progression have been identified over time. Some, such as the immune checkpoint inhibitors (ICIs) PD-1/PD-L1, have become valid markers as theranostic tools essential for prognosis and drug target therapy. However, despite the success of ICIs as an anti-cancer therapy for solid tumors, their efficacy in treating bone metastases has mainly proven ineffective, suggesting intrinsic resistance to this therapy in the bone microenvironment. This study explores the potential of immunological intratumoral biomarkers, focusing on placental growth factor (PlGF), Vascular Endothelial Growth Factor Receptor 1 (VEGFR1), and Programmed Cell Death Protein 1 (PD-1), in predicting bone metastasis formation. METHODS we analyzed PCa samples from patients with and without metastasis by immunohistochemical analysis. RESULTS Results revealed that PlGF expression is significantly higher in primary tumors of patients that developed metastasis within five years from the histological diagnosis. Additionally, PlGF expression correlates with increased VEGFR1 and PD-1 levels, as well as the presence of intratumoral M2 macrophages. CONCLUSIONS These findings suggest that PlGF contributes to an immunosuppressive environment, thus favoring tumor progression and metastatic process. Results here highlight the potential of integrating these molecular markers with existing prognostic tools to enhance the accuracy of metastasis prediction in PCa. By identifying patients at risk for metastasis, clinicians can tailor treatment strategies more effectively, potentially improving survival outcomes and quality of life. This study underscores the importance of further research into the role of intratumoral biomarkers in PCa management.
Collapse
Affiliation(s)
- Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy; (E.G.); (F.S.); (V.P.); (A.M.)
| | - Erica Giacobbi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy; (E.G.); (F.S.); (V.P.); (A.M.)
| | - Francesca Servadei
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy; (E.G.); (F.S.); (V.P.); (A.M.)
| | - Valeria Palumbo
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy; (E.G.); (F.S.); (V.P.); (A.M.)
| | - Camilla Palumbo
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Enrico Finazzi-Agrò
- Unit of Urology, Department of Surgical Sciences, Tor Vergata University, 00133 Rome, Italy; (E.F.-A.); (S.A.)
| | - Simone Albisinni
- Unit of Urology, Department of Surgical Sciences, Tor Vergata University, 00133 Rome, Italy; (E.F.-A.); (S.A.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy; (E.G.); (F.S.); (V.P.); (A.M.)
| | - Loredana Albonici
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
- Department of Biomedical Sciences, “Our Lady of Good Counsel” University, Rruga Dritan Hoxha, 1000 Tirana, Albania
| |
Collapse
|
28
|
Sulthana S, Shrestha D, Aryal S. Maximizing liposome tumor delivery by hybridizing with tumor-derived extracellular vesicles. NANOSCALE 2024; 16:16652-16663. [PMID: 39171636 PMCID: PMC11401241 DOI: 10.1039/d4nr02191f] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Extracellular vesicles (EVs) have gained widespread interest due to their potential in the diagnosis and treatment of inflammation, autoimmune diseases, and cancers. EVs are lipidic vesicles comprising vesicles of endosomal origin called exosomes, microvesicles from membrane shedding, and apoptotic bodies from programmed cell death membrane blebbing that carry complex sets of cargo from their cells of origin, including proteins, lipids, mRNA, and DNA. EVs are rich in integrin proteins that facilitate intrinsic cellular communication to deliver their cargo contents and can also be used as biomarkers to study respective cellular conditions. Within this background, we hypothesized that when these EVs are hybridized with synthetic liposomes, it would help navigate the hybrid construct in the complex biological environment to find its target. Toward this endeavor, we have hybridized a synthetic liposome with EVs (herein called LEVs) derived from mouse breast cancer (4T1 tumors) cells and incorporated a rhodamine-B/near-infrared fluorescent dye to investigate their potential for cellular targeting and tumor delivery. Using membrane extrusion, we have successfully hybridized both entities resulting in the formation of LEVs and characterized their colloidal properties and stability over a period. While EVs are broadly dispersed nano- and micron-sized vesicles, LEVs are engineered as monodispersed with an average hydrodynamic size of 140 ± 5. Using immunoblotting and ELISA, we monitored and quantified the EV-specific protein CD63 and other characteristic proteins such as CD9 and CD81, which were taken as a handle to ensure the reproducibility of EVs and thus LEVs. These LEVs were further challenged with mice bearing orthotopic 4T1 breast tumors and the LEV uptake was found to be maximum in tumors and organs like the liver, spleen, and lungs when compared to control PEGylated liposomes in live animal imaging. Likewise, the constructs were capable of finding lung metastasis as observed in ex vivo imaging. We anticipate that this study can open avenues for drug delivery solutions that are superior in target recognition.
Collapse
Affiliation(s)
- Shoukath Sulthana
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX 75799, USA.
| | - Dinesh Shrestha
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX 75799, USA.
| | - Santosh Aryal
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX 75799, USA.
| |
Collapse
|
29
|
Chen D, Tong W, Ang B, Bai Y, Dong W, Deng X, Wang C, Zhang Y. Revealing the crosstalk between LOX + fibroblast and M2 macrophage in gastric cancer by single-cell sequencing. BMC Cancer 2024; 24:1117. [PMID: 39251966 PMCID: PMC11382413 DOI: 10.1186/s12885-024-12861-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: 06/26/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND/AIMS Gastric cancer (GC) ranks among the prevalent types of cancer, and its progression is influenced by the tumor microenvironment (TME). A comprehensive comprehension of the TME associated with GC has the potential to unveil therapeutic targets of significance. METHODS The complexity and heterogeneity of TME interactions were revealed through our investigation using an integrated analysis of single-cell and bulk-tissue sequencing data. RESULTS We constructed a single-cell transcriptomic atlas of 150,913 cells isolated from GC patients. Our analysis revealed the intricate nature and heterogeneity of the GC TME and the metabolic properties of major cell types. Furthermore, two cell subtypes, LOX+ Fibroblasts and M2 Macrophages, were enriched in tumor tissue and related to the outcome of GC patients. In addition, LOX+ Fibroblasts were significantly associated with M2 macrophages. immunofluorescence double labeling indicated LOX+ Fibroblasts and M2 Macrophages were tightly localized in GC tissue. The two cell subpopulations strongly interacted in a hypoxic microenvironment, yielding an immunosuppressive phenotype. Our findings further suggest that LOX+ Fibroblasts may act as a trigger for inducing the differentiation of monocytes into M2 Macrophages via the IL6-IL6R signaling pathway. CONCLUSIONS Our study revealed the intricate and interdependent communication network between the fibroblast and macrophage subpopulations, which could offer valuable insights for targeted manipulation of the tumor microenvironment.
