151
|
Han T, Wu J, Liu Y, Zhou J, Miao R, Guo J, Xu Z, Xing Y, Bai Y, Hu D. Integrating bulk-RNA sequencing and single-cell sequencing analyses to characterize adenosine-enriched tumor microenvironment landscape and develop an adenosine-related prognostic signature predicting immunotherapy in lung adenocarcinoma. Funct Integr Genomics 2024; 24:19. [PMID: 38265702 DOI: 10.1007/s10142-023-01281-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 01/25/2024]
Abstract
The adenosine-signaling axis has been recognized as an important immunomodulatory pathway in tumor immunity. However, the biological role of the adenosine-signaling axis in the remodeling of the tumor microenvironment (TME) in lung adenocarcinoma (LUAD) remains unclear. Here, we quantified adenosine signaling (ado_sig) in LUAD samples using the GSVA method and assessed the prognostic value of adenosine in LUAD. Afterward, we explored the heterogeneity of the tumor-immune microenvironment at different adenosine levels. In addition, we analyzed the potential biological pathways engaged by adenosine. Next, we established single-cell transcriptional profiles of LUAD and analyzed cellular composition and cell-cell communication analysis under different adenosine microenvironments. Moreover, we established adenosine-related prognostic signatures (ARS) based on comprehensive bioinformatics analysis and evaluated the efficacy of ARS in predicting immunotherapy. The results demonstrated that adenosine signaling adversely impacted the survival of immune-enriched LUAD. The high-adenosine microenvironment exhibited elevated pro-tumor-immune infiltration, including M2 macrophages and displayed notably increased epithelial-mesenchymal transition (EMT) transformation. Furthermore, adenosine signaling displayed significant associations with the expression patterns and prognostic value of immunomodulators within the TME. Single-cell sequencing data revealed increased fibroblast occupancy and a prominent activation of the SPP1 signaling pathway in the high adenosine-signaling microenvironment. The ARS exhibited promising effectiveness in prognostication and predicting immunotherapy response in LUAD. In summary, overexpression of adenosine can cause a worsened prognosis in the LUAD with abundant immune infiltration. Moreover, increased adenosine levels are associated with pro-tumor-immune infiltration, active EMT transformation, pro-tumor angiogenesis, and other factors promoting cancer progression, which collectively contribute to the formation of an immunosuppressive microenvironment. Importantly, the ARS developed in this study demonstrate high efficacy in evaluating the response to immunotherapy.
Collapse
Affiliation(s)
- Tao Han
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China.
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China.
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Rui Miao
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Zhi Xu
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China
| | - Yingru Xing
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China
- Department of Clinical Laboratory, Anhui Zhongke Gengjiu Hospital, Hefei, People's Republic of China
| | - Ying Bai
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China.
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China.
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Chongren Building, No 168, Taifeng St, Huainan, People's Republic of China.
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, Huainan, People's Republic of China.
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, People's Republic of China.
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China.
| |
Collapse
|
152
|
Le Y, Gao H, Le J, Hornick JL, Bleday R, Wee J, Zhu Z. VentX promotes tumor specific immunity and efficacy of immune checkpoint inhibitors. iScience 2024; 27:108731. [PMID: 38299030 PMCID: PMC10829883 DOI: 10.1016/j.isci.2023.108731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/17/2023] [Accepted: 12/11/2023] [Indexed: 02/02/2024] Open
Abstract
Immune suppression within tumor microenvironments (TME) have been implicated in limited efficacy of immune check point inhibitors (ICIs) against solid tumors. Down-regulated VentX expression in tumor associated macrophages (TAMs) underlies phagocytotic anergic phenotype of TAMs, which govern immunological state of TME. In this study, using a tumor immune microenvironment enabling model system (TIME-EMS) of non-small cell lung cancer (NSCLC), we found that PD-1 antibody modestly activates cytotoxic T lymphocytes (CTLs) within the NSCLC-TME but not the status of TIME. We showed that the restoration of VentX expression in TAMs reignites the phagocytotic function of TAMs, which in turn, transforms TIME, activates CTLs in a tumor-specific manner and promotes efficacy of PD-1 antibody against NSCLC but not toxicity on normal lung epithelial cells. Supported by in vivo data on NSG-PDX models of primary human NSCLC, our study revealed potential venues to promote the efficacy of ICI against solid tumors through VentX-based mechanisms.
Collapse
Affiliation(s)
- Yi Le
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Hong Gao
- Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Joanna Le
- Department of Obstetrics and Gynecology, University of Massachusetts Medical Center
| | - Jason L. Hornick
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Ronald Bleday
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jon Wee
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Zhenglun Zhu
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
153
|
Zhao L, Chen J, Bai B, Song G, Zhang J, Yu H, Huang S, Wang Z, Lu G. Topical drug delivery strategies for enhancing drug effectiveness by skin barriers, drug delivery systems and individualized dosing. Front Pharmacol 2024; 14:1333986. [PMID: 38293666 PMCID: PMC10825035 DOI: 10.3389/fphar.2023.1333986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/27/2023] [Indexed: 02/01/2024] Open
Abstract
Topical drug delivery is widely used in various diseases because of the advantages of not passing through the gastrointestinal tract, avoiding gastrointestinal irritation and hepatic first-pass effect, and reaching the lesion directly to reduce unnecessary adverse reactions. The skin helps the organism to defend itself against a huge majority of external aggressions and is one of the most important lines of defense of the body. However, the skin's strong barrier ability is also a huge obstacle to the effectiveness of topical medications. Allowing the bioactive, composition in a drug to pass through the stratum corneum barrier as needed to reach the target site is the most essential need for the bioactive, composition to exert its therapeutic effect. The state of the skin barrier, the choice of delivery system for the bioactive, composition, and individualized disease detection and dosing planning influence the effectiveness of topical medications. Nowadays, enhancing transdermal absorption of topically applied drugs is the hottest research area. However, enhancing transdermal absorption of drugs is not the first choice to improve the effectiveness of all drugs. Excessive transdermal absorption enhances topical drug accumulation at non-target sites and the occurrence of adverse reactions. This paper introduces topical drug delivery strategies to improve drug effectiveness from three perspectives: skin barrier, drug delivery system and individualized drug delivery, describes the current status and shortcomings of topical drug research, and provides new directions and ideas for topical drug research.
