1
|
Zhong H, Zhou S, Yin S, Qiu Y, Liu B, Yu H. Tumor microenvironment as niche constructed by cancer stem cells: Breaking the ecosystem to combat cancer. J Adv Res 2025; 71:279-296. [PMID: 38866179 DOI: 10.1016/j.jare.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/27/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a distinct subpopulation of cancer cells with the capacity to constantly self-renew and differentiate, and they are the main driver in the progression of cancer resistance and relapse. The tumor microenvironment (TME) constructed by CSCs is the "soil" adapted to tumor growth, helping CSCs evade immune killing, enhance their chemical resistance, and promote cancer progression. AIM OF REVIEW We aim to elaborate the tight connection between CSCs and immunosuppressive components of the TME. We attempt to summarize and provide a therapeutic strategy to eradicate CSCs based on the destruction of the tumor ecological niche. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three main key concepts. First, we highlight that CSCs recruit and transform normal cells to construct the TME, which further provides ecological niche support for CSCs. Second, we describe the main characteristics of the immunosuppressive components of the TME, targeting strategies and summarize the progress of corresponding drugs in clinical trials. Third, we explore the multilevel insights of the TME to serve as an ecological niche for CSCs.
Collapse
Affiliation(s)
- Hao Zhong
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shiyue Zhou
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shuangshuang Yin
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| | - Haiyang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| |
Collapse
|
2
|
Tantray J, Patel A, Parveen H, Prajapati B, Prajapati J. Nanotechnology-based biomedical devices in the cancer diagnostics and therapy. Med Oncol 2025; 42:50. [PMID: 39828813 DOI: 10.1007/s12032-025-02602-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Nanotechnology has significantly transformed the field of cancer diagnostics and therapeutics by introducing advanced biomedical devices. These nanotechnology-based devices exhibit remarkable capabilities in detecting and treating various cancers, addressing the limitations of traditional approaches, such as limited specificity and sensitivity. This review aims to explore the advancements in nanotechnology-driven biomedical devices, emphasizing their role in the diagnosis and treatment of cancer. Through a comprehensive analysis, we evaluate various nanotechnology-based devices across different cancer types, detailing their diagnostic and therapeutic effectiveness. The review also discusses FDA-approved nanotechnology products, patents, and regulatory trends, highlighting the innovation and clinical impact in oncology. Nanotechnology-based devices, including nanobots, smart pills, and multifunctional nanoparticles, enable precise targeting and treatment, reducing adverse effects on healthy tissues. Devices such as DNA-based nanorobots, quantum dots, and biodegradable stents offer noninvasive diagnostic and therapeutic options, showing high efficacy in preclinical and clinical settings. FDA-approved products underscore the acceptance of these technologies. Nanotechnology-based biomedical devices offer a promising future for oncology, with the potential to revolutionize cancer care through early detection, targeted treatment, and minimal side effects. Continued research and technological improvements are essential to fully realize their potential in personalized cancer therapy.
Collapse
Affiliation(s)
- Junaid Tantray
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India
| | - Akhilesh Patel
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India
| | - Hiba Parveen
- Faculty of Pharmacy, Veer Madho Singh Bhandari Uttrakhand Technical University, Dehradun, India
| | - Bhupendra Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
| | - Jigna Prajapati
- Faculty of Computer Application, Ganpat University, Mehsana, Gujarat, 384012, India.
| |
Collapse
|
3
|
Zhou Y, Zhang Y, Zhou Y, Gu Y, Chen Y, Wang J. Bioinformation study of immune microenvironment characteristics of disulfidptosis-related subtypes in ovarian cancer and prognostic model construction. Discov Oncol 2025; 16:18. [PMID: 39775403 PMCID: PMC11711411 DOI: 10.1007/s12672-025-01752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE Ovarian cancer significantly impacts women's reproductive health and remains challenging to diagnose and treat. Despite advancements in understanding DNA repair mechanisms and identifying novel therapeutic targets, additional strategies are still needed. Recently, a novel form of cell death called disulfidptosis, which is triggered by glucose deprivation, has been linked to treatment resistance and changes in the tumor microenvironment (TME). However, its role in ovarian cancer is not well understood. METHODS Bioinformatics analysis was performed on RNA-seq data from TCGA and GEO databases to identify disulfidptosis-related genes in ovarian cancer. Differential expression analysis and pathway enrichment were conducted, followed by the development of a prognostic model using LASSO Cox regression, validated with GEO datasets (GSE13876, GSE26712). Clinical samples were analyzed using quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC) to validate gene expression. RESULTS This study identified disulfidptosis-related gene subtypes in ovarian cancer and demonstrated their influence on the tumor microenvironment (TME), immunotherapy responses, and patient prognosis. Six genes (IFNB1, IGF2, CD40LG, IL1B, IL21, CD38) associated with disulfidptosis were identified and incorporated into a prognostic model. This model predicted patient outcomes and was validated externally. Clinical validation showed its accuracy in predicting progression-free survival and resistance to platinum-based chemotherapy. CONCLUSION Our findings highlight the significant impact of disulfidptosis-related genes on the ovarian cancer tumor microenvironment, providing insights that could support the development of clinical evaluations and personalized treatment strategies.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yuhong Zhang
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University; Suzhou Municipal Hospital, Suzhou, Jiangsu, China
| | - Yang Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanzheng Gu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Juan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
4
|
Tan R, Wen M, Yang W, Zhan D, Zheng N, Liu M, Zhu F, Chen X, Wang M, Yang S, Xie B, He Q, Yuan K, Sun L, Wang Y, Qin J, Zhang Y. Integrated proteomics and scRNA-seq analyses of ovarian cancer reveal molecular subtype-associated cell landscapes and immunotherapy targets. Br J Cancer 2025; 132:111-125. [PMID: 39548315 PMCID: PMC11723995 DOI: 10.1038/s41416-024-02894-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) represents the most lethal gynaecological malignancy, yet understanding the connections between its molecular subtypes and their therapeutic implications remains incomplete. METHODS We conducted mass spectrometry-based proteomics analyses of 154 EOC tumour samples and 29 normal fallopian tubes, and single-cell RNA sequencing (scRNA-seq) analyses of an additional eight EOC tumours to classify proteomic subtypes and assess their cellular ecosystems and clinical significance. The efficacy of identified therapeutic targets was evaluated in patient-derived xenograft (PDX) and orthotopic mouse models. RESULTS We identified four proteomic subtypes with distinct clinical relevance: malignant proliferative (C1), immune infiltrating (C2), Fallopian-like (C3) and differentiated (C4) subtypes. C2 subtype was characterized by lymphocyte infiltration, notably an increased presence of GZMK CD8+ T cells and phagocytosis-like MRC+ macrophages. Additionally, we identified CD40 as a specific prognostic factor for C2 subtype. The interaction between CD40+ phagocytosis-like macrophages and CD40RL+ IL17R CD4+ T cells was correlated with a favourable prognosis. Finally, we established a druggable landscape for non-immune EOC patients and verified a TYMP inhibitor as a promising therapeutic strategy. CONCLUSIONS Our study refines the current immune subtype for EOC, highlighting CD40 agonists as promising therapies for C2 subtype patients and targeting TYMP for non-immune patients.
Collapse
Affiliation(s)
- Rong Tan
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China.
| | - Ming Wen
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenqing Yang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China
| | - Dongdong Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Beijing Pineal Diagnostics Co., Ltd., Beijing, China
| | - Nairen Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Fang Zhu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China
| | - Xiaodan Chen
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China
| | - Meng Wang
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
| | - Siyu Yang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China
| | - Bin Xie
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Qiongqiong He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Kai Yuan
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, China
| | - Lunquan Sun
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, China
- Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Yu Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China.
| |
Collapse
|
5
|
Macciò A, Madeddu C. Cancer is not a guest. Transl Oncol 2025; 51:102162. [PMID: 39490055 PMCID: PMC11576533 DOI: 10.1016/j.tranon.2024.102162] [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: 05/19/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
In a recent article titled "Embracing Cancer Complexity: Hallmarks of Systemic Disease" published in Cell, Swaton et al. propose the idea of cancer as a guest that develops within a host. They discuss the possible causes and events of neoplastic cell dysregulations within an organism, highlighting events such as cachexia and thrombosis. However, we believe that to understand cancer-associated phenomena better, cancer cannot be considered a guest. In reality, cancer is born, develops, and spreads within its environment. It does not come from outside but instead uses the same system in which it lives to promote its death plan. Indeed, today we know that cancer not only causes local symptoms in the affected organ but also leads to systemic symptoms, which are evidence of inflammation associated with cancer. Inflammation is vital in controlling oncogenesis and neoplastic proliferation during the resistance phase, which is a critical moment for the immune system to demonstrate its effectiveness. However, if the immune system causes immunopathological damage, it may lead to necrosis and eventually to the tolerance phase, which can result in systemic symptoms. Understanding these phenomena thoroughly explains thrombophilia, anemia, sarcopenia, and iron metabolism disruption in advanced-stage neoplastic patients. The concept of the microenvironment takes on a different meaning in this context. The same cells that should oppose cancer in the tolerance phase now participate in a process that self-maintains, favoring the growth of the cancer and its death plan. The exact knowledge of these mechanisms is a more modern translational approach to treating cancer and its related symptoms.
Collapse
Affiliation(s)
- Antonio Macciò
- Gynecology and Gynecologic Oncology Unit, Department of Oncological Surgery-ARNAS G. Brotzu; Department of Surgical Sciences, University of Cagliari, 09100 Cagliari, Italy.
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, 09100 Cagliari, Italy
| |
Collapse
|
6
|
Zhang S, Wang H. Targeting the lung tumour stroma: harnessing nanoparticles for effective therapeutic interventions. J Drug Target 2025; 33:60-86. [PMID: 39356091 DOI: 10.1080/1061186x.2024.2410462] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Lung cancer remains an influential global health concern, necessitating the development of innovative therapeutic strategies. The tumour stroma, which is known as tumour microenvironment (TME) has a central impact on tumour expansion and treatment resistance. The stroma of lung tumours consists of numerous cells and molecules that shape an environment for tumour expansion. This environment not only protects tumoral cells against immune system attacks but also enables tumour stroma to attenuate the action of antitumor drugs. This stroma consists of stromal cells like cancer-associated fibroblasts (CAFs), suppressive immune cells, and cytotoxic immune cells. Additionally, the presence of stem cells, endothelial cells and pericytes can facilitate tumour volume expansion. Nanoparticles are hopeful tools for targeted drug delivery because of their extraordinary properties and their capacity to devastate biological obstacles. This review article provides a comprehensive overview of contemporary advancements in targeting the lung tumour stroma using nanoparticles. Various nanoparticle-based approaches, including passive and active targeting, and stimuli-responsive systems, highlighting their potential to improve drug delivery efficiency. Additionally, the role of nanotechnology in modulating the tumour stroma by targeting key components such as immune cells, extracellular matrix (ECM), hypoxia, and suppressive elements in the lung tumour stroma.
Collapse
Affiliation(s)
- Shushu Zhang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Hui Wang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
7
|
Liu Y, Xiao H, Zeng H, Xiang Y. Beyond tumor‑associated macrophages involved in spheroid formation and dissemination: Novel insights for ovarian cancer therapy (Review). Int J Oncol 2024; 65:117. [PMID: 39513610 PMCID: PMC11575928 DOI: 10.3892/ijo.2024.5705] [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: 05/24/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
Ovarian cancer (OC) is the most common and deadly malignant tumor of the female reproductive system. When OC cells detach from the primary tumor and enter the ascitic microenvironment, they are present as individual cells or multicellular spheroids in ascites. These spheroids, composed of cancer and non‑malignant cells, are metastatic units and play a crucial role in the progression of OC. However, little is known about the mechanism of spheroid formation and dissemination. Tumor‑associated macrophages (TAMs) in the center of spheroids are key in spheroid formation and metastasis and provide a potential target for OC therapy. The present review summarizes the key biological features of spheroids, focusing on the role of TAMs in spheroid formation, survival and peritoneal metastasis, and the strategies targeting TAMs to provide new insights in treating OC.