Collapse
Affiliation(s)
- Dapeng Chen
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Wen Tong
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Bing Ang
- Oncology Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yi Bai
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Wenhui Dong
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xiyue Deng
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Chunjiong Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yamin Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China.
| |
Collapse
|
30
|
Abstract
Osteosarcoma is the most common primary bone cancer in children and young adults. Limited progress has been made in improving the survival outcomes in patients with osteosarcoma over the past four decades. Especially in metastatic or recurrent osteosarcoma, the survival rate is extremely unsatisfactory. The treatment of osteosarcoma urgently needs breakthroughs. In recent years, immunotherapy has achieved good therapeutic effects in various solid tumors. Due to the low immunogenicity and immunosuppressive microenvironment of osteosarcoma, immunotherapy has not yet been approved in osteosarcoma patients. However, immune-based therapies, including immune checkpoint inhibitors, chimeric antigen receptor T cells, and bispecfic antibodies are in active clinical development. In addition, other immunotherapy strategies including modified-NK cells/macrophages, DC vaccines, and cytokines are still in the early stages of research, but they will be hot topics for future study. In this review, we showed the functions of cell components including tumor-promoting and tumor-suppressing cells in the tumor microenvironment of osteosarcoma, and summarized the preclinical and clinical research results of various immunotherapy strategies in osteosarcoma, hoping to provide new ideas for future research in this field.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xudong Yao
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
31
|
Foley KG, Adli M, Kim JJ. Single-nuclei sequencing of uterine serous carcinoma reveals racial differences in immune signaling. Proc Natl Acad Sci U S A 2024; 121:e2402998121. [PMID: 39133838 PMCID: PMC11348309 DOI: 10.1073/pnas.2402998121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Significant racial disparities exist between Black and White patients with uterine serous carcinoma (USC). While the reasons for these disparities are unclear, several studies have demonstrated significantly different rates of driver mutations between racial groups, including TP53. However, limited research has investigated the transcriptional differences of tumors or the composition of the tumor microenvironment (TME) between these groups. Here, we report the single-nuclei RNA-sequencing profiles of primary USC tumors from diverse racial backgrounds. We find that there are significant differences between the tumors of Black and White patients. Tumors from Black patients exhibited higher expression of specific genes associated with aggressiveness, such as PAX8, and axon guidance and synaptic signaling pathways. We also demonstrated that T cell populations are reduced in the tumor tissue compared to matched benign, while anti-inflammatory macrophage populations are retained within the TME. Furthermore, we investigated the connection between PAX8 overexpression and immunosuppression in USC through regulation of several cytokines and chemokines. Notably, we show that PAX8 activity can influence macrophage gene expression and protein secretion. These studies provide a detailed understanding of the USC transcriptome and TME, and identify differences in tumor biology from patients of different racial backgrounds.
Collapse
Affiliation(s)
- K. Grace Foley
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL60611
| | - Mazhar Adli
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL60611
| | - J. Julie Kim
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL60611
| |
Collapse
|
32
|
Liu K, Du S, Yang J, Li J, Wang S, Zhang Z, Luo W, Chen C, Yang J, Han X. Engineered bacterial membrane vesicle as safe and efficient nano-heaters to reprogram tumor microenvironment for enhanced immunotherapy. J Control Release 2024; 374:127-139. [PMID: 39122216 DOI: 10.1016/j.jconrel.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
The immunosuppressive tumor microenvironment (TME) in solid tumors often impedes the efficacy of immunotherapy. Bacterial outer membrane vesicles (OMVs), as a promising cancer vaccine that can potently stimulate immune responses, have garnered interest as a potential platform for cancer therapy. However, the low yield of OMVs limits their utilization. To address this limitation, we developed a novel approach to synthesize OMV-like multifunctional synthetic bacterial vesicles (SBVs) by pretreating bacteria with ampicillin and lysing them through sonication. Compared to OMVs, the yield of SBVs increased by 40 times. Additionally, the unique synthesis process of SBVs allows for the encapsulation of bacterial intracellular contents, endowing SBVs with the capability of delivering catalase (CAT) for tumor hypoxia relief and activating the host cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) signaling pathway. To overcome the toxicity of lipopolysaccharide (LPS) on the SBVs surface, we decorated SBVs with a biocompatible polydopamine (PDA) shell, which allowed TME reprogramming using SBVs to be conducted without adverse side effects. Additionally, the photosensitizer indocyanine green (ICG) was loaded into the PDA shell to induce immunogenic cell death and further improve the efficacy of immunotherapy. In summary, the SBVs-based therapeutic platform SBV@PDA/ICG (SBV@P/I) can synergistically elicit safe and potent tumor-specific antitumor responses through combined immunotherapy and phototherapy.