Collapse
Affiliation(s)
- Lin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiamei Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bai Bai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guili Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiwei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhang Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guanghua Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
154
|
Chen B, Sun H, Zhang J, Xu J, Song Z, Zhan G, Bai X, Feng L. Cell-Based Micro/Nano-Robots for Biomedical Applications: A Review. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304607. [PMID: 37653591 DOI: 10.1002/smll.202304607] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/28/2023] [Indexed: 09/02/2023]
Abstract
Micro/nano-robots are powerful tools for biomedical applications and are applied in disease diagnosis, tumor imaging, drug delivery, and targeted therapy. Among the various types of micro-robots, cell-based micro-robots exhibit unique properties because of their different cell sources. In combination with various actuation methods, particularly externally propelled methods, cell-based microrobots have enormous potential for biomedical applications. This review introduces recent progress and applications of cell-based micro/nano-robots. Different actuation methods for micro/nano-robots are summarized, and cell-based micro-robots with different cell templates are introduced. Furthermore, the review focuses on the combination of cell-based micro/nano-robots with precise control using different external fields. Potential challenges, further prospects, and clinical translations are also discussed.
Collapse
Affiliation(s)
- Bo Chen
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, China
| | - Hongyan Sun
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, China
| | - Jiaying Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, China
| | - Junjie Xu
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, China
| | - Zeyu Song
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, China
| | - Guangdong Zhan
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, China
| | - Xue Bai
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China
| | - Lin Feng
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| |
Collapse
|
155
|
Liu Y, Peng C, Ahad F, Ali Zaidi SA, Muluh TA, Fu Q. Advanced Strategies of CAR-T Cell Therapy in Solid Tumors and Hematological Malignancies. Recent Pat Anticancer Drug Discov 2024; 19:557-572. [PMID: 38213150 DOI: 10.2174/0115748928277331231218115402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 01/13/2024]
Abstract
Chimeric antigen receptor T-cells, known as CAR-T cells, represent a promising breakthrough in the realm of adoptive cell therapy. These T-cells are genetically engineered to carry chimeric antigen receptors that specifically target tumors. They have achieved notable success in the treatment of blood-related cancers, breathing new life into this field of medical research. However, numerous obstacles limit chimeric antigen receptors T-cell therapy's efficacy, such as it cannot survive in the body long. It is prone to fatigue and exhaustion, leading to difficult tumor elimination and repeated recurrence, affecting solid tumors and hematological malignancies. The challenges posed by solid tumors, especially in the context of the complex solid-tumor microenvironment, require specific strategies. This review outlines recent advancements in improving chimeric antigen receptors T-cell therapy by focusing on the chimeric antigen receptors protein, modifying T-cells, and optimizing the interaction between T-cells and other components within the tumor microenvironment. This article aims to provide an extensive summary of the latest discoveries regarding CAR-T cell therapy, encompassing its application across various types of human cancers. Moreover, it will delve into the obstacles that have emerged in recent times, offering insights into the challenges faced by this innovative approach. Finally, it highlights novel therapeutic options in treating hematological and solid malignancies with chimeric antigen receptors T-cell therapies.
Collapse
Affiliation(s)
- Yangjie Liu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan, PRC China
| | - Cao Peng
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| | - Faiza Ahad
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Syed Aqib Ali Zaidi
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Qiuxia Fu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| |
Collapse
|
156
|
Jia D, He Y, Zhang Y. Long Non-coding RNAs Regulating Macrophage Polarization in Liver Cancer. Curr Pharm Des 2024; 30:2120-2128. [PMID: 38859791 DOI: 10.2174/0113816128311861240523075218] [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: 03/17/2024] [Accepted: 04/24/2024] [Indexed: 06/12/2024]
Abstract
Primary liver cancer is the second leading cause of cancer-related death worldwide. At present, liver cancer is often in an advanced stage once diagnosed, and treatment effects are generally poor. Therefore, there is an urgent need for other powerful treatments. Macrophages are an important component of the tumor microenvironment, and macrophage polarization is crucial to tumor proliferation and differentiation. Regulatory interactions between macrophage subtypes, such as M1 and M2, lead to a number of clinical outcomes, including tumor progression and metastasis. So, it is important to study the drivers of this process. Long non-coding RNA has been widely proven to be of great value in the early diagnosis and treatment of tumors. Many studies have shown that long non-coding RNA participates in macrophage polarization through its ability to drive M1 or M2 polarization, thereby participating in the occurrence and development of liver cancer. In this article, we systematically elaborated on the long non-coding RNAs involved in the polarization of liver cancer macrophages, hoping to provide a new idea for the early diagnosis and treatment of liver cancer. Liver cancer- related studies were retrieved from PubMed. Based on our identification of lncRNA and macrophage polarization as powerful therapies for liver cancer, we analyzed research articles in the PubMed system in the last ten years on the crosstalk between lncRNA and macrophage polarization. By targeting M1/M2 macrophage polarization, lncRNA may promote or suppress liver cancer, and the references are determined primarily by the article's impact factor. Consequently, the specific mechanism of action between lncRNA and M1/M2 macrophage polarization was explored, along with the role of their crosstalk in the occurrence, proliferation, and metastasis of liver cancer. LncRNA is bidirectionally expressed in liver cancer and can target macrophage polarization to regulate tumor behavior. LncRNA mainly functions as ceRNA and can participate in the crosstalk between liver cancer cells and macrophages through extracellular vesicles. LncRNA can potentially participate in the immunotherapy of liver cancer by targeting macrophages and becoming a new biomolecular marker of liver cancer.