Collapse
Affiliation(s)
- Yuchen Liu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Haoyue Xiao
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Hai Zeng
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| |
Collapse
|
8
|
Van Kleunen LB, Ahmadian M, Post MD, Wolsky RJ, Rickert C, Jordan KR, Hu J, Richer JK, Brubaker LW, Marjon N, Behbakht K, Sikora MJ, Bitler BG, Clauset A. The Spatial Structure of the Tumor Immune Microenvironment Can Explain and Predict Patient Response in High-Grade Serous Carcinoma. Cancer Immunol Res 2024; 12:1492-1507. [PMID: 39115368 PMCID: PMC11534564 DOI: 10.1158/2326-6066.cir-23-1109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/20/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024]
Abstract
Ovarian cancer is the deadliest gynecologic malignancy, and therapeutic options and mortality rates over the last three decades have largely not changed. Recent studies indicate that the composition of the tumor immune microenvironment (TIME) influences patient outcomes. To improve spatial understanding of the TIME, we performed multiplexed ion beam imaging on 83 human high-grade serous carcinoma tumor samples, identifying approximately 160,000 cells across 23 cell types. From the 77 of these samples that met inclusion criteria, we generated composition features based on cell type proportions, spatial features based on the distances between cell types, and spatial network features representing cell interactions and cell clustering patterns, which we linked to traditional clinical and IHC variables and patient overall survival (OS) and progression-free survival (PFS) outcomes. Among these features, we found several significant univariate correlations, including B-cell contact with M1 macrophages (OS HR = 0.696; P = 0.011; PFS HR = 0.734; P = 0.039). We then used high-dimensional random forest models to evaluate out-of-sample predictive performance for OS and PFS outcomes and to derive relative feature importance scores for each feature. The top model for predicting low or high PFS used TIME composition and spatial features and achieved an average AUC score of 0.71. The results demonstrate the importance of spatial structure in understanding how the TIME contributes to treatment outcomes. Furthermore, the present study provides a generalizable roadmap for spatial analyses of the TIME in ovarian cancer research.
Collapse
Affiliation(s)
| | - Mansooreh Ahmadian
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Miriam D Post
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Rebecca J Wolsky
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Christian Rickert
- Department of Immunology and Microbiology, The University of Colorado Anschutz Medical Campus
| | - Kimberly R. Jordan
- Department of Immunology and Microbiology, The University of Colorado Anschutz Medical Campus
| | - Junxiao Hu
- Department of Pediatrics, Cancer Center Biostatistics Core, University of Colorado Anschutz Medical Campus, CO, USA
| | - Jennifer K. Richer
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | | | - Nicole Marjon
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Kian Behbakht
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Matthew J. Sikora
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Benjamin G. Bitler
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Aaron Clauset
- Department of Computer Science, University of Colorado, Boulder, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
9
|
Yamanaka K, Koma YI, Urakami S, Takahashi R, Nagamata S, Omori M, Torigoe R, Yokoo H, Nakanishi T, Ishihara N, Tsukamoto S, Kodama T, Nishio M, Shigeoka M, Yokozaki H, Terai Y. YKL40/Integrin β4 Axis Induced by the Interaction between Cancer Cells and Tumor-Associated Macrophages Is Involved in the Progression of High-Grade Serous Ovarian Carcinoma. Int J Mol Sci 2024; 25:10598. [PMID: 39408927 PMCID: PMC11477481 DOI: 10.3390/ijms251910598] [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: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Macrophages in the tumor microenvironment, termed tumor-associated macrophages (TAMs), promote the progression of various cancer types. However, many mechanisms related to tumor-stromal interactions in epithelial ovarian cancer (EOC) progression remain unclear. High-grade serous ovarian carcinoma (HGSOC) is the most malignant EOC subtype. Herein, immunohistochemistry was performed on 65 HGSOC tissue samples, revealing that patients with a higher infiltration of CD68+, CD163+, and CD204+ macrophages had a poorer prognosis. We subsequently established an indirect co-culture system between macrophages and EOC cells, including HGSOC cells. The co-cultured macrophages showed increased expression of the TAM markers CD163 and CD204, and the co-cultured EOC cells exhibited enhanced proliferation, migration, and invasion. Cytokine array analysis revealed higher YKL40 secretion in the indirect co-culture system. The addition of YKL40 increased proliferation, migration, and invasion via extracellular signal-regulated kinase (Erk) signaling in EOC cells. The knockdown of integrin β4, one of the YKL40 receptors, suppressed YKL40-induced proliferation, migration, and invasion, as well as Erk phosphorylation in some EOC cells. Database analysis showed that high-level expression of YKL40 and integrin β4 correlated with a poor prognosis in patients with serous ovarian carcinoma. Therefore, the YKL40/integrin β4 axis may play a role in ovarian cancer progression.
Collapse
Affiliation(s)
- Keitaro Yamanaka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.T.); (S.N.); (Y.T.)
| | - Yu-ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Satoshi Urakami
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Ryosuke Takahashi
- Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.T.); (S.N.); (Y.T.)
| | - Satoshi Nagamata
- Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.T.); (S.N.); (Y.T.)
| | - Masaki Omori
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Rikuya Torigoe
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastrointestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Hiroki Yokoo
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastrointestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Takashi Nakanishi
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastrointestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Nobuaki Ishihara
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Shuichi Tsukamoto
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Takayuki Kodama
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Mari Nishio
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Manabu Shigeoka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Yoshito Terai
- Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.T.); (S.N.); (Y.T.)
| |
Collapse
|
10
|
Cruz LS, Robinson M, Stevenson D, Amador IC, Jordan GJ, Valencia S, Navarrete C, House CD. Chemotherapy Enriches for Proinflammatory Macrophage Phenotypes that Support Cancer Stem-Like Cells and Disease Progression in Ovarian Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:2638-2652. [PMID: 39287565 PMCID: PMC11464072 DOI: 10.1158/2767-9764.crc-24-0311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
High-grade serous ovarian cancer remains a poorly understood disease with a high mortality rate. Although most patients respond to cytotoxic therapies, a majority will experience recurrence. This may be due to a minority of drug-resistant cancer stem-like cells (CSC) that survive chemotherapy and are capable of repopulating heterogeneous tumors. It remains unclear how CSCs are supported in the tumor microenvironment (TME) particularly during chemotherapy exposure. Tumor-associated macrophages (TAM) make up half of the immune population of the ovarian TME and are known to support CSCs and contribute to cancer progression. TAMs are plastic cells that alter their phenotype in response to environmental stimuli and thus may influence CSC maintenance during chemotherapy. Given the plasticity of TAMs, we studied the effects of carboplatin on macrophage phenotypes using both THP1- and peripheral blood mononuclear cell (PBMC)-derived macrophages and whether this supports CSCs and ovarian cancer progression following treatment. We found that carboplatin exposure induces an M1-like proinflammatory phenotype that promotes SOX2 expression, spheroid formation, and CD117+ ovarian CSCs, and that macrophage-secreted CCL2/MCP-1 is at least partially responsible for this effect. Depletion of TAMs during carboplatin exposure results in fewer CSCs and prolonged survival in a xenograft model of ovarian cancer. This study supports a role for platinum-based chemotherapies in promoting a transient proinflammatory M1-like TAM that enriches for CSCs during treatment. Improving our understanding of TME responses to cytotoxic drugs and identifying novel mechanisms of CSC maintenance will enable the development of better therapeutic strategies for high-grade serous ovarian cancer. Significance: We show that chemotherapy enhances proinflammatory macrophage phenotypes that correlate with ovarian cancer progression. Given that macrophages are the most prominent immune cell within these tumors, this work provides the foundation for future translational studies targeting specific macrophage populations during chemotherapy, a promising approach to prevent relapse in ovarian cancer.
Collapse
Affiliation(s)
- Luisjesus S. Cruz
- Department of Biology, San Diego State University, San Diego, California.
| | - Mikella Robinson
- Department of Biology, San Diego State University, San Diego, California.
| | - Denay Stevenson
- Department of Biology, San Diego State University, San Diego, California.
| | - Isabella C. Amador
- Department of Biology, San Diego State University, San Diego, California.
| | - Gregory J. Jordan
- Department of Biology, San Diego State University, San Diego, California.
| | - Sofia Valencia
- Department of Biology, San Diego State University, San Diego, California.
| | - Carolina Navarrete
- Department of Biology, San Diego State University, San Diego, California.
| | - Carrie D. House
- Department of Biology, San Diego State University, San Diego, California.
- Moores Cancer Center, University of California San Diego, San Diego, California.
| |
Collapse
|
11
|
Son B, Lee W, Kim H, Shin H, Park HH. Targeted therapy of cancer stem cells: inhibition of mTOR in pre-clinical and clinical research. Cell Death Dis 2024; 15:696. [PMID: 39349424 PMCID: PMC11442590 DOI: 10.1038/s41419-024-07077-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024]
Abstract
Cancer stem cells (CSCs) are a type of stem cell that possesses not only the intrinsic abilities of stem cells but also the properties of cancer cells. Therefore, CSCs are known to have self-renewal and outstanding proliferation capacity, along with the potential to differentiate into specific types of tumor cells. Cancers typically originate from CSCs, making them a significant target for tumor treatment. Among the related cascades of the CSCs, mammalian target of rapamycin (mTOR) pathway is regarded as one of the most important signaling pathways because of its association with significant upstream signaling: phosphatidylinositol 3‑kinase/protein kinase B (PI3K/AKT) pathway and mitogen‑activated protein kinase (MAPK) cascade, which influence various activities of stem cells, including CSCs. Recent studies have shown that the mTOR pathway not only affects generation of CSCs but also the maintenance of their pluripotency. Furthermore, the maintenance of pluripotency or differentiation into specific types of cancer cells depends on the regulation of the mTOR signal in CSCs. Consequently, the clinical potential and importance of mTOR in effective cancer therapy are increasing. In this review, we demonstrate the association between the mTOR pathway and cancer, including CSCs. Additionally, we discuss a new concept for anti-cancer drug development aimed at overcoming existing drawbacks, such as drug resistance, by targeting CSCs through mTOR inhibition.
Collapse
Affiliation(s)
- Boram Son
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
| | - Wonhwa Lee
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyeonjeong Kim
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
12
|
Sezginer O, Unver N. Dissection of pro-tumoral macrophage subtypes and immunosuppressive cells participating in M2 polarization. Inflamm Res 2024; 73:1411-1423. [PMID: 38935134 PMCID: PMC11349836 DOI: 10.1007/s00011-024-01907-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Alternatively activated macrophage (M2) polarization can result in one of four subtypes based on cytokines and signaling pathways associated with macrophage activation: M2a, M2b, M2c, and M2d macrophages. The majority of M2 subtypes are anti-inflammatory and pro-angiogenic, secreting growth factors (VEGF, PDGF) and matrix metalloproteinases (MMP2, MMP9) which boost tumor growth, metastasis, and invasion. M2-polarized macrophages are associated with immune suppressor cells harboring Myeloid derived suppressor cells, Regulatory T cells (Tregs), Regulatory B cells as well as alternatively activated (N2) neutrophils. Treg cells selectively support the metabolic stability, mitochondrial integrity, and survival rate of M2-like TAMs in an indirect environment. Also, the contribution of Breg cells influences macrophage polarization towards the M2 direction. TAM is activated when TAN levels in the tumor microenvironment are insufficient or vice versa, suggesting that macrophage and its polarization are fine-tuned. Understanding the functions of immune suppressive cells, mediators, and signaling pathways involved with M2 polarization will allow us to identify potential strategies for targeting the TAM repolarization phenotype for innovative immunotherapy approaches. In this review, we have highlighted the critical factors for M2 macrophage polarization, differential cytokine/chemokine profiles of M1 and M2 macrophage subtypes, and other immune cells' impact on the polarization within the immunosuppressive niche.
Collapse
Affiliation(s)
- Onurcan Sezginer
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye
| | - Nese Unver
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Sihhiye, Ankara, 06100, Türkiye.
| |
Collapse
|
13
|
Du Q, Dickinson A, Nakuleswaran P, Maghami S, Alagoda S, Hook AL, Ghaemmaghami AM. Targeting Macrophage Polarization for Reinstating Homeostasis following Tissue Damage. Int J Mol Sci 2024; 25:7278. [PMID: 39000385 PMCID: PMC11242417 DOI: 10.3390/ijms25137278] [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: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Tissue regeneration and remodeling involve many complex stages. Macrophages are critical in maintaining micro-environmental homeostasis by regulating inflammation and orchestrating wound healing. They display high plasticity in response to various stimuli, showing a spectrum of functional phenotypes that vary from M1 (pro-inflammatory) to M2 (anti-inflammatory) macrophages. While transient inflammation is an essential trigger for tissue healing following an injury, sustained inflammation (e.g., in foreign body response to implants, diabetes or inflammatory diseases) can hinder tissue healing and cause tissue damage. Modulating macrophage polarization has emerged as an effective strategy for enhancing immune-mediated tissue regeneration and promoting better integration of implantable materials in the host. This article provides an overview of macrophages' functional properties followed by discussing different strategies for modulating macrophage polarization. Advances in the use of synthetic and natural biomaterials to fabricate immune-modulatory materials are highlighted. This reveals that the development and clinical application of more effective immunomodulatory systems targeting macrophage polarization under pathological conditions will be driven by a detailed understanding of the factors that regulate macrophage polarization and biological function in order to optimize existing methods and generate novel strategies to control cell phenotype.