Collapse
Affiliation(s)
- Kunguo Liu
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shiyu Du
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Jiawei Yang
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Juanjuan Li
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shijie Wang
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhibin Zhang
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Wen Luo
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Chao Chen
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jingjing Yang
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xin Han
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
33
|
Zhang J, Zhou J, Tang L, Ma J, Wang Y, Yang H, Wang X, Fan W. Custom-Design of Multi-Stimuli-Responsive Degradable Silica Nanoparticles for Advanced Cancer-Specific Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400353. [PMID: 38651235 DOI: 10.1002/smll.202400353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/24/2024] [Indexed: 04/25/2024]
Abstract
Chemotherapy is crucial in oncology for combating malignant tumors but often encounters obatacles such as severe adverse effects, drug resistance, and biocompatibility issues. The advantages of degradable silica nanoparticles in tumor diagnosis and treatment lie in their ability to target drug delivery, minimizing toxicity to normal tissues while enhancing therapeutic efficacy. Moreover, their responsiveness to both endogenous and exogenous stimuli opens up new possibilities for integrating multiple treatment modalities. This review scrutinizes the burgeoning utility of degradable silica nanoparticles in combination with chemotherapy and other treatment modalities. Commencing the elucidation of degradable silica synthesis and degradation mechanisms, emphasis is placed on the responsiveness of these materials to endogenous (e.g., pH, redox reactions, hypoxia, and enzymes) and exogenous stimuli (e.g., light and high-intensity focused ultrasound). Moreover, this exploration delves into strategies harnessing degradable silica nanoparticles in chemotherapy alone, coupled with radiotherapy, photothermal therapy, photodynamic therapy, gas therapy, immunotherapy, starvation therapy, and chemodynamic therapy, elucidating multimodal synergies. Concluding with an assessment of advances, challenges, and constraints in oncology, despite hurdles, future investigations are anticipated to augment the role of degradable silica in cancer therapy. These insights can serve as a compass for devising more efficacious combined tumor treatment strategies.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, P. R. China
| | - Jiani Zhou
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | | | - Jiayi Ma
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Ying Wang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Xiaoxiao Wang
- Biochemical Engineering Research Center, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243032, P. R. China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 211198, P. R. China
| |
Collapse
|
34
|
Zhao Y, Ni Q, Zhang W, Yu S. Progress in reeducating tumor-associated macrophages in tumor microenvironment. Discov Oncol 2024; 15:312. [PMID: 39060648 PMCID: PMC11282027 DOI: 10.1007/s12672-024-01186-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
Malignant tumor, one of the most threatening diseases to human health, has been comprehensively treated with surgery, radiotherapy, chemotherapy and targeted therapy, but the prognosis has not always been ideal. In the past decade, immunotherapy has shown increased efficacy in tumor treatment; however, for immunotherapy to achieve its fullest potential, obstacles are to be conquered, among which tumor microenvironment (TME) has been widely investigated. In remodeling the tumor immune microenvironment to inhibit tumor progression, macrophages, as the most abundant innate immune population, play an irreplaceable role in the immune response. Therefore, how to remodel TME and alter the recruitment and polarization status of tumor-associated macrophages (TAM) has been of wide interest. In this context, nanoparticles, photodynamic therapy and other therapeutic approaches capable of affecting macrophage polarization have emerged. In this paper, we categorize and organize the existing means and methods for reprogramming TAM to provide ideas for clinical application of novel tumor-related therapies.
Collapse
Affiliation(s)
- Yiming Zhao
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China
| | - Qianyang Ni
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China
| | - Weijian Zhang
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China
| | - Suyang Yu
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China.
| |
Collapse
|
35
|
Saadh MJ, Mustafa MA, Malathi H, Ahluwalia G, Kaur S, Al-Dulaimi MAAH, Alubiady MHS, Zain Al-Abdeen SH, Shakier HG, Ali MS, Ahmad I, Abosaoda MK. Targeting the pancreatic tumor microenvironment by plant-derived products and their nanoformulations. Med Oncol 2024; 41:201. [PMID: 39001987 DOI: 10.1007/s12032-024-02443-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024]
Abstract
Pancreatic cancer remains a significant health issue with limited treatment options. The tumor stroma, a complex environment made up of different cells and proteins, plays a crucial role in tumor growth and chemoresistance. Targeting tumor stroma, consisting of diverse non-tumor cells such as fibroblasts, extracellular matrix (ECM), immune cells, and also pre-vascular cells is encouraging for remodeling solid cancers, such as pancreatic cancer. Remodeling the stroma of pancreas tumors can be suggested as a strategy for reducing resistance to chemo/immunotherapy. Several studies have shown that phytochemicals from plants can affect the tumor environment and have anti-cancer properties. By targeting key pathways involved in stromal activation, phytochemicals may disrupt communication between the tumor and stroma and make tumor cells more sensitive to different treatments. Additionally, phytochemicals have immunomodulatory and anti-angiogenic properties, all of which contribute to their potential in treating pancreatic cancer. This review will provide a detailed look at how phytochemicals impact the tumor stroma and their effects on pancreatic tumor growth, spread, and response to treatment. It will also explore the potential of combining phytochemicals with other treatment options like chemotherapy, immunotherapy, and radiation.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, University of Imam Jaafar AL-Sadiq, Baghdad, Iraq
| | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Gunveen Ahluwalia
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, 303012, Rajasthan, India
| | - Sumeet Kaur
- Department of Applied Sciences, Chandigarh Engineering Colleges, Chandigarh Group of Colleges, Jhanjeri, 140307, Mohali, Punjab, India
| | | | | | | | | | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
36
|
Guo S, Li Z, Zhou R, Feng J, Huang L, Ren B, Zhu J, Huang Y, Wu G, Cai H, Zhang Q, Ke Y, Guan T, Chen P, Xu Y, Yan C, Ou C, Shen Z. MRI-Guided Tumor Therapy Based on Synergy of Ferroptosis, Immunosuppression Reversal and Disulfidptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309842. [PMID: 38431935 DOI: 10.1002/smll.202309842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/19/2024] [Indexed: 03/05/2024]
Abstract
Triple negative breast cancer (TNBC) cells have a high demand for oxygen and glucose to fuel their growth and spread, shaping the tumor microenvironment (TME) that can lead to a weakened immune system by hypoxia and increased risk of metastasis. To disrupt this vicious circle and improve cancer therapeutic efficacy, a strategy is proposed with the synergy of ferroptosis, immunosuppression reversal and disulfidptosis. An intelligent nanomedicine GOx-IA@HMON@IO is successfully developed to realize this strategy. The Fe release behaviors indicate the glutathione (GSH)-responsive degradation of HMON. The results of titanium sulfate assay, electron spin resonance (ESR) spectra, 5,5'-Dithiobis-(2-nitrobenzoic acid (DTNB) assay and T1-weighted magnetic resonance imaging (MRI) demonstrate the mechanism of the intelligent iron atom (IA)-based cascade reactions for GOx-IA@HMON@IO, generating robust reactive oxygen species (ROS). The results on cells and mice reinforce the synergistic mechanisms of ferroptosis, immunosuppression reversal and disulfidptosis triggered by the GOx-IA@HMON@IO with the following steps: 1) GSH peroxidase 4 (GPX4) depletion by disulfidptosis; 2) IA-based cascade reactions; 3) tumor hypoxia reversal; 4) immunosuppression reversal; 5) GPX4 depletion by immunotherapy. Based on the synergistic mechanisms of ferroptosis, immunosuppression reversal and disulfidptosis, the intelligent nanomedicine GOx-IA@HMON@IO can be used for MRI-guided tumor therapy with excellent biocompatibility and safety.