Collapse
Affiliation(s)
- Dengke Jia
- Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Yaping He
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yawu Zhang
- Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730000, China
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China
| |
Collapse
|
157
|
Hu Q, Wang S, Cheng R, Liu Y, Chang Z, Huang Y, Chen Y, Luo X, Zhou L, Wang B, Gao Y, Chen H, Liu R, Zhang L. Tannins in Phyllanthus emblica L. improves cisplatin efficacy in lung cancer cells by boosting endoplasmic reticulum stress to trigger immunogenic cell death. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155219. [PMID: 38056150 DOI: 10.1016/j.phymed.2023.155219] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Lung cancer is one of the deadliest cancers world-wide and immunotherapy has been considered as a promising therapeutic strategy. Previously, our study found that tannins in Phyllanthus emblica L. (PTF) could inhibit the growth of tumor by activating the immune response in liver cancer, and also exhibited a cytotoxicity on human lung cancer cells A549, H460, H1703 in vitro. OBJECTIVE To explore whether PTF inhibited the growth of lung cancer through its immune-regulating function and to clarify underlying mechanisms. METHODS The induction of immunogenic cell death (ICD) were characterized by calreticulin exposure, extracellular ATP secretion, and High Mobility Group Box 1(HMGB1) release both in vivo using LLC-derived xenograft tumor model and in vitro using both mouse LLC and human A549 cancer cells. RESULTS PTF inhibited lung cancer cells growth and tumorigenesis in vivo/vitro and promoted anti-tumor immune responses. We further found that PTF could induce ICD, which then activated Type I interferon responses and CXCL9/10-mediated chemotaxis. Mechanistically, PTF induced the formation of intracellular protein aggregates and following activation of PERK/ATF4/CHOP-dependent endoplasmic reticulum stress-related ICD. Moreover, PTF improved the antitumor efficacy of cisplatin by inducing ICD both in vitro and in vivo. Finally, we screened out 5 components from PTF, including gallocatechin, gallic acid, methyl gallate, ethyl gallate and ellagic acid, which could induce ICD in vitro and might be considered as the potential antitumor pharmacodynamic substances. CONCLUSION In conclusion, PTF inhibits the growth of lung cancer by triggering ICD and remodeling the tumor microenvironment, suggesting that PTF may have promising prospects as an adjacent immunotherapy for cancers.
Collapse
Affiliation(s)
- Qian Hu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Shukai Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Ruiyang Cheng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Yuqi Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Zihao Chang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Ya Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Yinxin Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Xiaowei Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Lipeng Zhou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Baojin Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Ye Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Hongjiao Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China.
| | - Lanzhen Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Road, Beijing 102488, PR China.
| |
Collapse
|
158
|
LaMarche NM, Hegde S, Park MD, Maier BB, Troncoso L, Le Berichel J, Hamon P, Belabed M, Mattiuz R, Hennequin C, Chin T, Reid AM, Reyes-Torres I, Nemeth E, Zhang R, Olson OC, Doroshow DB, Rohs NC, Gomez JE, Veluswamy R, Hall N, Venturini N, Ginhoux F, Liu Z, Buckup M, Figueiredo I, Roudko V, Miyake K, Karasuyama H, Gonzalez-Kozlova E, Gnjatic S, Passegué E, Kim-Schulze S, Brown BD, Hirsch FR, Kim BS, Marron TU, Merad M. An IL-4 signalling axis in bone marrow drives pro-tumorigenic myelopoiesis. Nature 2024; 625:166-174. [PMID: 38057662 PMCID: PMC11189607 DOI: 10.1038/s41586-023-06797-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 10/30/2023] [Indexed: 12/08/2023]
Abstract
Myeloid cells are known to suppress antitumour immunity1. However, the molecular drivers of immunosuppressive myeloid cell states are not well defined. Here we used single-cell RNA sequencing of human and mouse non-small cell lung cancer (NSCLC) lesions, and found that in both species the type 2 cytokine interleukin-4 (IL-4) was predicted to be the primary driver of the tumour-infiltrating monocyte-derived macrophage phenotype. Using a panel of conditional knockout mice, we found that only deletion of the IL-4 receptor IL-4Rα in early myeloid progenitors in bone marrow reduced tumour burden, whereas deletion of IL-4Rα in downstream mature myeloid cells had no effect. Mechanistically, IL-4 derived from bone marrow basophils and eosinophils acted on granulocyte-monocyte progenitors to transcriptionally programme the development of immunosuppressive tumour-promoting myeloid cells. Consequentially, depletion of basophils profoundly reduced tumour burden and normalized myelopoiesis. We subsequently initiated a clinical trial of the IL-4Rα blocking antibody dupilumab2-5 given in conjunction with PD-1/PD-L1 checkpoint blockade in patients with relapsed or refractory NSCLC who had progressed on PD-1/PD-L1 blockade alone (ClinicalTrials.gov identifier NCT05013450 ). Dupilumab supplementation reduced circulating monocytes, expanded tumour-infiltrating CD8 T cells, and in one out of six patients, drove a near-complete clinical response two months after treatment. Our study defines a central role for IL-4 in controlling immunosuppressive myelopoiesis in cancer, identifies a novel combination therapy for immune checkpoint blockade in humans, and highlights cancer as a systemic malady that requires therapeutic strategies beyond the primary disease site.
Collapse
Affiliation(s)
- Nelson M LaMarche
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samarth Hegde
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Barbara B Maier
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Leanna Troncoso
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Le Berichel
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pauline Hamon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raphaël Mattiuz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clotilde Hennequin
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Theodore Chin
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda M Reid
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Iván Reyes-Torres
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erika Nemeth
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruiyuan Zhang
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Oakley C Olson
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Deborah B Doroshow
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicholas C Rohs
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jorge E Gomez
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rajwanth Veluswamy
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicole Hall
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicholas Venturini
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), BIOPOLIS, Singapore, Singapore
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, France
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- SingHealth Duke-NUS Academic Medical Centre, Translational Immunology Institute, Singapore, Singapore
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mark Buckup
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Igor Figueiredo
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vladimir Roudko
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kensuke Miyake
- Inflammation, Infection and Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Edgar Gonzalez-Kozlova
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Seunghee Kim-Schulze
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian D Brown
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fred R Hirsch
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian S Kim
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Thomas U Marron
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
159
|
Huang X, Yang X, Xiang L, Chen Y. Serine metabolism in macrophage polarization. Inflamm Res 2024; 73:83-98. [PMID: 38070057 DOI: 10.1007/s00011-023-01815-y] [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: 07/20/2023] [Revised: 10/09/2023] [Accepted: 11/06/2023] [Indexed: 01/10/2024] Open
Abstract
OBJECTIVE Emerging studies have revealed that macrophages possess different dependences on the uptake, synthesis, and metabolism of serine for their activation and functionalization, necessitating our insight into how serine availability and utilization impact macrophage activation and inflammatory responses. METHODS This article summarizes the reports published domestically and internationally about the serine uptake, synthesis, and metabolic flux by the macrophages polarizing with distinct stimuli and under different pathologic conditions, and particularly analyzes how altered serine metabolism rewires the metabolic behaviors of polarizing macrophages and their genetic and epigenetic reprogramming. RESULTS Macrophages dynamically change serine metabolism to orchestrate their anabolism, redox balance, mitochondrial function, epigenetics, and post-translation modification, and thus match the distinct needs for both classical and alternative activation. CONCLUSION Serine metabolism coordinates multiple metabolic pathways to tailor macrophage polarization and their responses to different pathogenic attacks and thus holds the potential as therapeutic target for types of acute and chronic inflammatory diseases.