Collapse
Affiliation(s)
- Qiran Du
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Anna Dickinson
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Pruthvi Nakuleswaran
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK;
| | - Savindu Alagoda
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Andrew L. Hook
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Amir M. Ghaemmaghami
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| |
Collapse
|
14
|
Ulevicius J, Jasukaitiene A, Bartkeviciene A, Dambrauskas Z, Gulbinas A, Urboniene D, Paskauskas S. Preoperative Immune Cell Dysregulation Accompanies Ovarian Cancer Patients into the Postoperative Period. Int J Mol Sci 2024; 25:7087. [PMID: 39000195 PMCID: PMC11240929 DOI: 10.3390/ijms25137087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Ovarian cancer (OC) poses a significant global health challenge with high mortality rates, emphasizing the need for improved treatment strategies. The immune system's role in OC progression and treatment response is increasingly recognized, particularly regarding peripheral blood mononuclear cells (PBMCs) and cytokine production. This study aimed to investigate PBMC subpopulations (T and B lymphocytes, natural killer cells, monocytes) and cytokine production, specifically interleukin-1 beta (IL-1β), interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-10 (IL-10), interleukin-12 (IL-12), and tumor necrosis factor alpha (TNFα), in monocytes of OC patients both preoperatively and during the early postoperative period. Thirteen OC patients and 23 controls were enrolled. Preoperatively, OC patients exhibited changes in PBMC subpopulations, including decreased cytotoxic T cells, increased M2 monocytes, and the disbalance of monocyte cytokine production. These alterations persisted after surgery with subtle additional changes observed in PBMC subpopulations and cytokine expression in monocytes. Considering the pivotal role of these altered cells and cytokines in OC progression, our findings suggest that OC patients experience an enhanced pro-tumorigenic environment, which persists into the early postoperative period. These findings highlight the impact of surgery on the complex interaction between the immune system and OC progression. Further investigation is needed to clarify the underlying mechanisms during this early postoperative period, which may hold potential for interventions aimed at improving OC management.
Collapse
Affiliation(s)
- Jonas Ulevicius
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania; (A.J.); (A.B.); (Z.D.); (A.G.)
| | - Aldona Jasukaitiene
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania; (A.J.); (A.B.); (Z.D.); (A.G.)
| | - Arenida Bartkeviciene
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania; (A.J.); (A.B.); (Z.D.); (A.G.)
| | - Zilvinas Dambrauskas
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania; (A.J.); (A.B.); (Z.D.); (A.G.)
| | - Antanas Gulbinas
- Laboratory of Surgical Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania; (A.J.); (A.B.); (Z.D.); (A.G.)
| | - Daiva Urboniene
- Department of Laboratory Medicine, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania;
| | - Saulius Paskauskas
- Department of Obstetrics and Gynecology, Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
| |
Collapse
|
15
|
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: 0] [Impact Index Per Article: 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
|
16
|
Tcyganov EN, Kwak T, Yang X, Poli ANR, Hart C, Bhuniya A, Cassel J, Kossenkov A, Auslander N, Lu L, Sharma P, Mendoza MDGC, Zhigarev D, Cadungog MG, Jean S, Chatterjee-Paer S, Weiner D, Donthireddy L, Bristow B, Zhang R, Tyurin VA, Tyurina YY, Bayir H, Kagan VE, Salvino JM, Montaner LJ. Targeting LxCxE cleft pocket of retinoblastoma protein in M2 macrophages inhibits ovarian cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593562. [PMID: 38798466 PMCID: PMC11118332 DOI: 10.1101/2024.05.10.593562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Ovarian cancer remains a major health threat with limited treatment options available. It is characterized by immunosuppressive tumor microenvironment (TME) maintained by tumor- associated macrophages (TAMs) hindering anti-tumor responses and immunotherapy efficacy. Here we show that targeting retinoblastoma protein (Rb) by disruption of its LxCxE cleft pocket, causes cell death in TAMs by induction of ER stress, p53 and mitochondria-related cell death pathways. A reduction of pro-tumor Rb high M2-type macrophages from TME in vivo enhanced T cell infiltration and inhibited cancer progression. We demonstrate an increased Rb expression in TAMs in women with ovarian cancer is associated with poorer prognosis. Ex vivo, we show analogous cell death induction by therapeutic Rb targeting in TAMs in post-surgery ascites from ovarian cancer patients. Overall, our data elucidates therapeutic targeting of the Rb LxCxE cleft pocket as a novel promising approach for ovarian cancer treatment through depletion of TAMs and re-shaping TME immune landscape. Statement of significance Currently, targeting immunosuppressive myeloid cells in ovarian cancer microenvironment is the first priority need to enable successful immunotherapy, but no effective solutions are clinically available. We show that targeting LxCxE cleft pocket of Retinoblastoma protein unexpectedly induces preferential cell death in M2 tumor-associated macrophages. Depletion of immunosuppressive M2 tumor-associated macrophages reshapes tumor microenvironment, enhances anti-tumor T cell responses, and inhibits ovarian cancer. Thus, we identify a novel paradoxical function of Retinoblastoma protein in regulating macrophage viability as well as a promising target to enhance immunotherapy efficacy in ovarian cancer.
Collapse
|
17
|
Saad EE, Michel R, Borahay MA. Immunosuppressive tumor microenvironment and uterine fibroids: Role in collagen synthesis. Cytokine Growth Factor Rev 2024; 75:93-100. [PMID: 37839993 PMCID: PMC10922281 DOI: 10.1016/j.cytogfr.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
Uterine fibroids (UF), also called uterine leiomyoma, is one of the most prevalent uterine tumors. UF represents a serious women's health global problem with a significant physical, emotional, and socioeconomic impact. Risk factors for UF include racial disparities, age, race, hormonal factors, obesity, and lifestyle (diet, physical activity, and stress. There are several biological contributors to UF pathogenesis such as cellular proliferation, angiogenesis, and extracellular matrix (ECM) accumulation. This review addresses tumor immune microenvironment as a novel mediator of ECM deposition. Polarization of immune microenvironment towards the immunosuppressive phenotype has been associated with ECM deposition. Immunosuppressive cells include M2 macrophage, myeloid-derived suppressor cells (MDSCs), and Th17 cells, and their secretomes include interleukin 4 (IL-4), IL-10, IL-13, IL-17, IL-22, arginase 1, and transforming growth factor-beta (TGF-β1). The change in the immune microenvironment not only increase tumor growth but also aids in collagen synthesis and ECM disposition, which is one of the main hallmarks of UF pathogenesis. This review invites further investigations on the change in the UF immune microenvironment as well as a novel targeting approach instead of the traditional UF hormonal and supportive treatment.
Collapse
Affiliation(s)
- Eslam E Saad
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rachel Michel
- Department of Population, Family, and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, MD 21205, USA
| | - Mostafa A Borahay
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
18
|
Van Kleunen L, Ahmadian M, Post MD, Wolsky RJ, Rickert C, Jordan K, Hu J, Richer JK, Marjon NA, Behbakht K, Sikora MJ, Bitler BG, Clauset A. The spatial structure of the tumor immune microenvironment can explain and predict patient response in high-grade serous carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577350. [PMID: 38352574 PMCID: PMC10862769 DOI: 10.1101/2024.01.26.577350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
Despite ovarian cancer being the deadliest gynecological malignancy, there has been little change to therapeutic options and mortality rates over the last three decades. Recent studies indicate that the composition of the tumor immune microenvironment (TIME) influences patient outcomes but are limited by a lack of spatial understanding. We performed multiplexed ion beam imaging (MIBI) on 83 human high-grade serous carcinoma tumors - one of the largest protein-based, spatially-intact, single-cell resolution tumor datasets assembled - and used statistical and machine learning approaches to connect features of the TIME spatial organization to patient outcomes. Along with traditional clinical/immunohistochemical attributes and indicators of TIME composition, we found that several features of TIME spatial organization had significant univariate correlations and/or high relative importance in high-dimensional predictive models. The top performing predictive model for patient progression-free survival (PFS) used a combination of TIME composition and spatial features. Results demonstrate the importance of spatial structure in understanding how the TIME contributes to treatment outcomes. Furthermore, the present study provides a generalizable roadmap for spatial analyses of the TIME in ovarian cancer research.
Collapse
Affiliation(s)
- Lucy Van Kleunen
- Department of Computer Science, University of Colorado, Boulder, USA
| | - Mansooreh Ahmadian
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Miriam D Post
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Rebecca J Wolsky
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Christian Rickert
- Department of Immunology and Microbiology, The University of Colorado Anschutz Medical Campus
| | - Kimberly Jordan
- Department of Immunology and Microbiology, The University of Colorado Anschutz Medical Campus
| | - Junxiao Hu
- Department of Pediatrics, Cancer Center Biostatistics Core, University of Colorado Anschutz Medical Campus, CO, USA
| | - Jennifer K. Richer
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Nicole A. Marjon
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Kian Behbakht
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Matthew J. Sikora
- Department of Pathology, The University of Colorado Anschutz Medical Campus
| | - Benjamin G. Bitler
- Department of OB/GYN, The University of Colorado Anschutz Medical Campus
| | - Aaron Clauset
- Department of Computer Science, University of Colorado, Boulder, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
19
|
Zheng C, Li H, Zhao X, Yang S, Zhan J, Liu H, Jiang Y, shi L, Song Y, Lei Y, Yu T, Wang X, Li H, Wang X, Xu Y, Yao Z. Expression of PD-1 mitigates phagocytic activities TAM in osteosarcoma. Heliyon 2024; 10:e23498. [PMID: 38223729 PMCID: PMC10784140 DOI: 10.1016/j.heliyon.2023.e23498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/16/2023] [Accepted: 12/05/2023] [Indexed: 01/16/2024] Open
Abstract
The high expression of programmed death 1 (PD-1) is a hallmark of T cell exhaustion, consequently inhibiting the anti-tumor immunity, tumor-associated macrophages (TAMs) aggravate Osteosarcoma (OS) progression. However, PD-1 expression on TAMs in OS metastasis remains unclear. Here, we used scRNA-Seq of 15500 individual cells from human OS lung metastatic lesion, identified thirteen major cell clusters. Our data revealed that tumor-infiltrating lymphocytes (TILs) OS lung metastatic accompanied by accumulation of exhausted T cells and regulatory T cells (Tregs). CD3+ T cells from human OS lung metastatic exhibited lower proliferation than in primary tissue. Importantly, TAMs mainly comprise immunosuppressive M2 phenotype in OS metastasis. Mechanistically, we found that PD-1 of TAMs inhibits the phagocytic potency, further promoting the progression of OS metastasis. Therefore, the study provides a strong technical support for OS immunotherapy based on PD-1 inhibitors.
Collapse
Affiliation(s)
- Chenhong Zheng
- Department of Ultrasound, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, Yunnan, China
| | - Heng Li
- Department of Thoracic Surgery I&II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, 650118, China
| | - Xiaohui Zhao
- Department of Ultrasound, Hohhot First Hospital, Hohhot City, 010059, Inner Mongolia Autonomous Region, China Ultrasonic Department, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Siyu Yang
- Department of Ultrasound, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, Yunnan, China
| | - Jinqin Zhan
- Ultrasonic Department, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Huaie Liu
- Department of Geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Yan Jiang
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Li shi
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Yaxian Song
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Yujie Lei
- Department of Thoracic Surgery I&II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, 650118, China
| | - Tingdong Yu
- Department of Thoracic Surgery I&II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, 650118, China
| | - Xiaoxiong Wang
- Molecular Diagnostic Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Hongsheng Li
- Molecular Diagnostic Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Xi Wang
- Department of Thoracic Surgery I&II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, 650118, China
| | - Yushan Xu
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Zhihong Yao
- Bone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, 650118, China
- Department of Cancer Center Office, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| |
Collapse
|
20
|
Bui I, Bonavida B. Polarization of M2 Tumor-Associated Macrophages (TAMs) in Cancer Immunotherapy. Crit Rev Oncog 2024; 29:75-95. [PMID: 38989739 DOI: 10.1615/critrevoncog.2024053830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
We have witnessed in the last decade new milestones in the treatment of various resistant cancers with new immunotherapeutic modalities. These advances have resulted in significant objective durable clinical responses in a subset of cancer patients. These findings strongly suggested that immunotherapy should be considered for the treatment of all subsets of cancer patients. Accordingly, the mechanisms underlying resistance to immunotherapy must be explored and develop new means to target these resistant factors. One of the pivotal resistance mechanisms in the tumor microenvironment (TME) is the high infiltration of tumor-associated macrophages (TAMs) that are highly immunosuppressive and responsible, in large part, of cancer immune evasion. Thus, various approaches have been investigated to target the TAMs to restore the anti-tumor immune response. One approach is to polarize the M2 TAMS to the M1 phenotype that participates in the activation of the anti-tumor response. In this review, we discuss the various and differential properties of the M1 and M2 phenotypes, the molecular signaling pathways that participate in the polarization, and various approaches used to target the polarization of the M2 TAMs into the M1 anti-tumor phenotype. These approaches include inhibitors of histone deacetylases, PI3K inhibitors, STAT3 inhibitors, TLR agonists, and metabolic reprogramming. Clearly, due to the distinct features of various cancers and their heterogeneities, a single approach outlined above might only be effective against some cancers and not others. In addition, targeting by itself may not be efficacious unless used in combination with other therapeutic modalities.