Collapse
Affiliation(s)
- Shuai Guo
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Zongheng Li
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Ruilong Zhou
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Jie Feng
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Lin Huang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Bin Ren
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Jiaoyang Zhu
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Ya Huang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Guochao Wu
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Haobin Cai
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Qianqian Zhang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Yushen Ke
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Tianwang Guan
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Peier Chen
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Yikai Xu
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Chenggong Yan
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Caiwen Ou
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, China
| | - Zheyu Shen
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
37
|
Toledo B, Zhu Chen L, Paniagua-Sancho M, Marchal JA, Perán M, Giovannetti E. Deciphering the performance of macrophages in tumour microenvironment: a call for precision immunotherapy. J Hematol Oncol 2024; 17:44. [PMID: 38863020 PMCID: PMC11167803 DOI: 10.1186/s13045-024-01559-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/21/2024] [Indexed: 06/13/2024] Open
Abstract
Macrophages infiltrating tumour tissues or residing in the microenvironment of solid tumours are known as tumour-associated macrophages (TAMs). These specialized immune cells play crucial roles in tumour growth, angiogenesis, immune regulation, metastasis, and chemoresistance. TAMs encompass various subpopulations, primarily classified into M1 and M2 subtypes based on their differentiation and activities. M1 macrophages, characterized by a pro-inflammatory phenotype, exert anti-tumoural effects, while M2 macrophages, with an anti-inflammatory phenotype, function as protumoural regulators. These highly versatile cells respond to stimuli from tumour cells and other constituents within the tumour microenvironment (TME), such as growth factors, cytokines, chemokines, and enzymes. These stimuli induce their polarization towards one phenotype or another, leading to complex interactions with TME components and influencing both pro-tumour and anti-tumour processes.This review comprehensively and deeply covers the literature on macrophages, their origin and function as well as the intricate interplay between macrophages and the TME, influencing the dual nature of TAMs in promoting both pro- and anti-tumour processes. Moreover, the review delves into the primary pathways implicated in macrophage polarization, examining the diverse stimuli that regulate this process. These stimuli play a crucial role in shaping the phenotype and functions of macrophages. In addition, the advantages and limitations of current macrophage based clinical interventions are reviewed, including enhancing TAM phagocytosis, inducing TAM exhaustion, inhibiting TAM recruitment, and polarizing TAMs towards an M1-like phenotype. In conclusion, while the treatment strategies targeting macrophages in precision medicine show promise, overcoming several obstacles is still necessary to achieve an accessible and efficient immunotherapy.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén, E-23071, Spain
- Department of Medical Oncology, Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | - Linrui Zhu Chen
- Department of Medical Oncology, Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | - María Paniagua-Sancho
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, E-18100, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, Granada, E-18071, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E-18016, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, E-18100, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, Granada, E-18071, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E-18016, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén, E-23071, Spain.
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, E-18100, Spain.
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain.
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam UMC, VU University, Amsterdam, The Netherlands.
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, Pisa, 56017, Italy.
| |
Collapse
|
38
|
Xu D, Wang W, Wang D, Ding J, Zhou Y, Zhang W. Long noncoding RNA MALAT-1: A versatile regulator in cancer progression, metastasis, immunity, and therapeutic resistance. Noncoding RNA Res 2024; 9:388-406. [PMID: 38511067 PMCID: PMC10950606 DOI: 10.1016/j.ncrna.2024.01.015] [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: 11/14/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 03/22/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) are RNA transcripts longer than 200 nucleotides that do not code for proteins but have been linked to cancer development and metastasis. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT-1) influences crucial cancer hallmarks through intricate molecular mechanisms, including proliferation, invasion, angiogenesis, apoptosis, and the epithelial-mesenchymal transition (EMT). The current article highlights the involvement of MALAT-1 in drug resistance, making it a potential target to overcome chemotherapy refractoriness. It discusses the impact of MALAT-1 on immunomodulatory molecules, such as major histocompatibility complex (MHC) proteins and PD-L1, leading to immune evasion and hindering anti-tumor immune responses. MALAT-1 also plays a significant role in cancer immunology by regulating diverse immune cell populations. In summary, MALAT-1 is a versatile cancer regulator, influencing tumorigenesis, chemoresistance, and immunotherapy responses. Understanding its precise molecular mechanisms is crucial for developing targeted therapies, and therapeutic strategies targeting MALAT-1 show promise for improving cancer treatment outcomes. However, further research is needed to fully uncover the role of MALAT-1 in cancer biology and translate these findings into clinical applications.
Collapse
Affiliation(s)
- Dexin Xu
- Department of Orthopedics, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Wenhai Wang
- Department of Cardiology, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Duo Wang
- Department of Geriatrics, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Jian Ding
- Department of Electrodiagnosis, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Yunan Zhou
- Department of Orthopedics, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Wenbin Zhang
- Department of Cardiology, Jilin Province FAW General Hospital, Changchun, 130000, China
| |
Collapse
|
39
|
Li Y, Chen Y, Zhao C, Yang Y, Zhang M, Cheng H, Li Q, Wang M. Arenobufagin modulation of PCSK9-mediated cholesterol metabolism induces tumor-associated macrophages polarisation to inhibit hepatocellular carcinoma progression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155532. [PMID: 38493722 DOI: 10.1016/j.phymed.2024.155532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND The tumor microenvironment (TME) of hepatocellular carcinoma is heterogeneous enough to be prone to drug resistance and multidrug resistance during treatment, and reprogramming of cholesterol metabolism in TME mediates tumor-associated macrophages (TAMs) polarization, which has an impact on the regulation of malignant tumor progression. Arenobufagin (ARBU) was extracted and isolated from toad venom (purity ≥98 %), which is the main active ingredient of the traditional Chinese medicine Chan'su with good anti-tumor effects. PURPOSE To investigate the regulatory effect of ARBU on lipid metabolism in tumor microenvironment, interfere with macrophage polarization, and determine its mechanism of action on liver cancer progression. METHODS In this study, the inhibitory effect of ARBU on the proliferation of Hepa1-6 in C57 mice and the safety of administration were evaluated by establishing a transplanted tumor model of Hepa1-6 hepatocellular carcinoma mice and using 5-FU as a positive control drug. In addition, we constructed a co-culture system of Hepa1-6 cells and primary mouse macrophages to study the effects of ARBU on the polarization phenotypic transformation of macrophages and the proliferation and migration of hepatoma cells. The influence of ARBU on the metabolism of lipids in the hepatocellular carcinoma mouse model was investigated by combining it with lipidomics technology. The influence of ARBU on the PCSK9/LDL-R signaling pathway and macrophage polarization, which regulate cholesterol metabolism, was tested by using qRT-PCR, gene editing, IF, and WB. CONCLUSION ARBU significantly inhibited the proliferation of Hepa1-6 in vivo and in vitro, regulated cholesterol metabolism, and promoted the M1-type polarization of macrophages in the tumor microenvironment. ARBU inhibits cholesterol synthesis in the TME through the PCSK9/LDL-R signaling pathway, thereby blocking macrophage M2 polarization, promoting apoptosis of the tumor cells, and inhibiting their proliferation and migration.