Collapse
Affiliation(s)
| | - Xue Yang
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, 28 West Changsheng Road, Hengyang, 421001, Hunan, China
| | - Li Xiang
- Hengyang Medical School, Hengyang, China
| | - Yuping Chen
- Hengyang Medical School, Hengyang, China.
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, 28 West Changsheng Road, Hengyang, 421001, Hunan, China.
| |
Collapse
|
160
|
Chumak A, Fedosova N, Cheremshenko N, Karaman O, Симчич Т, Voyeykova I. EFFECT OF B. SUBTILIS ІМV B-7724 LECTIN ON THE ACTIVITY OF EFFECTORS OF CELLULAR ANTITUMOR IMMUNITY OF MICE WITH LEWIS LUNG CARCINOMA. Exp Oncol 2023; 45:328-336. [PMID: 38186022 DOI: 10.15407/exp-oncology.2023.03.328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Indexed: 01/09/2024]
Abstract
AIM To evaluate the effect of B. subtilis IMV B-7724 lectin on the functional activity of macrophages (Mph), natural killer (NK) cells and cytotoxic lymphocytes (CTL) of mice bearing Lewis lung carcinoma (LLC). MATERIALS AND METHODS The studies were performed on C57Bl/6J mice; LLC was used as an experimental transplantable tumor. The lectin from B. subtilis IMV B-7724 was administered to LLC-bearing mice subcutaneously at a dose of 1 mg/kg of body weight for 10 days. The immunological testing was performed on days 14, 21, and 28 after tumor grafting. The cytotoxic activity of Mph, NK, and CTL was estimated in MTT-assay; the content of the stable metabolites of nitric oxide (NO) was measured by a standard Griess reaction; the arginase activity (Arg) was determined based on the measurement of urea. RESULTS The administration of the B. subtilis IMV B-7724 lectin to LLC-bearing mice exerted its antitumor and antimetastatic effects partially via a significant (p < 0.05) increase of Mph and NK activities after the completion of the treatment. In the group of animals injected with lectin, the NO/Arg ratio increased significantly, indicating the prevalence of Mph with proinflammatory and antitumor properties. The cytotoxic activity of Mph exceeded the indices of untreated mice and intact control by 1.8 times and 5.3 times respectively; of NK - by 2.8 and 1.3 times respectively. The effect of treatment on the CTL activity was less pronounced. CONCLUSION Antitumor and antimetastatic activity of the lectin from B. subtilis IMV B-7724 ensured the preservation of the cytotoxic activity of the main effectors of antitumor immunity (Mph, NK, and CTL) throughout LLC growth.
Collapse
Affiliation(s)
- A Chumak
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - N Fedosova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - N Cheremshenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - O Karaman
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Т Симчич
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - I Voyeykova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
161
|
Wang S, Chen S, Li H, Ben S, Zhao T, Zheng R, Wang M, Gu D, Liu L. Causal genetic regulation of DNA replication on immune microenvironment in colorectal tumorigenesis: Evidenced by an integrated approach of trans-omics and GWAS. J Biomed Res 2023; 38:37-50. [PMID: 38111199 PMCID: PMC10818172 DOI: 10.7555/jbr.37.20230081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 12/20/2023] Open
Abstract
The interplay between DNA replication stress and immune microenvironment alterations is known to play a crucial role in colorectal tumorigenesis, but a comprehensive understanding of their association with and relevant biomarkers involved in colorectal tumorigenesis is lacking. To address this gap, we conducted a study aiming to investigate this association and identify relevant biomarkers. We analyzed transcriptomic and proteomic profiles of 904 colorectal tumor tissues and 342 normal tissues to examine pathway enrichment, biological activity, and the immune microenvironment. Additionally, we evaluated genetic effects of single variants and genes on colorectal cancer susceptibility using data from genome-wide association studies (GWASs) involving both East Asian (7062 cases and 195745 controls) and European (24476 cases and 23073 controls) populations. We employed mediation analysis to infer the causal pathway, and applied multiplex immunofluorescence to visualize colocalized biomarkers in colorectal tumors and immune cells. Our findings revealed that both DNA replication activity and the flap structure-specific endonuclease 1 ( FEN1) gene were significantly enriched in colorectal tumor tissues, compared with normal tissues. Moreover, a genetic variant rs4246215 G>T in FEN1 was associated with a decreased risk of colorectal cancer (odds ratio = 0.94, 95% confidence interval: 0.90-0.97, P meta = 4.70 × 10 -9). Importantly, we identified basophils and eosinophils that both exhibited a significantly decreased infiltration in colorectal tumors, and were regulated by rs4246215 through causal pathways involving both FEN1 and DNA replication. In conclusion, this trans-omics incorporating GWAS data provides insights into a plausible pathway connecting DNA replication and immunity, expanding biological knowledge of colorectal tumorigenesis and therapeutic targets.
Collapse
Affiliation(s)
- Sumeng Wang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Silu Chen
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Huiqin Li
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Shuai Ben
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Tingyu Zhao
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Rui Zheng
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Meilin Wang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Dongying Gu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Lingxiang Liu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
162
|
Yin H, Xie C, Zuo Z, Xie D, Wang Q. A CTL-Inspired Killing System Using Ultralow-Dose Chemical-Drugs to Induce a Pyroptosis-Mediated Antitumor Immune Function. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2309839. [PMID: 38102944 DOI: 10.1002/adma.202309839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/29/2023] [Indexed: 12/17/2023]
Abstract
A Cytotoxic T lymphocyte-inspired system capable of using ultralow-dose chemical drugs to manipulate cell death is needed to investigate the antitumor immunotherapy. Recent studies reveal pyroptosis promotes antitumor immune function. However, high-dose chemotherapy leads to cytokine release syndrome by pyroptosis. Therefore, pyroptosis-inducing ultralow-dose chemotherapy is potential in preclinical and clinical research, but its efficacy, safety, and the antitumor immune responses are not clear. Here, a near-infrared light controllable killing system (BIK system) is established by which ultralow-dose doxorubicin can be spatiotemporally transported to tumor cells and mediate efficient pyroptosis. This BIK system reduces total drug consumption to less than one-thirtieth the common dose in vitro. Moreover, this BIK system exhibited good tumor targeting and tumor penetration. This system is applied for pyroptosis-induced antitumor therapies, which shows less than ≈25 µg kg-1 doxorubicin is sufficient for tumor regression with negligible injuries to major organs. The antitumor immune function are proven to correlate with the impressive efficacy of pyroptosis-inducing ultralow-dose chemotherapy. This study provides new insights into the design of nanoassisted systems for activating the antitumor immunity by microstimulation; the application of the BIK system suggests that ultralow-dose chemotherapy is sufficient for inducing a robust pyroptosis-mediated antitumor immunity.