Collapse
Affiliation(s)
- Indy Bui
- University of California Los Angeles
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
21
|
Blanc-Durand F, Clemence Wei Xian L, Tan DSP. Targeting the immune microenvironment for ovarian cancer therapy. Front Immunol 2023; 14:1328651. [PMID: 38164130 PMCID: PMC10757966 DOI: 10.3389/fimmu.2023.1328651] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
Ovarian cancer (OC) is an aggressive malignancy characterized by a complex immunosuppressive tumor microenvironment (TME). Immune checkpoint inhibitors have emerged as a breakthrough in cancer therapy by reactivating the antitumor immune response suppressed by tumor cells. However, in the case of OC, these inhibitors have failed to demonstrate significant improvements in patient outcomes, and existing biomarkers have not yet identified promising subgroups. Consequently, there remains a pressing need to understand the interplay between OC tumor cells and their surrounding microenvironment to develop effective immunotherapeutic approaches. This review aims to provide an overview of the OC TME and explore its potential as a therapeutic strategy. Tumor-infiltrating lymphocytes (TILs) are major actors in OC TME. Evidence has been accumulating regarding the spontaneous TILS response against OC antigens. Activated T-helpers secrete a wide range of inflammatory cytokines with a supportive action on cytotoxic T-cells. Simultaneously, mature B-cells are recruited and play a significant antitumor role through opsonization of target antigens and T-cell recruitment. Macrophages also form an important subset of innate immunity (M1-macrophages) while participating in the immune-stimulation context. Finally, OC has shown to engage a significant natural-killer-cells immune response, exerting direct cytotoxicity without prior sensitization. Despite this initial cytotoxicity, OC cells develop various strategies to induce an immune-tolerant state. To this end, multiple immunosuppressive molecules are secreted to impair cytotoxic cells, recruit regulatory cells, alter antigen presentation, and effectively evade immune response. Consequently, OC TME is predominantly infiltrated by immunosuppressive cells such as FOXP3+ regulatory T-cells, M2-polarized macrophages and myeloid-derived suppressor cells. Despite this strong immunosuppressive state, PD-1/PD-L1 inhibitors have failed to improve outcomes. Beyond PD-1/PD-L1, OC expresses multiple other immune checkpoints that contribute to immune evasion, and each representing potential immune targets. Novel immunotherapies are attempting to overcome the immunosuppressive state and induce specific immune responses using antibodies adoptive cell therapy or vaccines. Overall, the OC TME presents both opportunities and obstacles. Immunotherapeutic approaches continue to show promise, and next-generation inhibitors offer exciting opportunities. However, tailoring therapies to individual immune characteristics will be critical for the success of these treatments.
Collapse
Affiliation(s)
- Felix Blanc-Durand
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - Lai Clemence Wei Xian
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - David S. P. Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University Centre for Cancer Research (N2CR) and Cancer Science Institute (CSI), National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Lv Y, Liao Z, Bi Q, Xie C, Wei X, Yun Y, He Y, Tang Q. Irreversible repolarization of tumour-associated macrophages by low-Pi stress inhibits the progression of hepatocellular carcinoma. J Cell Mol Med 2023; 27:2906-2921. [PMID: 37471521 PMCID: PMC10538272 DOI: 10.1111/jcmm.17861] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023] Open
Abstract
Numerous studies have shown the positive correlation between high levels of Pi and tumour progression. A critical goal of macrophage-based cancer therapeutics is to reduce anti-inflammatory macrophages (M2) and increase proinflammatory antitumour macrophages (M1). This study aimed to investigate the relationship between macrophage polarization and low-Pi stress. First, the spatial populations of M2 and M1 macrophages in 22 HCC patient specimens were quantified and correlated with the local Pi concentration. The levels of M2 and M1 macrophage markers expressed in the peritumour area were higher than the intratumour levels, and the expression of M2 markers was positively correlated with Pi concentration. Next, monocytes differentiated from THP-1 cells were polarized against different Pi concentrations to investigate the activation or silencing of the expression of p65, IκB-α and STAT3 as well as their phosphorylation. Results showed that low-Pi stress irreversibly repolarizes tumour-associated macrophages (TAMs) towards the M1 phenotype by silencing stat6 and activating p65. Moreover, HepG-2 and SMCC-7721 cells were cultured in conditioned medium to investigate the innate anticancer immune effects on tumour progression. Both cancer cell lines showed reduced proliferation, migration and invasion, as epithelial-mesenchymal transition (EMT) was inactivated. In vivo therapeutic effect on the innate and adaptive immune processes was validated in a subcutaneous liver cancer model by the intratumoural injection of sevelamer. Tumour growth was significantly inhibited by the partial deprivation of intratumoural Pi as the tumour microenvironment under low-Pi stress is more immunostimulatory. The anticancer immune response, activated by low-Pi stress, suggests a new macrophage-based immunotherapeutic modality.
Collapse
Affiliation(s)
- Yang‐feng Lv
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public HealthNanchang UniversityNanchangChina
- Institute for Advanced Study, Nanchang UniversityNanchangChina
| | - Zi‐qiang Liao
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public HealthNanchang UniversityNanchangChina
- Institute for Advanced Study, Nanchang UniversityNanchangChina
| | - Qiu‐chen Bi
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public HealthNanchang UniversityNanchangChina
- Institute for Advanced Study, Nanchang UniversityNanchangChina
| | - Chuan‐sheng Xie
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public HealthNanchang UniversityNanchangChina
| | - Xiao‐yong Wei
- Department of Hepatobiliary SurgeryJiangxi Provincial Cancer HospitalNanchangChina
| | - Yi Yun
- Biobank CenterThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Yuan‐qiao He
- Center of Laboratory Animal Science, Jiangxi Province Key Laboratory of Laboratory AnimalNanchang UniversityNanchangChina
| | - Qun Tang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public HealthNanchang UniversityNanchangChina
- Institute for Advanced Study, Nanchang UniversityNanchangChina
| |
Collapse
|
23
|
Hung CN, Chen M, DeArmond DT, Chiu CHL, Limboy CA, Tan X, Kusi M, Chou CW, Lin LL, Zhang Z, Wang CM, Chen CL, Mitsuya K, Osmulski PA, Gaczynska ME, Kirma NB, Vadlamudi RK, Gibbons DL, Warner S, Brenner AJ, Mahadevan D, Michalek JE, Huang THM, Taverna JA. AXL-initiated paracrine activation of pSTAT3 enhances mesenchymal and vasculogenic supportive features of tumor-associated macrophages. Cell Rep 2023; 42:113067. [PMID: 37659081 PMCID: PMC10577802 DOI: 10.1016/j.celrep.2023.113067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 07/14/2023] [Accepted: 08/18/2023] [Indexed: 09/04/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are integral to the development of complex tumor microenvironments (TMEs) and can execute disparate cellular programs in response to extracellular cues. However, upstream signaling processes underpinning this phenotypic plasticity remain to be elucidated. Here, we report that concordant AXL-STAT3 signaling in TAMs is triggered by lung cancer cells or cancer-associated fibroblasts in the cytokine milieu. This paracrine action drives TAM differentiation toward a tumor-promoting "M2-like" phenotype with upregulation of CD163 and putative mesenchymal markers, contributing to TAM heterogeneity and diverse cellular functions. One of the upregulated markers, CD44, mediated by AXL-IL-11-pSTAT3 signaling cascade, enhances macrophage ability to interact with endothelial cells and facilitate formation of primitive vascular networks. We also found that AXL-STAT3 inhibition can impede the recruitment of TAMs in a xenograft mouse model, thereby suppressing tumor growth. These findings suggest the potential application of AXL-STAT3-related markers to quantitatively assess metastatic potential and inform therapeutic strategies in lung cancer.
Collapse
Affiliation(s)
- Chia-Nung Hung
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Meizhen Chen
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Daniel T DeArmond
- Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX, USA
| | - Cheryl H-L Chiu
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Catherine A Limboy
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Xi Tan
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Meena Kusi
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Chih-Wei Chou
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Li-Ling Lin
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Zhao Zhang
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Chiou-Miin Wang
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Chun-Liang Chen
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA; Office of Nursing Research & Scholarship, School of Nursing, University of Texas Health Science Center, San Antonio, TX, USA
| | - Kohzoh Mitsuya
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Pawel A Osmulski
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Maria E Gaczynska
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Nameer B Kirma
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Ratna K Vadlamudi
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Don L Gibbons
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Andrew J Brenner
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Division of Hematology and Oncology, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Daruka Mahadevan
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Division of Hematology and Oncology, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Joel E Michalek
- Department of Population Health Sciences, University of Texas Health Science Center, San Antonio, TX, USA
| | - Tim H-M Huang
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA; Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA.
| | - Josephine A Taverna
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA; Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Division of Hematology and Oncology, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
24
|
Mhaidly N, Journe F, Najem A, Stock L, Trelcat A, Dequanter D, Saussez S, Descamps G. Macrophage Profiling in Head and Neck Cancer to Improve Patient Prognosis and Assessment of Cancer Cell-Macrophage Interactions Using Three-Dimensional Coculture Models. Int J Mol Sci 2023; 24:12813. [PMID: 37628994 PMCID: PMC10454490 DOI: 10.3390/ijms241612813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Tumor-associated macrophages are key components of the tumor microenvironment and play important roles in the progression of head and neck cancer, leading to the development of effective strategies targeting immune cells in tumors. Our study demonstrated the prognostic potential of a new scoring system (Macroscore) based on the combination of the ratio and the sum of the high and low densities of M1 (CD80+) and M2 (CD163+) macrophages in a series of head and neck cancer patients, including a training population (n = 54) and a validation population (n = 19). Interestingly, the Macroscore outperformed TNM criteria and p16 status, showing a significant association with poor patient prognosis, and demonstrated significant predictive value for overall survival. Additionally, 3D coculture spheroids were established to analyze the crosstalk between cancer cells and monocytes/macrophages. Our data revealed that cancer cells can induce monocyte differentiation into protumoral M2 macrophages, creating an immunosuppressive microenvironment. This coculture also induced the production of immunosuppressive cytokines, such as IL10 and IL8, known to promote M2 polarization. Finally, we validated the ability of the macrophage subpopulations to induce apoptosis (M1) or support proliferation (M2) of cancer cells. Overall, our research highlights the potential of the Macroscore as a valuable prognostic biomarker to enhance the clinical management of patients and underscores the relevance of a spheroid model in gaining a better understanding of the mechanisms underlying cancer cell-macrophage interactions.
Collapse
Affiliation(s)
- Nour Mhaidly
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (F.J.); (L.S.); (A.T.); (S.S.)
| | - Fabrice Journe
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (F.J.); (L.S.); (A.T.); (S.S.)
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium;
| | - Ahmad Najem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium;
| | - Louis Stock
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (F.J.); (L.S.); (A.T.); (S.S.)
| | - Anne Trelcat
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (F.J.); (L.S.); (A.T.); (S.S.)
| | - Didier Dequanter
- Department of Otolaryngology and Head and Neck Surgery, CHU Saint-Pierre, 1000 Brussels, Belgium;
| | - Sven Saussez
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (F.J.); (L.S.); (A.T.); (S.S.)
- Department of Otolaryngology and Head and Neck Surgery, CHU Saint-Pierre, 1000 Brussels, Belgium;
| | - Géraldine Descamps
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (F.J.); (L.S.); (A.T.); (S.S.)
| |
Collapse
|
25
|
Xi Y, Zhang Y, Zheng K, Zou J, Gui L, Zou X, Chen L, Hao J, Zhang Y. A chemotherapy response prediction model derived from tumor-promoting B and Tregs and proinflammatory macrophages in HGSOC. Front Oncol 2023; 13:1171582. [PMID: 37519793 PMCID: PMC10382026 DOI: 10.3389/fonc.2023.1171582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Background Most patients with high-grade serous ovarian cancer (HGSOC) experienced disease recurrence with cumulative chemoresistance, leading to treatment failure. However, few biomarkers are currently available in clinical practice that can accurately predict chemotherapy response. The tumor immune microenvironment is critical for cancer development, and its transcriptomic profile may be associated with treatment response and differential outcomes. The aim of this study was to develop a new predictive signature for chemotherapy in patients with HGSOC. Methods Two HGSOC single-cell RNA sequencing datasets from patients receiving chemotherapy were reinvestigated. The subtypes of endoplasmic reticulum stress-related XBP1+ B cells, invasive metastasis-related ACTB+ Tregs, and proinflammatory-related macrophage subtypes with good predictive power and associated with chemotherapy response were identified. These results were verified in an independent HGSOC bulk RNA-seq dataset for chemotherapy. Further validation in clinical cohorts used quantitative real-time PCR (qRT-PCR). Results By combining cluster-specific genes for the aforementioned cell subtypes, we constructed a chemotherapy response prediction model containing 43 signature genes that achieved an area under the receiver operator curve (AUC) of 0.97 (p = 2.1e-07) for the GSE156699 cohort (88 samples). A huge improvement was achieved compared to existing prediction models with a maximum AUC of 0.74. In addition, its predictive capability was validated in multiple independent bulk RNA-seq datasets. The qRT-PCR results demonstrate that the expression of the six genes has the highest diagnostic value, consistent with the trend observed in the analysis of public data. Conclusions The developed chemotherapy response prediction model can be used as a valuable clinical decision tool to guide chemotherapy in HGSOC patients.