Collapse
Affiliation(s)
- Yueyue Li
- Key Laboratory of Xin'an Medicine, Anhui Province Key Laboratory of R&D of Chinese Medicine, Ministry of Education, Anhui University of Traditional Chinese Medicine, 103 Meishan Road, Shushan District, Hefei City, Anhui Province 230038, China
| | - Yang Chen
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province 230022, China
| | - Cheng Zhao
- Anqing Petrochemical Hospital of Nanjing Gulou Hospital Group, Medical Oncology, Anqing City, Anhui Province 264000, China
| | - Yuting Yang
- Key Laboratory of Xin'an Medicine, Anhui Province Key Laboratory of R&D of Chinese Medicine, Ministry of Education, Anhui University of Traditional Chinese Medicine, 103 Meishan Road, Shushan District, Hefei City, Anhui Province 230038, China
| | - Mei Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province 230022, China
| | - Hui Cheng
- Key Laboratory of Xin'an Medicine, Anhui Province Key Laboratory of R&D of Chinese Medicine, Ministry of Education, Anhui University of Traditional Chinese Medicine, 103 Meishan Road, Shushan District, Hefei City, Anhui Province 230038, China
| | - Qinglin Li
- Key Laboratory of Xin'an Medicine, Anhui Province Key Laboratory of R&D of Chinese Medicine, Ministry of Education, Anhui University of Traditional Chinese Medicine, 103 Meishan Road, Shushan District, Hefei City, Anhui Province 230038, China.
| | - Meng Wang
- Key Laboratory of Xin'an Medicine, Anhui Province Key Laboratory of R&D of Chinese Medicine, Ministry of Education, Anhui University of Traditional Chinese Medicine, 103 Meishan Road, Shushan District, Hefei City, Anhui Province 230038, China.
| |
Collapse
|
40
|
Chen H, Li J, Cao D, Tang H. Construction of a Prognostic Model for Hepatocellular Carcinoma Based on Macrophage Polarization-Related Genes. J Hepatocell Carcinoma 2024; 11:857-878. [PMID: 38751862 PMCID: PMC11095518 DOI: 10.2147/jhc.s453080] [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: 12/22/2023] [Accepted: 05/07/2024] [Indexed: 05/18/2024] Open
Abstract
Background The progression of hepatocellular carcinoma (HCC) is related to macrophage polarization (MP). Our aim was to identify genes associated with MP in HCC patients and develop a prognostic model based on these genes. Results We successfully developed a prognostic model consisting of six MP-related genes (SCN4A, EBF3, ADGRB2, HOXD9, CLEC1B, and MSC) to calculate the risk score for each patient. Patients were then classified into high- and low-risk groups based on their median risk score. The performance of the MP-related prognostic model was evaluated using Kaplan-Meier and ROC curves, which yielded favorable results. Additionally, the nomogram demonstrated good clinical effectiveness and displayed consistent survival predictions with actual observations. Gene Set Enrichment Analysis (GSEA) revealed enrichment of pathways related to KRAS signaling downregulation, the G2M checkpoint, and E2F targets in the high-risk group. Conversely, pathways associated with fatty acid metabolism, xenobiotic metabolism, bile acid metabolism, and adipogenesis were enriched in the low-risk group. The risk score positively correlated with the number of invasion-related genes. Immune checkpoint expression differed significantly between the two groups. Patients in the high-risk group exhibited increased sensitivity to mitomycin C, cisplatin, gemcitabine, rapamycin, and paclitaxel, while those in the low-risk group showed heightened sensitivity to doxorubicin. These findings suggest that the high-risk group may have more invasive HCC with greater susceptibility to specific drugs. IHC staining revealed higher expression levels of SCN4A in HCC tissues. Furthermore, experiments conducted on HepG2 cells demonstrated that supernatants from cells with reduced SCN4A expression promoted M2 macrophage polarization marker, CD163 in THP-1 cells. Reduced SCN4A expression induced HCC-related genes, while increased SCN4A expression reduced their expression in HepG2 cells. Conclusion The MP-related prognostic model comprising six MPRGs can effectively predict HCC prognosis, infer invasiveness, and guide drug therapy. SCN4A is identified as a suppressor gene in HCC.