Collapse
Affiliation(s)
- Hao Yin
- Institute for Advanced Research, Wenzhou Medical University, Wenzhou, Zhejiang, 305006, P. R. China
| | - Congying Xie
- Department of Radiation and Medical Oncology, Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 305006, P. R. China
| | - Zhigui Zuo
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 305006, P. R. China
| | - Danli Xie
- Institute for Advanced Research, Wenzhou Medical University, Wenzhou, Zhejiang, 305006, P. R. China
| | - Qinyang Wang
- Institute for Advanced Research, Wenzhou Medical University, Wenzhou, Zhejiang, 305006, P. R. China
- Department of Radiation and Medical Oncology, Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 305006, P. R. China
| |
Collapse
|
163
|
Zhu W, Liu X, Yang L, He Q, Huang D, Tan X. Ferroptosis and tumor immunity: In perspective of the major cell components in the tumor microenvironment. Eur J Pharmacol 2023; 961:176124. [PMID: 37925133 DOI: 10.1016/j.ejphar.2023.176124] [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: 05/22/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/06/2023]
Abstract
Ferroptosis is an iron-dependent form of cell death driven by lipid peroxidation, which is morphologically, biochemically, and genetically distinct from apoptosis, necrosis, and autophagy. Mounting studies on the essential role of ferroptosis have been published in the progression of solid tumors, metastasis, therapy, and therapy resistance. Studies showed that ferroptosis is a "double-edged sword" in tumor immunity, which means it may have both tumor-antagonizing and tumor-promoting functions. The tumor microenvironment (TME) comprises not only tumor cells but also surrounding immune cells, stromal cells, as well as noncellular components such as the extracellular matrix (ECM), cytokines, growth factors, and extracellular vesicles (EVs). In the complex and diverse condition in TME where tumor cells grow, changes in each constituent may impact tumor destiny differently. Recently, several studies have revealed the interaction between ferroptosis and different constituents in TME. Both tumor cells and nontumor cells have a dual role in tumor immunity and influence tumor progression through ferroptosis. Herein, this review aims at summarizing the role of ferroptosis in tumor immunity based on TME, focusing on the mechanisms of the interaction between the ferroptosis and the different constituents in TME, illuminating how ferroptosis plays its role in promoting or antagonizing tumors by acting with varying components in TME and proposing several questions in immunomodulatory effects of ferroptosis and ferroptosis-associated immunotherapy.
Collapse
Affiliation(s)
- Wanling Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaowei Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qiang He
- Department of Cosmetic Surgery, Sichuan Provincial People's Hospital Medical Group Chengdu Newme Medical Cosmetic Hospital, 610041, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Xuelian Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
164
|
Qu B, Liu J, Peng Z, Xiao Z, Li S, Wu J, Li S, Luo J. CircSOD2 polarizes macrophages towards the M1 phenotype to alleviate cisplatin resistance in gastric cancer cells by targeting the miR-1296/STAT1 axis. Gene 2023; 887:147733. [PMID: 37625563 DOI: 10.1016/j.gene.2023.147733] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/06/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Cisplatin is the first-line drug for gastric cancer (GC). Cisplatin resistance is the most important cause of poor prognosis for GC. Increasing evidence has identified the important role of macrophage polarization in chemoresistance. CircRNAs are newly discovered non-coding RNAs, characterized by covalently closed loops with high stability. Previous studies have reported a significant difference between circRNA profiles expressed in classically activated M1 macrophages, and those expressed in alternatively activated M2 macrophages. However, the underlying mechanism behind the regulation of GC cisplatin resistance by macrophages remains unclear. In our study, we observed the aberrant high expression of circSOD2 in M1 macrophages derived from THP-1. These expression patterns were confirmed in macrophages from patients with GC. Detection of the M1 and M2 markers confirmed that overexpression of circSOD2 enhances M1 polarization. The viability of cisplatin-treated GC cells was significantly reduced in the presence of macrophages overexpressing circSOD2, and cisplatin-induced apoptosis increased dramatically. In vivo experiments showed that macrophages expressing circSOD2 enhanced the effect of cisplatin. Moreover, we demonstrated that circSOD2 acts as a microRNA sponge for miR-1296 and regulates the expression of its target gene STAT1 (signal transducer and activator of transcription 1). CircSOD2 exerts its function through the miR-1296/STAT1 axis. Inhibition of circSOD2/miR-1296/STAT1 may therefore reduce M1 polarization. Overexpression of circSOD2 promotes the polarization of M1 macrophages and enhances the effect of cisplatin in GC. CircSOD2 is a novel positive regulator of M1 macrophages and may serve as a potential target for GC chemotherapy.
Collapse
Affiliation(s)
- Bing Qu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jiasheng Liu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhiyang Peng
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhe Xiao
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shijun Li
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jianguo Wu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shengbo Li
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jianfei Luo
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
165
|
Xia Z, Mu W, Yuan S, Fu S, Liu Y, Zhang N. Cell Membrane Biomimetic Nano-Delivery Systems for Cancer Therapy. Pharmaceutics 2023; 15:2770. [PMID: 38140108 PMCID: PMC10748133 DOI: 10.3390/pharmaceutics15122770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Nano-delivery systems have demonstrated great promise in the therapy of cancer. However, the therapeutic efficacy of conventional nanomedicines is hindered by the clearance of the blood circulation system and the physiological barriers surrounding the tumor. Inspired by the unique capabilities of cells within the body, such as immune evasion, prolonged circulation, and tumor-targeting, there has been a growing interest in developing cell membrane biomimetic nanomedicine delivery systems. Cell membrane modification on nanoparticle surfaces can prolong circulation time, activate tumor-targeting, and ultimately improve the efficacy of cancer treatment. It shows excellent development potential. This review will focus on the advancements in various cell membrane nano-drug delivery systems for cancer therapy and the obstacles encountered during clinical implementation. It is hoped that such discussions will inspire the development of cell membrane biomimetic nanomedical systems.