Collapse
Affiliation(s)
- Yue Xi
- Department of Reproductive Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yingchun Zhang
- Department of Reproductive Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Kun Zheng
- Department of Urology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiawei Zou
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lv Gui
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xin Zou
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Liang Chen
- Department of Gynecological Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Hao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiming Zhang
- Department of Reproductive Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
26
|
De Simone G, Soldani C, Morabito A, Franceschini B, Ferlan F, Costa G, Pastorelli R, Donadon M, Brunelli L. Implication of metabolism in the polarization of tumor-associated-macrophages: the mass spectrometry-based point of view. Front Immunol 2023; 14:1193235. [PMID: 37503340 PMCID: PMC10368868 DOI: 10.3389/fimmu.2023.1193235] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/22/2023] [Indexed: 07/29/2023] Open
Abstract
Tumor-associated macrophages (TAMs) represent one of the main tumor-infiltrating immune cell types and are generally categorized into either of two functionally contrasting subtypes, namely classical activated M1 macrophages and alternatively activated M2 macrophages. TAMs showed different activation states that can be represent by the two extremes of the complex profile of macrophages biology, the M1-like phenotype (pro-inflammatory activity) and the M2-like phenotype (anti-inflammatory activity). Based on the tumor type, and grades, TAMs can acquire different functions and properties; usually, the M1-like phenotype is typical of early tumor stages and is associated to an anti-tumor activity, while M2-like phenotype has a pro-inflammatory activity and is related to a poor patients' prognosis. The classification of macrophages into M1/M2 groups based on well-defined stimuli does not model the infinitely more complex tissue milieu where macrophages (potentially of different origin) would be exposed to multiple signals in different sequential order. This review aims to summarize the recent mass spectrometry-based (MS-based) metabolomics findings about the modifications of metabolism in TAMs polarization in different tumors. The published data shows that MS-based metabolomics is a promising tool to help better understanding TAMs metabolic phenotypes, although it is still poorly applied for TAMs metabolism. The knowledge of key metabolic alterations in TAMs is an essential step for discovering TAMs polarization novel biomarkers and developing novel therapeutic approaches targeting TAM metabolism to repolarize TAMs towards their anti-tumor phenotype.
Collapse
Affiliation(s)
- Giulia De Simone
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biotechnologies and Biosciences, Università degli Studi Milano Bicocca, Milan, Italy
| | - Cristiana Soldani
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Aurelia Morabito
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Barbara Franceschini
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Fabrizio Ferlan
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Guido Costa
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Roberta Pastorelli
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Matteo Donadon
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
- Department of General Surgery, University Maggiore Hospital, Novara, Italy
| | - Laura Brunelli
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
27
|
Liu Y, Yang J, Hilliard TS, Wang Z, Johnson J, Wang W, Harper EI, Ott C, O'Brien C, Campbell L, Crowley B, Grisoli S, Stavrou NM, Juncker-Jensen A, Stack MS. Host obesity alters the ovarian tumor immune microenvironment and impacts response to standard of care chemotherapy. J Exp Clin Cancer Res 2023; 42:165. [PMID: 37438818 PMCID: PMC10337170 DOI: 10.1186/s13046-023-02740-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND The majority of women with epithelial ovarian cancer (OvCa) are diagnosed with metastatic disease, resulting in a poor 5-year survival of 31%. Obesity is a recognized non-infectious pandemic that increases OvCa incidence, enhances metastatic success and reduces survival. We have previously demonstrated a link between obesity and OvCa metastatic success in a diet-induced obesity mouse model wherein a significantly enhanced tumor burden was associated with a decreased M1/M2 tumor-associated macrophage ratio (Liu Y et al. Can, Res. 2015; 75:5046-57). METHODS The objective of this study was to use pre-clinical murine models of diet-induced obesity to evaluate the effect of a high fat diet (HFD) on response to standard of care chemotherapy and to assess obesity-associated changes in the tumor microenvironment. Archived tumor tissues from ovarian cancer patients of defined body mass index (BMI) were also evaluated using multiplexed immunofluorescence analysis of immune markers. RESULTS We observed a significantly diminished response to standard of care paclitaxel/carboplatin chemotherapy in HFD mice relative to low fat diet (LFD) controls. A corresponding decrease in the M1/M2 macrophage ratio and enhanced tumor fibrosis were observed both in murine DIO studies and in human tumors from women with BMI > 30. CONCLUSIONS Our data suggest that the reported negative impact of obesity on OvCa patient survival may be due in part to the effect of the altered M1/M2 tumor-associated macrophage ratio and enhanced fibrosis on chemosensitivity. These data demonstrate a contribution of host obesity to ovarian tumor progression and therapeutic response and support future combination strategies targeting macrophage polarization and/or fibrosis in the obese host.
Collapse
Affiliation(s)
- Yueying Liu
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Jing Yang
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Tyvette S Hilliard
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Zhikun Wang
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Jeff Johnson
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Wanrui Wang
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Elizabeth I Harper
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Connor Ott
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Caitlin O'Brien
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Leigh Campbell
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Brian Crowley
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | - Stephen Grisoli
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA
| | | | | | - M Sharon Stack
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA.
- Harper Cancer Research Institute, University of Notre Dame, A200E Harper Hall, 1234 N. Notre Dame Ave, South Bend, IN, 46617, USA.
| |
Collapse
|
28
|
Wilczyński JR, Wilczyński M, Paradowska E. "DEPHENCE" system-a novel regimen of therapy that is urgently needed in the high-grade serous ovarian cancer-a focus on anti-cancer stem cell and anti-tumor microenvironment targeted therapies. Front Oncol 2023; 13:1201497. [PMID: 37448521 PMCID: PMC10338102 DOI: 10.3389/fonc.2023.1201497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer, especially high-grade serous type, is the most lethal gynecological malignancy. The lack of screening programs and the scarcity of symptomatology result in the late diagnosis in about 75% of affected women. Despite very demanding and aggressive surgical treatment, multiple-line chemotherapy regimens and both approved and clinically tested targeted therapies, the overall survival of patients is still unsatisfactory and disappointing. Research studies have recently brought some more understanding of the molecular diversity of the ovarian cancer, its unique intraperitoneal biology, the role of cancer stem cells, and the complexity of tumor microenvironment. There is a growing body of evidence that individualization of the treatment adjusted to the molecular and biochemical signature of the tumor as well as to the medical status of the patient should replace or supplement the foregoing therapy. In this review, we have proposed the principles of the novel regimen of the therapy that we called the "DEPHENCE" system, and we have extensively discussed the results of the studies focused on the ovarian cancer stem cells, other components of cancer metastatic niche, and, finally, clinical trials targeting these two environments. Through this, we have tried to present the evolving landscape of treatment options and put flesh on the experimental approach to attack the high-grade serous ovarian cancer multidirectionally, corresponding to the "DEPHENCE" system postulates.
Collapse
Affiliation(s)
- Jacek R Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, Lodz, Poland
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother's Health Center-Research Institute, Lodz, Poland
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland
| |
Collapse
|
29
|
Dossou AS, Mantsch ME, Kapic A, Burnett WL, Sabnis N, Coffer JL, Berg RE, Fudala R, Lacko AG. Mannose-Coated Reconstituted Lipoprotein Nanoparticles for the Targeting of Tumor-Associated Macrophages: Optimization, Characterization, and In Vitro Evaluation of Effectiveness. Pharmaceutics 2023; 15:1685. [PMID: 37376134 DOI: 10.3390/pharmaceutics15061685] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Reconstituted high-density lipoprotein nanoparticles (rHDL NPs) have been utilized as delivery vehicles to a variety of targets, including cancer cells. However, the modification of rHDL NPs for the targeting of the pro-tumoral tumor-associated macrophages (TAMs) remains largely unexplored. The presence of mannose on nanoparticles can facilitate the targeting of TAMs which highly express the mannose receptor at their surface. Here, we optimized and characterized mannose-coated rHDL NPs loaded with 5,6-dimethylxanthenone-4-acetic acid (DMXAA), an immunomodulatory drug. Lipids, recombinant apolipoprotein A-I, DMXAA, and different amounts of DSPE-PEG-mannose (DPM) were combined to assemble rHDL-DPM-DMXAA NPs. The introduction of DPM in the nanoparticle assembly altered the particle size, zeta potential, elution pattern, and DMXAA entrapment efficiency of the rHDL NPs. Collectively, the changes in physicochemical characteristics of rHDL NPs upon the addition of the mannose moiety DPM indicated that the rHDL-DPM-DMXAA NPs were successfully assembled. The rHDL-DPM-DMXAA NPs induced an immunostimulatory phenotype in macrophages pre-exposed to cancer cell-conditioned media. Furthermore, rHDL-DPM NPs delivered their payload more readily to macrophages than cancer cells. Considering the effects of the rHDL-DPM-DMXAA NPs on macrophages, the rHDL-DPM NPs have the potential to serve as a drug delivery platform for the selective targeting of TAMs.
Collapse
Affiliation(s)
- Akpedje S Dossou
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center (UNTHSC), Fort Worth, TX 76107, USA
| | - Morgan E Mantsch
- College of Natural Sciences, University of Texas at Austin, Austin, TX 78705, USA
| | - Ammar Kapic
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center (UNTHSC), Fort Worth, TX 76107, USA
| | - William L Burnett
- College of Science and Engineering, Texas Christian University (TCU), Fort Worth, TX 76129, USA
| | - Nirupama Sabnis
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center (UNTHSC), Fort Worth, TX 76107, USA
| | - Jeffery L Coffer
- College of Science and Engineering, Texas Christian University (TCU), Fort Worth, TX 76129, USA
| | - Rance E Berg
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center (UNTHSC), Fort Worth, TX 76107, USA
| | - Rafal Fudala
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center (UNTHSC), Fort Worth, TX 76107, USA
| | - Andras G Lacko
- Department of Microbiology, Immunology and Genetics, UNT Health Science Center (UNTHSC), Fort Worth, TX 76107, USA
| |
Collapse
|
30
|
Pankowska KA, Będkowska GE, Chociej-Stypułkowska J, Rusak M, Dąbrowska M, Osada J. Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance. Int J Mol Sci 2023; 24:ijms24119279. [PMID: 37298230 DOI: 10.3390/ijms24119279] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological cancers, largely due to the fast development of metastasis and drug resistance. The immune system is a critical component of the OC tumor microenvironment (TME) and immune cells such as T cells, NK cells, and dendritic cells (DC) play a key role in anti-tumor immunity. However, OC tumor cells are well known for evading immune surveillance by modulating the immune response through various mechanisms. Recruiting immune-suppressive cells such as regulatory T cells (Treg cells), macrophages, or myeloid-derived suppressor cells (MDSC) inhibit the anti-tumor immune response and promote the development and progression of OC. Platelets are also involved in immune evasion by interaction with tumor cells or through the secretion of a variety of growth factors and cytokines to promote tumor growth and angiogenesis. In this review, we discuss the role and contribution of immune cells and platelets in TME. Furthermore, we discuss their potential prognostic significance to help in the early detection of OC and to predict disease outcome.
Collapse
Affiliation(s)
- Katarzyna Aneta Pankowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Grażyna Ewa Będkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Chociej-Stypułkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Małgorzata Rusak
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Milena Dąbrowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Osada
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| |
Collapse
|
31
|
Valerio TI, Furrer CL, Sadeghipour N, Patrock SJX, Tillery SA, Hoover AR, Liu K, Chen WR. Immune modulations of the tumor microenvironment in response to phototherapy. JOURNAL OF INNOVATIVE OPTICAL HEALTH SCIENCES 2023; 16:2330007. [PMID: 38550850 PMCID: PMC10976517 DOI: 10.1142/s1793545823300070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/01/2024]
Abstract
The tumor microenvironment (TME) promotes pro-tumor and anti-inflammatory metabolisms and suppresses the host immune system. It prevents immune cells from fighting against cancer effectively, resulting in limited efficacy of many current cancer treatment modalities. Different therapies aim to overcome the immunosuppressive TME by combining various approaches to synergize their effects for enhanced anti-tumor activity and augmented stimulation of the immune system. Immunotherapy has become a major therapeutic strategy because it unleashes the power of the immune system by activating, enhancing, and directing immune responses to prevent, control, and eliminate cancer. Phototherapy uses light irradiation to induce tumor cell death through photothermal, photochemical, and photo-immunological interactions. Phototherapy induces tumor immunogenic cell death, which is a precursor and enhancer for anti-tumor immunity. However, phototherapy alone has limited effects on long-term and systemic anti-tumor immune responses. Phototherapy can be combined with immunotherapy to improve the tumoricidal effect by killing target tumor cells, enhancing immune cell infiltration in tumors, and rewiring pathways in the TME from anti-inflammatory to pro-inflammatory. Phototherapy-enhanced immunotherapy triggers effective cooperation between innate and adaptive immunities, specifically targeting the tumor cells, whether they are localized or distant. Herein, the successes and limitations of phototherapy combined with other cancer treatment modalities will be discussed. Specifically, we will review the synergistic effects of phototherapy combined with different cancer therapies on tumor elimination and remodeling of the immunosuppressive TME. Overall, phototherapy, in combination with other therapeutic modalities, can establish anti-tumor pro-inflammatory phenotypes in activated tumor-infiltrating T cells and B cells and activate systemic anti-tumor immune responses.