Collapse
Affiliation(s)
- Han Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, People’s Republic of China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Jianhao Li
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, People’s Republic of China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Dan Cao
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, People’s Republic of China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, People’s Republic of China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
41
|
Bergerud KMB, Berkseth M, Pardoll DM, Ganguly S, Kleinberg LR, Lawrence J, Odde DJ, Largaespada DA, Terezakis SA, Sloan L. Radiation Therapy and Myeloid-Derived Suppressor Cells: Breaking Down Their Cancerous Partnership. Int J Radiat Oncol Biol Phys 2024; 119:42-55. [PMID: 38042450 PMCID: PMC11082936 DOI: 10.1016/j.ijrobp.2023.11.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Radiation therapy (RT) has been a primary treatment modality in cancer for decades. Increasing evidence suggests that RT can induce an immunosuppressive shift via upregulation of cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). MDSCs inhibit antitumor immunity through potent immunosuppressive mechanisms and have the potential to be crucial tools for cancer prognosis and treatment. MDSCs interact with many different pathways, desensitizing tumor tissue and interacting with tumor cells to promote therapeutic resistance. Vascular damage induced by RT triggers an inflammatory signaling cascade and potentiates hypoxia in the tumor microenvironment (TME). RT can also drastically modify cytokine and chemokine signaling in the TME to promote the accumulation of MDSCs. RT activation of the cGAS-STING cytosolic DNA sensing pathway recruits MDSCs through a CCR2-mediated mechanism, inhibiting the production of type 1 interferons and hampering antitumor activity and immune surveillance in the TME. The upregulation of hypoxia-inducible factor-1 and vascular endothelial growth factor mobilizes MDSCs to the TME. After recruitment, MDSCs promote immunosuppression by releasing reactive oxygen species and upregulating nitric oxide production through inducible nitric oxide synthase expression to inhibit cytotoxic activity. Overexpression of arginase-1 on subsets of MDSCs degrades L-arginine and downregulates CD3ζ, inhibiting T-cell receptor reactivity. This review explains how radiation promotes tumor resistance through activation of immunosuppressive MDSCs in the TME and discusses current research targeting MDSCs, which could serve as a promising clinical treatment strategy in the future.
Collapse
Affiliation(s)
| | - Matthew Berkseth
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sudipto Ganguly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lawrence R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Departments of Pediatrics and Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | | | - Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
42
|
Jabbarzadeh Kaboli P, Chen HF, Babaeizad A, Roustai Geraylow K, Yamaguchi H, Hung MC. Unlocking c-MET: A comprehensive journey into targeted therapies for breast cancer. Cancer Lett 2024; 588:216780. [PMID: 38462033 DOI: 10.1016/j.canlet.2024.216780] [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: 12/20/2023] [Revised: 02/18/2024] [Accepted: 02/29/2024] [Indexed: 03/12/2024]
Abstract
Breast cancer is the most common malignancy among women, posing a formidable health challenge worldwide. In this complex landscape, the c-MET (cellular-mesenchymal epithelial transition factor) receptor tyrosine kinase (RTK), also recognized as the hepatocyte growth factor (HGF) receptor (HGFR), emerges as a prominent protagonist, displaying overexpression in nearly 50% of breast cancer cases. Activation of c-MET by its ligand, HGF, secreted by neighboring mesenchymal cells, contributes to a cascade of tumorigenic processes, including cell proliferation, metastasis, angiogenesis, and immunosuppression. While c-MET inhibitors such as crizotinib, capmatinib, tepotinib and cabozantinib have garnered FDA approval for non-small cell lung cancer (NSCLC), their potential within breast cancer therapy is still undetermined. This comprehensive review embarks on a journey through structural biology, multifaceted functions, and intricate signaling pathways orchestrated by c-MET across cancer types. Furthermore, we highlight the pivotal role of c-MET-targeted therapies in breast cancer, offering a clinical perspective on this promising avenue of intervention. In this pursuit, we strive to unravel the potential of c-MET as a beacon of hope in the fight against breast cancer, unveiling new horizons for therapeutic innovation.
Collapse
Affiliation(s)
- Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Hsiao-Fan Chen
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Ali Babaeizad
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Hirohito Yamaguchi
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan; Department of Biotechnology, Asia University, Taichung, 413, Taiwan.
| |
Collapse
|
43
|
Yan Q, Liu J, Liu Y, Wen Z, Jin D, Wang F, Gao L. Tumor-associated macrophage-derived exosomal miR21-5p promotes tumor angiogenesis by regulating YAP1/HIF-1α axis in head and neck squamous cell carcinoma. Cell Mol Life Sci 2024; 81:179. [PMID: 38602536 PMCID: PMC11009780 DOI: 10.1007/s00018-024-05210-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/12/2024]
Abstract
Extracellular vesicles (EVs) have recently received increasing attention as essential mediators of communication between tumor cells and their microenvironments. Tumor-associated macrophages (TAMs) play a proangiogenic role in various tumors, especially head and neck squamous cell carcinoma (HNSCC), and angiogenesis is closely related to tumor growth and metastasis. This research focused on exploring the mechanisms by which EVs derived from TAMs modulate tumor angiogenesis in HNSCC. Our results indicated that TAMs infiltration correlated positively with microvascular density in HNSCC. Then we collected and identified EVs from TAMs. In the microfluidic chip, TAMs derived EVs significantly enhanced the angiogenic potential of pHUVECs and successfully induced the formation of perfusable blood vessels. qPCR and immunofluorescence analyses revealed that EVs from TAMs transferred miR-21-5p to endothelial cells (ECs). And targeting miR-21-5p of TAMs could effectively inhibit TAM-EVs induced angiogenesis. Western blot and tube formation assays showed that miR-21-5p from TAM-EVs downregulated LATS1 and VHL levels but upregulated YAP1 and HIF-1α levels, and the inhibitors of YAP1 and HIF-1α could both reduce the miR-21-5p enhanced angiogenesis in HUVECs. The in vivo experiments further proved that miR-21-5p carried by TAM-EVs promoted the process of tumor angiogenesis via YAP1/HIF-1α axis in HNSCC. Conclusively, TAM-derived EVs transferred miR-21-5p to ECs to target the mRNA of LATS1 and VHL, which inhibited YAP1 phosphorylation and subsequently enhanced YAP1-mediated HIF-1α transcription and reduced VHL-mediated HIF-1α ubiquitination, contributing to angiogenesis in HNSCC. These findings present a novel regulatory mechanism of tumor angiogenesis, and miR-21-5p/YAP1/HIF-1α might be a potential therapeutic target for HNSCC.
Collapse
Affiliation(s)
- Quan Yan
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Dalian Key Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, People's Republic of China
| | - Jing Liu
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Dalian Key Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, People's Republic of China
| | - Yiding Liu
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Dalian Key Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, People's Republic of China
| | - Zhihao Wen
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Dalian Key Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, People's Republic of China
| | - Dong Jin
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Dalian Key Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, People's Republic of China
| | - Fu Wang
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China.
- Dalian Key Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, People's Republic of China.
- The Affiliated Stomatological Hospital of Dalian Medical University, Dalian, People's Republic of China.
| | - Lu Gao
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China.
- Dalian Key Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, People's Republic of China.