Collapse
Affiliation(s)
- Zhenxing Xia
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Weiwei Mu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shijun Yuan
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shunli Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Yongjun Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Na Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| |
Collapse
|
166
|
Zhu Z, He L, Bai Y, Xia L, Sun X, Qi C. Yeast β-glucan modulates macrophages and improves antitumor NK-cell responses in cancer. Clin Exp Immunol 2023; 214:50-60. [PMID: 37455659 PMCID: PMC10711352 DOI: 10.1093/cei/uxad080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/24/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023] Open
Abstract
As the largest proportion of myeloid immune cells in tumors, macrophages play an important role in tumor growth and regression according to their different phenotypes, thus reprogramming macrophages has become a new research direction for cancer immunotherapy. Yeast-derived whole β-glucan particles (WGPs) can induce M0 macrophages to differentiate into M1 macrophages and convert M2 macrophages and tumor-associated macrophages (TAMs) into M1 macrophages. In vitro, studies have confirmed that WGP-treated macrophages increase the activating receptors in natural killer cells (NK cells) and enhance the cytotoxicity of NK cells. The extracellular regulated protein kinases (ERK) signaling pathway is involved in WGP-mediated regulation of the macrophage phenotype. Further in vivo studies show that oral WGP can significantly delay tumor growth, which is related to the increased proportion of macrophages and NK cells, the macrophage phenotype reversal, and the enhancement of NK cell immune function. NK-cell depletion reduces the therapeutic efficacy of WGP in tumor-bearing mice. These findings revealed that in addition to T cells, NK cells also participate in the antitumor process of WGP. It was confirmed that WGP regulates the macrophage phenotype to regulate NK-cell function.
Collapse
Affiliation(s)
- Zhichao Zhu
- Laboratory of Oncology, Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Liuyang He
- Laboratory of Oncology, Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Yu Bai
- Laboratory of Oncology, Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Lei Xia
- Laboratory of Oncology, Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Xiao Sun
- Laboratory of Oncology, Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Chunjian Qi
- Laboratory of Oncology, Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
167
|
Song Y, Liu H, Pan S, Xiang X, Ye M, Zhu X. Emerging role of mesenchymal stromal cells in gynecologic cancer therapy. Stem Cell Res Ther 2023; 14:347. [PMID: 38049868 PMCID: PMC10696729 DOI: 10.1186/s13287-023-03585-0] [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: 04/22/2023] [Accepted: 11/23/2023] [Indexed: 12/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) show considerable promise in regenerative medicine with superior anti-fibrotic, immunomodulatory, and angiogenic functions. More recently, discovered with the tumor tropism, MSCs have been exploited as the basis of targeted cancer therapy. In this scenario, MSCs can directly home to tumor tissues and play anti-tumor properties. In addition, MSCs, MSC-derived exosomes and MSC-derived membranes are often developed as carriers for precisely delivering cytotoxic agents to cancer sites, including chemotherapeutic drugs, therapeutic genes, or oncolytic viruses. However, it has revealed the tumorigenic risk of MSCs as an important component within the tumor microenvironment, hampering the translation of MSC-based cancer therapies into clinical settings. Therefore, in this review, we introduce the specific tumor-tropic ability of MSCs and underlying mechanisms. We also summarize the current application of MSC-based therapeutic approaches in treating gynecologic cancers, mainly including cervical, ovarian, and endometrial cancers. Moreover, we discuss the main challenges that the current MSC-based cancer therapies are facing.
Collapse
Affiliation(s)
- Yizuo Song
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Hejing Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Shuya Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Xinli Xiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Miaomiao Ye
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China.
| |
Collapse
|
168
|
Lafuente-Gómez N, de Lázaro I, Dhanjani M, García-Soriano D, Sobral MC, Salas G, Mooney DJ, Somoza Á. Multifunctional magnetic nanoparticles elicit anti-tumor immunity in a mouse melanoma model. Mater Today Bio 2023; 23:100817. [PMID: 37822453 PMCID: PMC10562177 DOI: 10.1016/j.mtbio.2023.100817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023] Open
Abstract
Immunotherapy has emerged as a promising strategy to eradicate cancer cells. Particularly, the development of cancer vaccines to induce a potent and sustained antigen-specific T cell response has become a center of attention. Herein, we describe a novel immunotherapy based on magnetic nanoparticles (MNP) covalently modified with the OVA254-267 antigen and a CpG oligonucleotide via disulfide bonds. The MNP-CpG-COVA significantly enhances dendritic cell activation and CD8+ T cell antitumoral response against B16-OVA melanoma cells in vitro. Notably, the immune response induced by the covalently modified MNP is more potent and sustained over time than that triggered by the free components, highlighting the advantage of nanoformulations in immunotherapies. What is more, the nanoparticles are stable in the blood after in vivo administration and induce potent levels of systemic tumor-specific effector CD8 + T cells. Overall, our findings highlight the potential of covalently functionalized MNP to induce robust immune responses against mouse melanoma.
Collapse
Affiliation(s)
- Nuria Lafuente-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Immunology Service, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa, Madrid, 28006, Spain
| | - Irene de Lázaro
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, New York University, New York, NY, 10010, USA
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York University, New York, NY, 10010, USA
| | - Mónica Dhanjani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
| | - David García-Soriano
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
| | - Miguel C. Sobral
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Gorka Salas
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Unidad de Nanobiotecnología Asociada al Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain
| | - David J. Mooney
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Unidad de Nanobiotecnología Asociada al Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain
| |
Collapse
|
169
|
Shen Z, Teng M, Han L, Bian D, Zhang J, Zhu X, Qing Y, Hu S, Chen Y, Yao W, Yu H, Zhang L, Zhang P. The impact of oncogenic driver mutations on neoadjuvant immunotherapy outcomes in patients with resectable non-small cell lung cancer. Cancer Immunol Immunother 2023; 72:4235-4247. [PMID: 37932425 PMCID: PMC10992055 DOI: 10.1007/s00262-023-03560-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/14/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Neoadjuvant immunotherapy has been demonstrated to be effective and safe in resectable non-small cell lung cancer (NSCLC) patients. However, the presence of different oncogenic driver mutations may affect the tumor microenvironment and consequently influence the clinical benefit from immunotherapy. METHODS This retrospective study included consecutive NSCLC patients (stage IIA to IIIB) who underwent radical surgery after receiving neoadjuvant immunotherapy at a single high-volume center between December 2019 and August 2022. Pathological response and long-term outcomes were compared based on the driver oncogene status, and RNA sequencing analysis was conducted to investigate the transcriptomic characteristics before and after treatment. RESULTS Of the 167 patients included in this study, 47 had oncogenic driver mutations. KRAS driver mutations were identified in 28 patients, representing 59.6% of oncogenic driver mutations. Of these, 17 patients had a major pathological response, which was significantly higher than in the non-KRAS driver mutation group (60.7% vs. 31.6%, P = 0.049). Multivariate Cox regression analysis further revealed that the KRAS driver mutation group was an independent prognostic factor for prolonged disease-free survival (hazard ratio: 0.10, P = 0.032). The median proportion of CD8+ T cells was significantly higher in the KRAS driver mutation NSCLCs than in the non-driver mutation group (18% vs. 13%, P = 0.030). Furthermore, immune-related pathways were enriched in the KRAS driver mutation NSCLCs and activated after immunotherapy. CONCLUSION Our study suggests that NSCLC patients with KRAS driver mutations have a superior response to neoadjuvant immunotherapy, possibly due to their higher immunogenicity. The findings highlight the importance of considering oncogenic driver mutations in selecting neoadjuvant treatment strategies for NSCLC patients.