Collapse
Affiliation(s)
- Trisha I. Valerio
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Coline L. Furrer
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Negar Sadeghipour
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
- School of Electrical and Computer Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Sophia-Joy X. Patrock
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Sayre A. Tillery
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Ashley R. Hoover
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Wei R. Chen
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| |
Collapse
|
32
|
Deng Y, Fu Y, Chua SL, Khoo BL. Biofilm Potentiates Cancer-Promoting Effects of Tumor-Associated Macrophages in a 3D Multi-Faceted Tumor Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205904. [PMID: 36748304 DOI: 10.1002/smll.202205904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/01/2023] [Indexed: 05/11/2023]
Abstract
Components of the tumor microenvironment (TME), such as tumor-associated macrophages (TAMs), influence tumor progression. The specific polarization and phenotypic transition of TAMs in the tumor microenvironment lead to two-pronged impacts that can promote or hinder cancer development and treatment. Here, a novel microfluidic multi-faceted bladder tumor model (TAMPIEB ) is developed incorporating TAMs and cancer cells to evaluate the impact of bacterial distribution on immunomodulation within the tumor microenvironment in vivo. It is demonstrated for the first time that biofilm-induced inflammatory conditions within tumors promote the transition of macrophages from a pro-inflammatory M1-like to an anti-inflammatory/pro-tumor M2-like state. Consequently, multiple roles and mechanisms by which biofilms promote cancer by inducing pro-tumor phenotypic switch of TAMs are identified, including cancer hallmarks such as reducing susceptibility to apoptosis, enhancing cell viability, and promoting epithelial-mesenchymal transition and metastasis. Furthermore, biofilms formed by extratumoral bacteria can shield tumors from immune attack by TAMs, which can be visualized through various imaging assays in situ. The study sheds light on the underlying mechanism of biofilm-mediated inflammation on tumor progression and provides new insights into combined anti-biofilm therapy and immunotherapy strategies in clinical trials.
Collapse
Affiliation(s)
- Yanlin Deng
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, 999077, Hong Kong
| | - Yatian Fu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, 999077, Hong Kong
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Kowloon, 999077, Hong Kong
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, Kowloon, 999077, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong SAR, Kowloon, 999077, China
- Shenzhen Key Laboratory of Food Biological Safety Control, Kowloon, 999077, Hong Kong
- Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Hong Kong SAR, Kowloon, 999077, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, 999077, Hong Kong
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Kowloon, 999077, Hong Kong
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen-Futian Research Institute, Shenzhen, 518057, China
| |
Collapse
|
33
|
Liu J, Zhang X, Wang H, Zuo X, Hong L. Comprehensive Analysis of Purine-Metabolism-Related Gene Signature for Predicting Ovarian Cancer Prognosis, Immune Landscape, and Potential Treatment Options. J Pers Med 2023; 13:jpm13050776. [PMID: 37240946 DOI: 10.3390/jpm13050776] [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: 03/02/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Purine metabolism is an important branch of metabolic reprogramming and has received increasing attention in cancer research. Ovarian cancer is an extremely dangerous gynecologic malignancy for which there are no adequate tools to predict prognostic risk. Here, we identified a prognostic signature consisting of nine genes related to purine metabolism, including ACSM1, CACNA1C, EPHA4, TPM3, PDIA4, JUNB, EXOSC4, TRPM2, and CXCL9. The risk groups defined by the signature are able to distinguish the prognostic risk and the immune landscape of patients. In particular, the risk scores offer promising personalized drug options. By combining risk scores with clinical characteristics, we have created a more detailed composite nomogram that allows for a more complete and individualized prediction of prognosis. In addition, we demonstrated metabolic differences between platinum-resistant and platinum-sensitive ovarian cancer cells. In summary, we have performed the first comprehensive analysis of genes related to purine metabolism in ovarian cancer patients and created a feasible prognostic signature that will aid in risk prediction and support personalized medicine.
Collapse
Affiliation(s)
- Jingchun Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaoyi Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Haoyu Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaohu Zuo
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
34
|
Nie S, Ni N, Chen N, Gong M, Feng E, Liu J, Liu Q. Development of a necroptosis-related gene signature and the immune landscape in ovarian cancer. J Ovarian Res 2023; 16:82. [PMID: 37095524 PMCID: PMC10127035 DOI: 10.1186/s13048-023-01155-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 04/06/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Necroptosis is a novel type of programmed cell death distinct from apoptosis. However, the role of necroptosis in ovarian cancer (OC) remains unclear. The present study investigated the prognostic value of necroptosis-related genes (NRGs) and the immune landscape in OC. METHODS The gene expression profiling and clinical information were downloaded from the TCGA and GTEx databases. Differentially expressed NRGs (DE-NRGs) between OC and normal tissueswere identified. The regression analyses were conducted to screen the prognostic NRGs and construct the predictive risk model. Patients were then divided into high- and low-risk groups, and the GO and KEGG analyses were performed to explore bioinformatics functions between the two groups. Subsequently, the risk level and immune status correlations were assessed through the ESTIMATE and CIBERSORT algorithms. The tumor mutation burden (TMB) and the drug sensitivity were also analyzed based on the two-NRG signature in OC. RESULTS Totally 42 DE-NRGs were identified in OC. The regression analyses screened out two NRGs (MAPK10 and STAT4) with prognostic values for overall survival. The ROC curve showed a better predictive ability in five-year OS using the risk score. Immune-related functions were significantly enriched in the high- and low-risk group. Macrophages M1, T cells CD4 memory activated, T cells CD8, and T cells regulatory infiltration immune cells were associated with the low-risk score. The lower tumor microenvironment score was demonstrated in the high-risk group. Patients with lower TMB in the low-risk group showed a better prognosis, and a lower TIDE score suggested a better immune checkpoint inhibitor response in the high-risk group. Besides, cisplatin and paclitaxel were found to be more sensitive in the low-risk group. CONCLUSIONS MAPK10 and STAT4 can be important prognosis factors in OC, and the two-gene signature performs well in predicting survival outcomes. Our study provided novel ways of OC prognosis estimation and potential treatment strategy.
Collapse
Affiliation(s)
- Sipei Nie
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Na Ni
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Ningxin Chen
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Min Gong
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Ercui Feng
- Department of Preventive Health Care, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, China
| | - Jinhui Liu
- Department of Gynecology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China.
| | - Qiaoling Liu
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, Jiangsu, China.
| |
Collapse
|
35
|
Lin CY, Huang KY, Kao SH, Lin MS, Lin CC, Yang SC, Chung WC, Chang YH, Chein RJ, Yang PC. Small-molecule PIK-93 modulates the tumor microenvironment to improve immune checkpoint blockade response. SCIENCE ADVANCES 2023; 9:eade9944. [PMID: 37027467 PMCID: PMC10081850 DOI: 10.1126/sciadv.ade9944] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/08/2023] [Indexed: 06/19/2023]
Abstract
Immune checkpoint inhibitors (ICIs) targeting PD-L1 immunotherapy are state-of-the-art treatments for advanced non-small cell lung cancer (NSCLC). However, the treatment response of certain patients with NSCLC is unsatisfactory because of an unfavorable tumor microenvironment (TME) and poor permeability of antibody-based ICIs. In this study, we aimed to discover small-molecule drugs that can modulate the TME to enhance ICI treatment efficacy in NSCLC in vitro and in vivo. We identified a PD-L1 protein-modulating small molecule, PIK-93, using a cell-based global protein stability (GPS) screening system. PIK-93 mediated PD-L1 ubiquitination by enhancing the PD-L1-Cullin-4A interaction. PIK-93 reduced PD-L1 levels on M1 macrophages and enhanced M1 antitumor cytotoxicity. Combined PIK-93 and anti-PD-L1 antibody treatment enhanced T cell activation, inhibited tumor growth, and increased tumor-infiltrating lymphocyte (TIL) recruitment in syngeneic and human peripheral blood mononuclear cell (PBMC) line-derived xenograft mouse models. PIK-93 facilitates a treatment-favorable TME when combined with anti-PD-L1 antibodies, thereby enhancing PD-1/PD-L1 blockade cancer immunotherapy.
Collapse
Affiliation(s)
- Chia-Yi Lin
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Kuo-Yen Huang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shih-Han Kao
- Resuscitation Science Center of Emphasis, Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ming-Shiu Lin
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Chih-Chien Lin
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Shuenn-Chen Yang
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Wei-Chia Chung
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Ya-Hsuan Chang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Rong-Jie Chein
- Institute of Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Pan-Chyr Yang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei 115, Taiwan
- Genomics Research Center, Academia Sinica, Nankang, Taipei 115, Taiwan
| |
Collapse
|
36
|
Sadhukhan P, Seiwert TY. The role of macrophages in the tumor microenvironment and tumor metabolism. Semin Immunopathol 2023; 45:187-201. [PMID: 37002376 DOI: 10.1007/s00281-023-00988-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/08/2023] [Indexed: 04/03/2023]
Abstract
The complexity and plasticity of the tumor microenvironment (TME) make it difficult to fully understand the intratumoral regulation of different cell types and their activities. Macrophages play a crucial role in the signaling dynamics of the TME. Among the different subtypes of macrophages, tumor-associated macrophages (TAMs) are often associated with poor prognosis, although some subtypes of TAMs can at the same time improve treatment responsiveness and lead to favorable clinical outcomes. TAMs are key regulators of cancer cell proliferation, metastasis, angiogenesis, extracellular matrix remodeling, tumor metabolism, and importantly immunosuppression in the TME by modulating various chemokines, cytokines, and growth factors. TAMs have been identified as a key contributor to resistance to chemotherapy and cancer immunotherapy. In this review article, we aim to discuss the mechanisms by which TAMs regulate innate and adaptive immune signaling in the TME and summarize recent preclinical research on the development of therapeutics targeting TAMs and tumor metabolism.
Collapse
Affiliation(s)
- Pritam Sadhukhan
- Johns Hopkins University, Skip Viragh Outpatient Cancer Building, Baltimore, MD, 21287, USA
| | - Tanguy Y Seiwert
- Johns Hopkins University, Skip Viragh Outpatient Cancer Building, Baltimore, MD, 21287, USA.
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
37
|
Truxova I, Cibula D, Spisek R, Fucikova J. Targeting tumor-associated macrophages for successful immunotherapy of ovarian carcinoma. J Immunother Cancer 2023; 11:jitc-2022-005968. [PMID: 36822672 PMCID: PMC9950980 DOI: 10.1136/jitc-2022-005968] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is among the top five causes of cancer-related death in women, largely reflecting early, prediagnosis dissemination of malignant cells to the peritoneum. Despite improvements in medical therapies, particularly with the implementation of novel drugs targeting homologous recombination deficiency, the survival rates of patients with EOC remain low. Unlike other neoplasms, EOC remains relatively insensitive to immune checkpoint inhibitors, which is correlated with a tumor microenvironment (TME) characterized by poor infiltration by immune cells and active immunosuppression dominated by immune components with tumor-promoting properties, especially tumor-associated macrophages (TAMs). In recent years, TAMs have attracted interest as potential therapeutic targets by seeking to reverse the immunosuppression in the TME and enhance the clinical efficacy of immunotherapy. Here, we review the key biological features of TAMs that affect tumor progression and their relevance as potential targets for treating EOC. We especially focus on the therapies that might modulate the recruitment, polarization, survival, and functional properties of TAMs in the TME of EOC that can be harnessed to develop superior combinatorial regimens with immunotherapy for the clinical care of patients with EOC.