- The Affiliated Stomatological Hospital of Dalian Medical University, Dalian, People's Republic of China.
| |
Collapse
|
44
|
Huang Z, Xiao Z, Yu L, Liu J, Yang Y, Ouyang W. Tumor-associated macrophages in non-small-cell lung cancer: From treatment resistance mechanisms to therapeutic targets. Crit Rev Oncol Hematol 2024; 196:104284. [PMID: 38311012 DOI: 10.1016/j.critrevonc.2024.104284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) remains one of the leading causes of cancer-related deaths worldwide. Different treatment approaches are typically employed based on the stage of NSCLC. Common clinical treatment methods include surgical resection, drug therapy, and radiation therapy. However, with the introduction and utilization of immune checkpoint inhibitors, cancer treatment has entered a new era, completely revolutionizing the treatment landscape for various cancers and significantly improving overall patient survival. Concurrently, treatment resistance often poses a critical challenge, with many patients experiencing disease progression following an initial response due to treatment resistance. Increasing evidence suggests that the tumor microenvironment (TME) plays a pivotal role in treatment resistance. Tumor-associated macrophages (TAMs) within the TME can promote treatment resistance in NSCLC by secreting various cytokines activating signaling pathways, and interacting with other immune cells. Therefore, this article will focus on elucidating the key mechanisms of TAMs in treatment resistance and analyze how targeting TAMs can reduce the levels of treatment resistance in NSCLC, providing a comprehensive understanding of the principles and approaches to overcome treatment resistance in NSCLC.
Collapse
Affiliation(s)
- Zhenjun Huang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Ziqi Xiao
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Liqing Yu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jiayu Liu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yihan Yang
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang 330006, Jiangxi Province, China.
| | - Wenhao Ouyang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
45
|
Choi Y, Lee D, Kim NY, Seo I, Park NJY, Chong GO. Role of Tumor-Associated Macrophages in Cervical Cancer: Integrating Classical Perspectives with Recent Technological Advances. Life (Basel) 2024; 14:443. [PMID: 38672714 PMCID: PMC11051155 DOI: 10.3390/life14040443] [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: 02/16/2024] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in the tumor microenvironment, influencing cancer progression and contributing to poor prognosis. However, in cervical cancer (CC), their significance and involvement are relatively less studied than in other gynecological cancers such as ovarian and endometrial cancer. This review aims to provide an overview of TAMs, covering their origins and phenotypes and their impact on CC progression, along with major TAM-targeted therapeutic approaches. Furthermore, we advocate for the integration of cutting-edge research methodologies, such as single-cell RNA sequencing and spatial RNA sequencing, to enable in-depth and comprehensive investigations into TAMs in CC, which would be beneficial in leading to more personalized and effective immunotherapy strategies for patients with CC.
Collapse
Affiliation(s)
- Yeseul Choi
- Graduate Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.C.); (D.L.); (N.Y.K.)
| | - Donghyeon Lee
- Graduate Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.C.); (D.L.); (N.Y.K.)
| | - Na Young Kim
- Graduate Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.C.); (D.L.); (N.Y.K.)
| | - Incheol Seo
- Department of Immunology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea;
| | - Nora Jee-Young Park
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea;
- Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu 41404, Republic of Korea
| | - Gun Oh Chong
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea;
- Department of Obstetrics and Gynecology, Kyungpook National University Chilgok Hospital, Daegu 41404, Republic of Korea
| |
Collapse
|
46
|
Li SL, Hou HY, Chu X, Zhu YY, Zhang YJ, Duan MD, Liu J, Liu Y. Nanomaterials-Involved Tumor-Associated Macrophages' Reprogramming for Antitumor Therapy. ACS NANO 2024; 18:7769-7795. [PMID: 38420949 DOI: 10.1021/acsnano.3c12387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Tumor-associated macrophages (TAMs) play pivotal roles in tumor development. As primary contents of tumor environment (TME), TAMs secrete inflammation-related substances to regulate tumoral occurrence and development. There are two kinds of TAMs: the tumoricidal M1-like TAMs and protumoral M2-like TAMs. Reprogramming TAMs from immunosuppressive M2 to immunocompetent M1 phenotype is considered a feasible way to improve immunotherapeutic efficiency. Notably, nanomaterials show great potential for biomedical fields due to their controllable structures and properties. There are many types of nanomaterials that exhibit great regulatory activities for TAMs' reprogramming. In this review, the recent progress of nanomaterials-involved TAMs' reprogramming is comprehensively discussed. The various nanomaterials for TAMs' reprogramming and the reprogramming strategies are summarized and introduced. Additionally, the challenges and perspectives of TAMs' reprogramming for efficient therapy are discussed, aiming to provide inspiration for TAMs' regulator design and promote the development of TAMs-mediated immunotherapy.
Collapse
Affiliation(s)
- Shu-Lan Li
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Hua-Ying Hou
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Xu Chu
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Ying Zhu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Juan Zhang
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Meng-Die Duan
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Junyi Liu
- Albany Medical College, New York 12208, United States
| | - Yi Liu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, P. R. China
| |
Collapse
|
47
|
Yoo YJ, Eun R, Park HM, Kim S, Park SH, Heo J, Lim YT. Nanoengineered Macrophages Armed with TLR7/8 Agonist Enhance Remodeling of Immunosuppressive Tumor Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307694. [PMID: 37967333 DOI: 10.1002/smll.202307694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/01/2023] [Indexed: 11/17/2023]
Abstract
Although adoptive cell-based therapy is illuminated as one of the promising approaches in cancer immunotherapy, it shows low antitumor efficacy because transferred cells adapt and alter toward a pro-tumoral phenotype in response to the tumor's immunosuppressive milieu. Herein, nanoengineered macrophages anchored with functional liposome armed with cholesterol-conjugated Toll-like receptor 7/8 agonist (masked TLR7/8a, m7/8a) are generated to overcome the shortcomings of current macrophage-based therapies and enhance the remodeling of the immunosuppressive tumor microenvironment (TME). The liposome-anchored macrophages (LAMΦ-m7/8a), are fabricated by anchoring dibenzocyclooctyne-modified liposome(m7/8a) onto azido-expressing macrophages via a bio-orthogonal click reaction, are continuously invigorated due to the slow internalization of liposome(m7/8a) and sustained activation. LAMΦ-m7/8a secreted ≈3 and 33-fold more IL-6 and TNF-α than conventional M1-MΦ, maintained the M1 phenotype, and phagocytosed tumor cells for up to 48 h in vitro. Both intratumoral and intravenous injections of LAMΦ-m7/8a induced effective antitumor efficacy when treated in combination with doxorubicin-loaded liposomes in 4T1-tumor bearing mice. It not only increases the infiltration of antigen-specific CD8+ T cells secreting granzyme B, IFN-γ, and TNF-α within the TME, but also reduces myeloid-derived suppressor cells. These results suggest that LAMΦ-m7/8a may provide a suitable alternative to next-generation cell-based therapy platform.