Collapse
Affiliation(s)
- Ziyun Shen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Meixin Teng
- Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Lu Han
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Dongliang Bian
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Jing Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Xinsheng Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Yang Qing
- Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Shiqi Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yan Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Wangchao Yao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Huansha Yu
- Experimental Animal Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Lele Zhang
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
| |
Collapse
|
170
|
Shimasaki N, Shimizu E, Nakamura Y, Iguchi H, Ueda A, Umekage M, Haneda S, Mazda O. Size control of induced pluripotent stem cells colonies in two-dimensional culture for differentiation into functional monocyte-like cells. Cytotherapy 2023; 25:1338-1348. [PMID: 37676216 DOI: 10.1016/j.jcyt.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 07/17/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND AIMS Monocytes, derived from hematopoietic stem cells (HSCs), play a pivotal role in the immune response to cancer. Although they are an attractive source of cell therapy for cancer, a method for ex vivo expansion has not yet been established. Monocytes differentiated from pluripotent stem cells (PSCs), including induced pluripotent stem cells (iPSCs), can be an alternative source of HSC-derived monocytes because of their self-renewal and pluripotency. To develop a standardized method for the generation of iPSC-derived monocytes for future clinical applications, we aim to control the size of the iPSC colony. METHODS To this end, we developed a plate with multiple dots containing a chemical substrate for the iPSC scaffold. iPSCs placed in the plate expanded only on the dots and created colonies of the same size. The cells were then differentiated into monocytes by adding cytokines to the colonies. RESULTS The dot plate substantially reduced variability in monocyte-like cell generation when compared with cultivating cells on a plate with the substrate covering the entire surface area. Furthermore, more monocyte-like cells were obtained by adjusting the dot size and the distance between the dots. The iPSC-derived monocyte-like cells phagocytosed cancer cells and secreted proinflammatory cytokines. The cells also expressed Fc receptors and exerted immunoglobulin G-mediated killing of cancer cells with the corresponding antibodies. CONCLUSIONS The dot plate enabled the control of iPSC colony size in two-dimensional culture, which resulted in a reduction in the generation-variation of functional monocyte-like cells. This standardized method for generating iPSC-derived monocyte-like cells using the dot plate could also facilitate the development of an automated closed system on a large scale for clinical applications.
Collapse
Affiliation(s)
- Noriko Shimasaki
- Center for iPS Cell Research and Application Foundation, Kyoto University, Kyoto, Japan; Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Center for Pediatric Cancer Treatment, Nagoya University Hospital, Nagoya, Japan.
| | - Eiko Shimizu
- Center for iPS Cell Research and Application Foundation, Kyoto University, Kyoto, Japan
| | - Yuta Nakamura
- R&D Center Corporate, Sekisui Chemical Co., Ltd., Osaka, Japan
| | - Hiroki Iguchi
- R&D Center Corporate, Sekisui Chemical Co., Ltd., Osaka, Japan
| | - Anna Ueda
- Center for iPS Cell Research and Application Foundation, Kyoto University, Kyoto, Japan
| | - Masafumi Umekage
- Center for iPS Cell Research and Application Foundation, Kyoto University, Kyoto, Japan
| | - Satoshi Haneda
- R&D Center Corporate, Sekisui Chemical Co., Ltd., Osaka, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
171
|
Qi YQ, Xiong F, Chen YJ. The correlation between tumor-associated macrophages and the prognosis of east Asian hepatocellular carcinoma patients: A systematic review and meta-analysis. Pathol Res Pract 2023; 252:154919. [PMID: 37939428 DOI: 10.1016/j.prp.2023.154919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Previous related studies have found that the levels of tumor-associated macrophages (TAMs) were correlated with prognoses in hepatocellular carcinoma. However, the prognostic value of TAMs for East Asian HCC patients remains inconclusive. METHODS Our objectives were to systematically review the performance and explore the prognostic and clinical value of TAMs in patients with HCC. A total of 23 relevant studies of 4389 patients were included into our meta-analysis. And the work has been reported in line with PRISMA guidelines. RESULTS The results demonstrated that increased expression level of peritumoral infiltrated CD68+ macrophages had a poor prognostic value on overall survival (OS), disease free survival (DFS) and recurrence-free survival (RFS). However, there was no correlation between disease-free survival (DFS) and the abundance of CD68+ TAMs both in intratumoral regions. Additionally, low density of CD169+, high density of CD206, and high density of CD204+ TAMs had a worse prognostic value on OS while the CD163+ TAMs had no diagnostic value on OS. The densities of CD68+ TAMs exhibited significantly correlation with AFP level and vascular invasion. The levels of CD169+ TAMs showed apparent relation to vascular invasion and TNM stages. CONCLUSION These findings indicate that TAMs may accomplish as significant prognostic biomarkers for East Asian HCC patients. However, further researches should be performed to estimate the clinical value of TAMs in HCC.