Collapse
Affiliation(s)
| | - David Cibula
- Gynecologic Oncology Center, Department of Obstetrics and Gynecology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic,Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic .,Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
38
|
Yan C, Li K, Meng F, Chen L, Zhao J, Zhang Z, Xu D, Sun J, Zhou M. Integrated immunogenomic analysis of single-cell and bulk tissue transcriptome profiling unravels a macrophage activation paradigm associated with immunologically and clinically distinct behaviors in ovarian cancer. J Adv Res 2023; 44:149-160. [PMID: 36725186 PMCID: PMC9936412 DOI: 10.1016/j.jare.2022.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/14/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Increasing evidence demonstrates that the activation states and diverse spectrum of macrophage subtypes display dynamic heterogeneity in the tumor microenvironment, which plays a critical role in a variety of cancer types. OBJECTIVES To investigate the heterogeneity and the homeostasis of different macrophage subtypes, as well as their effect on biological and clinical manifestations of ovarian cancer (OV). METHOD Integrated immunogenomic analysis of single-cell and bulk tissuetranscriptome profiling was performed to systematically investigate the association between macrophage activation and prognostic and therapeutic efficacy. Consensus clustering analysis was used to define novel macrophage subtypes. An artificial neural network was used to simulate the dynamic activation of macrophages. RESULTS The pan-cohort results suggested that high relative infiltration abundance of M0 and M1 macrophages was associated with improved outcome and therapeutic efficacy. However, it was the opposite for M2 macrophages. Unsupervised consensus clustering analysis revealed two OV subgroups characterized by a balance between M0, M1 and M2 macrophages with distinct clinical and immunological behaviors. Finally, a macrophage polarization-derived artificial neural network model was proposed to serve as a robust prognostic factor and predictive biomarker for therapeutic efficacy, which was validated in different independent patient cohorts. CONCLUSION The present study provides a new understanding of macrophage heterogeneity and its association with OV prognosis and underlines the future clinical potential of a macrophage activation model for tumor prevention and treatment.
Collapse
Affiliation(s)
- Congcong Yan
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Ke Li
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Fanling Meng
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin 150081, P. R. China
| | - Lu Chen
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Jingting Zhao
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Zicheng Zhang
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Dandan Xu
- School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, P. R. China.
| | - Jie Sun
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, P. R. China.
| | - Meng Zhou
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, P. R. China.
| |
Collapse
|
39
|
Zheng Y, Jiang B, Guo H, Zhang Z, Chen B, Zhang Z, Wu S, Zhao J. The combinational nano-immunotherapy of ferumoxytol and poly(I:C) inhibits melanoma via boosting anti-angiogenic immunity. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 49:102658. [PMID: 36708910 DOI: 10.1016/j.nano.2023.102658] [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: 10/18/2022] [Revised: 12/11/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
Angiogenesis plays a key role in the progression and metastasis of melanoma, and the pro-angiogenic effect of macrophages is one major reason for the failure of current anti-angiogenic therapies. Here, a nano-immunotherapy combining ferumoxytol and poly(I:C) (ferumoxytol/poly(I:C)) has been developed to boost the anti-angiogenic activities of macrophages to inhibit melanoma. Our findings demonstrated that ferumoxytol/poly(I:C) was a highly efficacious anti-tumor therapy with limited toxicity. Both in vivo and in vitro experiments indicated that this combination was successful in impeding angiogenesis. Ferumoxytol/poly(I:C) was demonstrated to reduce the viability of endothelial cells, thus hindering tube formation. Particularly, ferumoxytol/poly(I:C) was able to polarize macrophages to the M1 phenotype and decrease the expression of vascular endothelial growth factor, which in turn amplified the anti-angiogenic properties of ferumoxytol/poly(I:C). This combination of ferumoxytol/poly(I:C) nano-immunotherapy enriches the anti-angiogenic therapeutic nature of ferumoxytol and will shed new light on the treatment of melanoma.
Collapse
Affiliation(s)
- Yunuo Zheng
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China
| | - Bo Jiang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China; Department of Urology, Xuzhou Central Hospital, Xuzhou 221009, China
| | - Hongmei Guo
- Department of Ultrasonography, Weinan Maternal and Child Health Hospital, Weinan 714000, Shaanxi, China
| | - Zhonghai Zhang
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China; Department of Physiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Bo Chen
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Zhengkui Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, China.
| | - Shaoyuan Wu
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China.
| | - Jiaojiao Zhao
- Jiangsu Key Laboratory of Phylogenomics and Comparative Genomics, Jiangsu Joint International Center of Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China.
| |
Collapse
|
40
|
Schweer D, Anand N, Anderson A, McCorkle J, Neupane K, Nail AN, Harvey B, Hill KS, Ueland F, Richards C, Kolesar J. Human macrophage-engineered vesicles for utilization in ovarian cancer treatment. Front Oncol 2023; 12:1042730. [PMID: 36713536 PMCID: PMC9875020 DOI: 10.3389/fonc.2022.1042730] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Background Ovarian cancer is a deadly female malignancy with a high rate of recurrent and chemotherapy-resistant disease. Tumor-associated macrophages (TAMs) are a significant component of the tumor microenvironment and include high levels of M2-protumor macrophages that promote chemoresistance and metastatic spread. M2 macrophages can be converted to M1 anti-tumor macrophages, representing a novel therapeutic approach. Vesicles engineered from M1 macrophages (MEVs) are a novel method for converting M2 macrophages to M1 phenotype-like macrophages. Methods Macrophages were isolated and cultured from human peripheral blood mononuclear cells. Macrophages were stimulated to M1 or M2 phenotypes utilizing LPS/IFN-γ and IL-4/IL-13, respectively. M1 MEVs were generated with nitrogen cavitation and ultracentrifugation. Co-culture of ovarian cancer cells with macrophages and M1 MEVs was followed by cytokine, PCR, and cell viability analysis. Murine macrophage cell line, RAW264.7 cells were cultured and used to generate M1 MEVs for use in ovarian cancer xenograft models. Results M1 MEVs can effectively convert M2 macrophages to an M1-like state both in isolation and when co-cultured with ovarian cancer cells in vitro, resulting in a reduced ovarian cancer cell viability. Additionally, RAW264.7 M1 MEVs can localize to ovarian cancer tumor xenografts in mice. Conclusion Human M1 MEVs can repolarize M2 macrophages to a M1 state and have anti-cancer activity against ovarian cancer cell lines. RAW264.7 M1 MEVs localize to tumor xenografts in vivo murine models.
Collapse
Affiliation(s)
- David Schweer
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Namrata Anand
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States,Department of Pharmacy and Practice, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Abigail Anderson
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - J. Robert McCorkle
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Khaga Neupane
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - Alexandra N. Nail
- Department of Pharmacy and Practice, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Brock Harvey
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - Kristen S. Hill
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Frederick Ueland
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Christopher Richards
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - Jill Kolesar
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, University of Kentucky, Lexington, KY, United States,Department of Pharmacy and Practice, College of Pharmacy, University of Kentucky, Lexington, KY, United States,*Correspondence: Jill Kolesar,
| |
Collapse
|
41
|
Tursi NJ, Xu Z, Helble M, Walker S, Liaw K, Chokkalingam N, Kannan T, Wu Y, Tello-Ruiz E, Park DH, Zhu X, Wise MC, Smith TRF, Majumdar S, Kossenkov A, Kulp DW, Weiner DB. Engineered antibody cytokine chimera synergizes with DNA-launched nanoparticle vaccines to potentiate melanoma suppression in vivo. Front Immunol 2023; 14:1072810. [PMID: 36911698 PMCID: PMC9997082 DOI: 10.3389/fimmu.2023.1072810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/30/2023] [Indexed: 02/25/2023] Open
Abstract
Cancer immunotherapy has demonstrated great promise with several checkpoint inhibitors being approved as the first-line therapy for some types of cancer, and new engineered cytokines such as Neo2/15 now being evaluated in many studies. In this work, we designed antibody-cytokine chimera (ACC) scaffolding cytokine mimetics on a full-length tumor-specific antibody. We characterized the pharmacokinetic (PK) and pharmacodynamic (PD) properties of first-generation ACC TA99-Neo2/15, which synergized with DLnano-vaccines to suppress in vivo melanoma proliferation and induced significant systemic cytokine activation. A novel second-generation ACC TA99-HL2-KOA1, with retained IL-2Rβ/γ binding and attenuated but preserved IL-2Rα binding, induced lower systemic cytokine activation with non-inferior protection in murine tumor studies. Transcriptomic analyses demonstrated an upregulation of Type I interferon responsive genes, particularly ISG15, in dendritic cells, macrophages and monocytes following TA99-HL2-KOA1 treatment. Characterization of additional ACCs in combination with cancer vaccines will likely be an important area of research for treating melanoma and other types of cancer.
Collapse
Affiliation(s)
- Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ziyang Xu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michaela Helble
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Susanne Walker
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Kevin Liaw
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Neethu Chokkalingam
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Toshitha Kannan
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Yuanhan Wu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Edgar Tello-Ruiz
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Daniel H Park
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Xizhou Zhu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Megan C Wise
- Inovio Pharmaceuticals, Bluebell, PA, United States
| | | | - Sonali Majumdar
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Andrew Kossenkov
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Daniel W Kulp
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
42
|
Wu X, Lu W, Xu C, Jiang C, Zhuo Z, Wang R, Zhang D, Cui Y, Chang L, Zuo X, Wang Y, Mei H, Zhang W, Zhang M, Li C. Macrophages Phenotype Regulated by IL-6 Are Associated with the Prognosis of Platinum-Resistant Serous Ovarian Cancer: Integrated Analysis of Clinical Trial and Omics. J Immunol Res 2023; 2023:6455704. [PMID: 37124547 PMCID: PMC10132904 DOI: 10.1155/2023/6455704] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/06/2022] [Accepted: 03/22/2023] [Indexed: 05/02/2023] Open
Abstract
Background The treatment of platinum-resistant recurrent ovarian cancer (PROC) is a clinical challenge and a hot topic. Tumor microenvironment (TME) as a key factor promoting ovarian cancer progression. Macrophage is a component of TME, and it has been reported that macrophage phenotype is related to the development of PROC. However, the mechanism underlying macrophage polarization and whether macrophage phenotype can be used as a prognostic indicator of PROC remains unclear. Methods We used ESTIMATE to calculate the number of immune and stromal components in high-grade serous ovarian cancer (HGSOC) cases from The Cancer Genome Atlas database. The differential expression genes (DEGs) were analyzed via protein-protein interaction network, Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) analysis to reveal major pathways of DEGs. CD80 was selected for survival analysis. IL-6 was selected for gene set enrichment analysis (GSEA). A subsequent cohort study was performed to confirm the correlation of IL-6 expression with macrophage phenotype in peripheral blood and to explore the clinical utility of macrophage phenotype for the prognosis of PROC patients. Results A total of 993 intersecting genes were identified as candidates for further survival analysis. Further analysis revealed that CD80 expression was positively correlated with the survival of HGSOC patients. The results of GO and KEGG analysis suggested that macrophage polarization could be regulated via chemokine pathway and cytokine-cytokine receptor interaction. GSEA showed that the genes were mainly enriched in IL-6-STAT-3. Correlation analysis for the proportion of tumor infiltration macrophages revealed that M2 was correlated with IL-6. The results of a cohort study demonstrated that the regulation of macrophage phenotype by IL-6 is bidirectional. The high M1% was a protective factor for progression-free survival. Conclusion Thus, the macrophage phenotype is a prognostic indicator in PROC patients, possibly via a hyperactive IL-6-related pathway, providing an additional clue for the therapeutic intervention of PROC.