Collapse
Affiliation(s)
- Yeon Jeong Yoo
- Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ryounho Eun
- Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hye Min Park
- Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Suhyeon Kim
- Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sei Hyun Park
- Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Janghun Heo
- Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yong Taik Lim
- Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
48
|
Cheng S, Wang H, Kang X, Zhang H. Immunotherapy Innovations in the Fight against Osteosarcoma: Emerging Strategies and Promising Progress. Pharmaceutics 2024; 16:251. [PMID: 38399305 PMCID: PMC10892906 DOI: 10.3390/pharmaceutics16020251] [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: 11/13/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Immunosuppressive elements within the tumor microenvironment are the primary drivers of tumorigenesis and malignant advancement. The presence, as well as the crosstalk between myeloid-derived suppressor cells (MDSCs), osteosarcoma-associated macrophages (OS-Ms), regulatory T cells (Tregs), and endothelial cells (ECs) with osteosarcoma cells cause the poor prognosis of OS. In addition, the consequent immunosuppressive factors favor the loss of treatment potential. Nanoparticles offer a means to dynamically and locally manipulate immuno-nanoparticles, which present a promising strategy for transforming OS-TME. Additionally, chimeric antigen receptor (CAR) technology is effective in combating OS. This review summarizes the essential mechanisms of immunosuppressive cells in the OS-TME and the current immune-associated strategies. The last part highlights the limitations of existing therapies and offers insights into future research directions.
Collapse
Affiliation(s)
- Shigao Cheng
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Orthopedics, Hunan Loudi Central Hospital, Loudi 417000, China
| | - Huiyuan Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuejia Kang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Hui Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
49
|
Foglietta F, Panzanelli P, Pizzo R, Giacone M, Pepa CD, Durando G, Serpe L, Canaparo R. Evaluation of the cytotoxic and immunomodulatory effects of sonodynamic therapy in human pancreatic cancer spheroids. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 251:112842. [PMID: 38232641 DOI: 10.1016/j.jphotobiol.2024.112842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/27/2023] [Accepted: 01/07/2024] [Indexed: 01/19/2024]
Abstract
Sonodynamic therapy (SDT) exploits the energy generated by ultrasound (US) to activate sound-sensitive drugs (sonosensitizers), leading to the generation of reactive oxygen species (ROS) and cancer cell death. Two-dimensional (2D) and three-dimensional (3D) cultures of human pancreatic cancer BxPC-3 cells were chosen as the models with which to investigate the therapeutic effects of the US-activated sonosensitizer IR-780 as pancreatic cancer is still one of the most lethal types of cancer. The effects of SDT, including ROS production, cancer cell death and immunogenic cell death (ICD), were extensively investigated. When subjected to US, IR-780 triggered significant ROS production and caused cancer cell death after 24 h (p ≤ 0.01). Additionally, the activation of dendritic cells (DCs) led to an effective immune response against the cancer cells undergoing SDT-induced death. BxPC-3 spheroids were developed and studied extensively to validate the findings observed in 2D BxPC-3 cell cultures. An analysis of the pancreatic cancer spheroid section revealed significant SDT-induced cancer cell death after 48 h after the treatment (p ≤ 0.01), with this being accompanied by the presence of SDT-induced damage-associated molecular patterns (DAMPs), such as calreticulin (CRT) and high mobility group box 1 (HMGB1). In conclusion, the data obtained demonstrates the anticancer efficacy of SDT and its immunomodulatory potential via action as an ICD-inducer.
Collapse
Affiliation(s)
- Federica Foglietta
- Department of Drug Science and Technology, University of Torino, Via Pietro Giuria 13, 10125 Torino, Italy.
| | - Patrizia Panzanelli
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Torino, Italy.
| | - Riccardo Pizzo
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Torino, Italy.
| | - Marta Giacone
- Department of Drug Science and Technology, University of Torino, Via Pietro Giuria 13, 10125 Torino, Italy.
| | - Carlo Della Pepa
- Department of Drug Science and Technology, University of Torino, Via Pietro Giuria 13, 10125 Torino, Italy.
| | - Gianni Durando
- National Institute of Metrological Research (INRIM), Strada delle Cacce 91, 10135 Torino, Italy.
| | - Loredana Serpe
- Department of Drug Science and Technology, University of Torino, Via Pietro Giuria 13, 10125 Torino, Italy.
| | - Roberto Canaparo
- Department of Drug Science and Technology, University of Torino, Via Pietro Giuria 13, 10125 Torino, Italy.
| |
Collapse
|
50
|
Qian Y, Yin Y, Zheng X, Liu Z, Wang X. Metabolic regulation of tumor-associated macrophage heterogeneity: insights into the tumor microenvironment and immunotherapeutic opportunities. Biomark Res 2024; 12:1. [PMID: 38185636 PMCID: PMC10773124 DOI: 10.1186/s40364-023-00549-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/12/2023] [Indexed: 01/09/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are a heterogeneous population that play diverse functions in tumors. Their identity is determined not only by intrinsic factors, such as origins and transcription factors, but also by external signals from the tumor microenvironment (TME), such as inflammatory signals and metabolic reprogramming. Metabolic reprogramming has rendered TAM to exhibit a spectrum of activities ranging from pro-tumorigenic to anti-tumorigenic, closely associated with tumor progression and clinical prognosis. This review implicates the diversity of TAM phenotypes and functions, how this heterogeneity has been re-evaluated with the advent of single-cell technologies, and the impact of TME metabolic reprogramming on TAMs. We also review current therapies targeting TAM metabolism and offer new insights for TAM-dependent anti-tumor immunotherapy by focusing on the critical role of different metabolic programs in TAMs.
Collapse
Affiliation(s)
- Yujing Qian
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yujia Yin
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaocui Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhaoyuan Liu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|