Collapse
Affiliation(s)
- Xiaoqing Wu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Wenping Lu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chaojie Xu
- The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Cuihong Jiang
- Department of Oncology, Guang'anmen Hospital South Campus, China Academy of Chinese Medical Sciences, Beijing 102627, China
| | - Zhili Zhuo
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ruipeng Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Dongni Zhang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yongjia Cui
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Lei Chang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xi Zuo
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ya'nan Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Heting Mei
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Weixuan Zhang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Mengfan Zhang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chen Li
- Department of Biology, Chemistry, and Pharmacy, Free University of Berlin, Berlin 14195, Germany
| |
Collapse
|
43
|
NUDT1 Could Be a Prognostic Biomarker and Correlated with Immune Infiltration in Clear Cell Renal Cell Carcinoma. Appl Bionics Biomech 2022; 2022:3669296. [PMID: 36606241 PMCID: PMC9808898 DOI: 10.1155/2022/3669296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/25/2022] [Indexed: 12/28/2022] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is a malignant tumor with high morbidity and mortality. As a member of the Nudix hydrolase superfamily, Nudix (nucleoside diphosphate-linked moiety X)-type motif 1 (NUDT1) is closely related to the occurrence and development of cancer. Our study aims to explore the role of NUDT1 in ccRCC and its relationship with immune infiltration. Methods The NUDT1 expression matrix and corresponding clinical information were obtained from The Cancer Genome Atlas (TCGA) database. The expression difference of NUDT1 in ccRCC and its relationship with the clinical characteristics were investigated using R software. Kaplan-Meier (K-M) analysis, univariate Cox regression, multivariate Cox regression, receiver operating characteristic (ROC) curve, and nomogram were utilized to evaluate the survival and prognosis of patients. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were utilized to explore the function of differential genes in low- or high-expression group of NUDT1. TCGA dataset and Tumor IMmune Estimation Resource (TIMER) database were utilized to explore the relationship between NUDT1 and immune infiltration. Finally, TCGA dataset was utilized for gene set enrichment analysis (GSEA). Results NUDT1 was not only overexpressed in ccRCC but also significantly correlated with clinicopathological features (P < 0.05). K-M survival analysis showed that upregulated NUDT1 was closely related to the decrease of overall survival (OS) and progression-free survival (PFS) in ccRCC patients. Multivariate Cox regression revealed that NUDT1 was a independent prognostic indicator (HR = 1.437, 95% CI: 1.065-1.939, P=0.018). The ROC curve showed that NUDT1 had a certain accuracy in predicting the outcome of ccRCC patiens. Furthermore, a total of 150 coexpressed genes and 1,886 differentially expressed genes (DEGs) were identified. GO/KEGG and GSEA results suggested that NUDT1 and its DEGs were involved in the immune-related pathways. NUDT1 expression was positively correlated with infiltrating levels of regulatory T cells (Tregs), CD8+ T cells, follicular helper T cells, and M0 macrophages. In addition, NUDT1 was positively related to immune checkpoints, such as PD-1, LAG3, CTLA4, and CD70, in ccRCC. Conclusion NUDT1 plays a key role in the prognosis and immune cell infiltration of ccRCC patients, indicating its potential use as a prognostic biomarker and therapeutic target.
Collapse
|
44
|
Gazzillo A, Polidoro MA, Soldani C, Franceschini B, Lleo A, Donadon M. Relationship between Epithelial-to-Mesenchymal Transition and Tumor-Associated Macrophages in Colorectal Liver Metastases. Int J Mol Sci 2022; 23:16197. [PMID: 36555840 PMCID: PMC9783529 DOI: 10.3390/ijms232416197] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
The liver is the most common metastatic site in colorectal cancer (CRC) patients. Indeed, 25-30% of the cases develop colorectal liver metastasis (CLM), showing an extremely poor 5-year survival rate and resistance to conventional anticancer therapies. Tumor-associated macrophages (TAMs) provide a nurturing microenvironment for CRC metastasis, promoting epithelial-to-mesenchymal transition (EMT) through the TGF-β signaling pathway, thus driving tumor cells to acquire mesenchymal properties that allow them to migrate from the primary tumor and invade the new metastatic site. EMT is known to contribute to the disruption of blood vessel integrity and the generation of circulating tumor cells (CTCs), thus being closely related to high metastatic potential in numerous solid cancers. Despite the fact that it is well-recognized that the crosstalk between tumor cells and the inflammatory microenvironment is crucial in the EMT process, the association between the EMT and the role of TAMs is still poorly understood. In this review, we elaborated on the role that TAMs exert in the induction of EMT during CLM development. Since TAMs are the major source of TGF-β in the liver, we also focused on novel insights into their role in TGF-β-induced EMT.
Collapse
Affiliation(s)
- Aurora Gazzillo
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Michela Anna Polidoro
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Cristiana Soldani
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Barbara Franceschini
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Ana Lleo
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Matteo Donadon
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, NO, Italy
- Department of General Surgery, University Maggiore Hospital Della Carità, 28100 Novara, NO, Italy
| |
Collapse
|
45
|
Lee JJ, Kang HJ, Kim SS, Charton C, Kim J, Lee JK. Unraveling the Transcriptomic Signatures of Homologous Recombination Deficiency in Ovarian Cancers. Adv Biol (Weinh) 2022; 6:e2200060. [PMID: 36116121 DOI: 10.1002/adbi.202200060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 08/03/2022] [Indexed: 01/28/2023]
Abstract
Homologous recombination deficiency (HRD) is a crucial driver of tumorigenesis by inducing impaired repair of double-stranded DNA breaks. Although HRD possibly triggers the production of numerous tumor neoantigens that sufficiently stimulate and activate various tumor-immune responses, a comprehensive understanding of the HRD-associated tumor microenvironment is elusive. To investigate the effect of HRD on the selective enrichment of transcriptomic signatures, 294 cases from The Cancer Genome Atlas-Ovarian Cancer project with both RNA-sequencing and SNP array data are analyzed. Differentially expressed gene analysis and network analysis are performed to identify HRD-specific signatures. Gene-sets associated with mitochondrial activation, including enhanced oxidative phosphorylation (OxPhos), are significantly enriched in the HRD-high group. Furthermore, a wide range of immune cell activation signatures is enriched in HRD-high cases of high-grade serous ovarian cancer (HGSOC). On further cell-type-specific analysis, M1-like macrophage genes are significantly enriched in HRD-high HGSOC cases, whereas M2-macrophage-related genes are not. The immune-response-associated genomic features, including tumor mutation rate, neoantigens, and tumor mutation burdens, correlated with HRD scores. In conclusion, the results of this study highlight the biological properties of HRD, including enhanced energy metabolism, increased tumor neoantigens and tumor mutation burdens, and consequent exacerbation of immune responses, particularly the enrichment of M1-like macrophages in HGSOC cases.
Collapse
Affiliation(s)
- Jae Jun Lee
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, 13620, Republic of Korea
| | - Hyun Ju Kang
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Stephanie S Kim
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, 13620, Republic of Korea
| | - Clémentine Charton
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, 13620, Republic of Korea
| | - Jinho Kim
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, 13620, Republic of Korea
| | - Jin-Ku Lee
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| |
Collapse
|
46
|
Xu T, Liu Z, Huang L, Jing J, Liu X. Modulating the tumor immune microenvironment with nanoparticles: A sword for improving the efficiency of ovarian cancer immunotherapy. Front Immunol 2022; 13:1057850. [PMID: 36532066 PMCID: PMC9751906 DOI: 10.3389/fimmu.2022.1057850] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
With encouraging antitumor effects, immunotherapy represented by immune checkpoint blockade has developed into a mainstream cancer therapeutic modality. However, only a minority of ovarian cancer (OC) patients could benefit from immunotherapy. The main reason is that most OC harbor a suppressive tumor immune microenvironment (TIME). Emerging studies suggest that M2 tumor-associated macrophages (TAMs), T regulatory cells (Tregs), myeloid-derived suppressor cells (MDSCs), and cancer-associated fibroblasts (CAFs) are enriched in OC. Thus, reversing the suppressive TIME is considered an ideal candidate for improving the efficiency of immunotherapy. Nanoparticles encapsulating immunoregulatory agents can regulate immunocytes and improve the TIME to boost the antitumor immune response. In addition, some nanoparticle-mediated photodynamic and photothermal therapy can directly kill tumor cells and induce tumor immunogenic cell death to activate antigen-presenting cells and promote T cell infiltration. These advantages make nanoparticles promising candidates for modulating the TIME and improving OC immunotherapy. In this review, we analyzed the composition and function of the TIME in OC and summarized the current clinical progress of OC immunotherapy. Then, we expounded on the promising advances in nanomaterial-mediated immunotherapy for modulating the TIME in OC. Finally, we discussed the obstacles and challenges in the clinical translation of this novel combination treatment regimen. We believe this resourceful strategy will open the door to effective immunotherapy of OC and benefit numerous patients.
Collapse
Affiliation(s)
| | | | | | - Jing Jing
- *Correspondence: Xiaowei Liu, ; Jing Jing,
| | | |
Collapse
|
47
|
Immunotherapeutic effects of intratumorally injected Zymosan-Adenovirus conjugates encoding constant active IRF3 in a melanoma mouse model. Immunol Res 2022; 71:197-212. [PMID: 36418765 DOI: 10.1007/s12026-022-09336-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/27/2022] [Indexed: 11/27/2022]
Abstract
M2-like tumor-associated macrophages (TAMs) play a significant role in immunosuppressive conditions in the tumor microenvironment (TME). TAM reprogramming, a dual-pronged therapy, reduces immunosuppression and induces immune favorable conditions in the TME. In this study, recombinant adenoviruses encoding active forms of interferon regulatory factor 3 (IRF3) were conjugated to zymosan particles to target phagocytic cells to create a pro-inflammatory immunomodulatory therapy. We determined TAM reprogramming by upregulation and downregulation of M1- and M2-associated genes, respectively, as well as cytokine and transcription factor expression in vitro. The overall shift to immune favorable conditions in the TME was suggested by metabolic, cytokine, and immune cell gene expression. Our data indicated that the zymosan:adenovirus (Zym:Ad) particle itself induced a shift from M2-like to M1-like TAMs, a shift in immune status of the TME, and systemic tumor immunity as determined using a double tumor melanoma mouse model and splenocyte functional assay. Notably, direct intratumoral injection of Zym:Ad IRF3 reduced tumor growth more significantly than Zym:Ad GFP, indicating additional therapeutic benefits due to incorporation of constant active IRF3.
Collapse
|
48
|
Rajtak A, Ostrowska-Leśko M, Żak K, Tarkowski R, Kotarski J, Okła K. Integration of local and systemic immunity in ovarian cancer: Implications for immunotherapy. Front Immunol 2022; 13:1018256. [PMID: 36439144 PMCID: PMC9684707 DOI: 10.3389/fimmu.2022.1018256] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/18/2022] [Indexed: 08/21/2023] Open
Abstract
Cancer is a disease that induces many local and systemic changes in immunity. The difficult nature of ovarian cancer stems from the lack of characteristic symptoms that contributes to a delayed diagnosis and treatment. Despite the enormous progress in immunotherapy, its efficacy remains limited. The heterogeneity of tumors, lack of diagnostic biomarkers, and complex immune landscape are the main challenges in the treatment of ovarian cancer. Integrative approaches that combine the tumor microenvironment - local immunity - together with periphery - systemic immunity - are urgently needed to improve the understanding of the disease and the efficacy of treatment. In fact, multiparametric analyses are poised to improve our understanding of ovarian tumor immunology. We outline an integrative approach including local and systemic immunity in ovarian cancer. Understanding the nature of both localized and systemic immune responses will be crucial to boosting the efficacy of immunotherapies in ovarian cancer patients.
Collapse
Affiliation(s)
- Alicja Rajtak
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Marta Ostrowska-Leśko
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
- Chair and Department of Toxicology, Medical University of Lublin, Lublin, Poland
| | - Klaudia Żak
- 1st Chair and Department of Oncological Gynaecology and Gynaecology, Student Scientific Association, Medical University of Lublin, Lublin, Poland
| | - Rafał Tarkowski
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Jan Kotarski
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Karolina Okła
- 1st Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
- Department of Surgery, University of Michigan Rogel Cancer Center, Ann Arbor, MI, United States
| |
Collapse
|
49
|
Maccio A, Madeddu C. Correspondence on "Quality is more important than quantity: pre-operative sarcopenia is associated with poor survival in advanced ovarian cancer" by Polen-De et al. Int J Gynecol Cancer 2022; 32:1493. [PMID: 36253005 PMCID: PMC9664104 DOI: 10.1136/ijgc-2022-003916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2022] [Indexed: 11/04/2022] Open
Affiliation(s)
- Antonio Maccio
- Department of Surgical Sciences, University of Cagliari, Monserrato, Italy
- Department of Gynecologic Oncology, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
50
|
Focus on organoids: cooperation and interconnection with extracellular vesicles - Is this the future of in vitro modeling? Semin Cancer Biol 2022; 86:367-381. [PMID: 34896267 DOI: 10.1016/j.semcancer.2021.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 01/27/2023]
Abstract
Organoids are simplified in vitro model systems of organs that are used for modeling tissue development and disease, drug screening, cell therapy, and personalized medicine. Despite considerable success in the design of organoids, challenges remain in achieving real-life applications. Organoids serve as unique and organized groups of micro physiological systems that are capable of self-renewal and self-organization. Moreover, they exhibit similar organ functionality(ies) as that of tissue(s) of origin. Organoids can be designed from adult stem cells, induced pluripotent stem cells, or embryonic stem cells. They consist of most of the important cell types of the desired tissue/organ along with the topology and cell-cell interactions that are highly similar to those of an in vivo tissue/organ. Organoids have gained interest in human biomedical research, as they demonstrate high promise for use in basic, translational, and applied research. As in vitro models, organoids offer significant opportunities for reducing the reliance and use of experimental animals. In this review, we will provide an overview of organoids, as well as those intercellular communications mediated by extracellular vesicles (EVs), and discuss the importance of organoids in modeling a tumor immune microenvironment (TIME). Organoids can also be exploited to develop a better understanding of intercellular communications mediated by EVs. Also, organoids are useful in mimicking TIME, thereby offering a better-controlled environment for studying various associated biological processes and immune cell types involved in tumor immunity, such as T-cells, macrophages, dendritic cells, and myeloid-derived suppressor cells, among others.
Collapse
|