Collapse
Affiliation(s)
- Yong-Qiang Qi
- Department of Biliary-pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Fei Xiong
- Department of Biliary-pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yong-Jun Chen
- Department of Biliary-pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
172
|
Ruella M, Korell F, Porazzi P, Maus MV. Mechanisms of resistance to chimeric antigen receptor-T cells in haematological malignancies. Nat Rev Drug Discov 2023; 22:976-995. [PMID: 37907724 PMCID: PMC10965011 DOI: 10.1038/s41573-023-00807-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 11/02/2023]
Abstract
Chimeric antigen receptor (CAR)-T cells have recently emerged as a powerful therapeutic approach for the treatment of patients with chemotherapy-refractory or relapsed blood cancers, including acute lymphoblastic leukaemia, diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma and multiple myeloma. Nevertheless, resistance to CAR-T cell therapies occurs in most patients. In this Review, we summarize the resistance mechanisms to CAR-T cell immunotherapy by analysing CAR-T cell dysfunction, intrinsic tumour resistance and the immunosuppressive tumour microenvironment. We discuss current research strategies to overcome multiple resistance mechanisms, including optimization of the CAR design, improvement of in vivo T cell function and persistence, modulation of the immunosuppressive tumour microenvironment and synergistic combination strategies.
Collapse
Affiliation(s)
- Marco Ruella
- Division of Hematology and Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Felix Korell
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Patrizia Porazzi
- Division of Hematology and Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
173
|
Xue L, Wang C, Qian Y, Zhu W, Liu L, Yang X, Zhang S, Luo D. Tryptophan metabolism regulates inflammatory macrophage polarization as a predictive factor for breast cancer immunotherapy. Int Immunopharmacol 2023; 125:111196. [PMID: 37972471 DOI: 10.1016/j.intimp.2023.111196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Metabolic reprogramming plays a pivotal role in regulating macrophage polarization and function. However, the impact of macrophage tryptophan metabolism on polarization within the breast cancer microenvironment remains elusive. In this study, we used single-cell transcriptome analysis and found that macrophages had the highest tryptophan metabolic activity in breast cancer, melanoma, and head and neck squamous cell carcinoma (HNSC). Further analysis revealed that the tryptophan metabolic activity of macrophages was positively correlated with the M1 macrophage scores in breast cancer. Pancancer analysis found positive correlations between tryptophan metabolism and the M1 macrophage score in almost all tumor types. Spatial transcriptome analysis revealed higher tryptophan metabolism in regions with higher M1 macrophage score in breast cancer tissues. Immune infiltration analysis revealed that the high tryptophan metabolism group exhibited a higher immune score, an increased proportion of CD8+ T cells, augmented cytolytic activity mediated by CD8+ T cells, and elevated expression of immune checkpoint molecules. Spatial immunophenotype cohort analysis exhibited that breast cancer patients expected to respond to immunotherapy had stronger tryptophan metabolism, with a 73.8 % area under the ROC curve. Single-cell transcriptome analysis of the immunotherapy cohort found that patients responding to immunotherapy had higher macrophage tryptophan metabolism prior to treatment initiation. Finally, in vitro experiments demonstrated elevated expression of tryptophan metabolic enzymes in M1 macrophages. Moreover, tryptophan facilitated the expression of M1 polarization markers, whereas inhibitors of tryptophan metabolic enzymes, such as NLG919, LM10, and Ro 61-8048, inhibited the expression of M1 polarization markers. In conclusion, this study identified a dual role for macrophage tryptophan metabolism in breast cancer; on the one hand, it promotes macrophage M1 polarization, while on the other hand, it serves as a promising predictor for the effectiveness of immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Linxuan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, China
| | - Chao Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, China
| | - Yulu Qian
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, China
| | - Wenqiang Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, China
| | - Lina Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, China
| | - Xiaohong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, China
| | - Shuhua Zhang
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
174
|
Hadiloo K, Taremi S, Heidari M, Esmaeilzadeh A. The CAR macrophage cells, a novel generation of chimeric antigen-based approach against solid tumors. Biomark Res 2023; 11:103. [PMID: 38017494 PMCID: PMC10685521 DOI: 10.1186/s40364-023-00537-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
Today, adoptive cell therapy has many successes in cancer therapy, and this subject is brilliant in using chimeric antigen receptor T cells. The CAR T cell therapy, with its FDA-approved drugs, could treat several types of hematological malignancies and thus be very attractive for treating solid cancer. Unfortunately, the CAR T cell cannot be very functional in solid cancers due to its unique features. This treatment method has several harmful adverse effects that limit their applications, so novel treatments must use new cells like NK cells, NKT cells, and macrophage cells. Among these cells, the CAR macrophage cells, due to their brilliant innate features, are more attractive for solid tumor therapy and seem to be a better candidate for the prior treatment methods. The CAR macrophage cells have vital roles in the tumor microenvironment and, with their direct effect, can eliminate tumor cells efficiently. In addition, the CAR macrophage cells, due to being a part of the innate immune system, attended the tumor sites. With the high infiltration, their therapy modulations are more effective. This review investigates the last achievements in CAR-macrophage cells and the future of this immunotherapy treatment method.
Collapse
Affiliation(s)
- Kaveh Hadiloo
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Department of Immunology, Zanjan, Iran
| | - Siavash Taremi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahmood Heidari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran.
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
175
|
Liang Y, Xu Q, Gao Q. Advancing CAR-based immunotherapies in solid tumors: CAR- macrophages and neutrophils. Front Immunol 2023; 14:1291619. [PMID: 38090576 PMCID: PMC10715261 DOI: 10.3389/fimmu.2023.1291619] [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: 09/09/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Macrophages and neutrophils are the main components of the innate immune system and play important roles in promoting angiogenesis, extracellular matrix remodeling, cancer cell proliferation, and metastasis in the tumor microenvironment (TME). They can also be harnessed to mediate cytotoxic tumor killing effects and orchestrate effective anti-tumor immune responses with proper stimulation and modification. Therefore, macrophages and neutrophils have strong potential in cancer immunotherapy. In this review, we briefly outlined the applications of macrophages or neutrophils in adoptive cell therapies, and focused on chimeric antigen receptor (CAR)-engineered macrophages (CAR-Ms) and neutrophils (CAR-Ns). We summarized the construction strategies, the preclinical and clinical studies of CAR-Ms and CAR-Ns. In the end, we briefly discussed the limitations and challenges of CAR-Ms and CAR-Ns, as well as future research directions to extend their applications in treating solid tumors.
Collapse
Affiliation(s)
- Yanling Liang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Qumiao Xu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Qianqian Gao
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
176
|
Shen Z, Liu X, Fan G, Na J, Liu Q, Lin F, Zhang Z, Zhong L. Improving the therapeutic efficacy of oncolytic viruses for cancer: targeting macrophages. J Transl Med 2023; 21:842. [PMID: 37993941 PMCID: PMC10666393 DOI: 10.1186/s12967-023-04709-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023] Open
Abstract
Oncolytic viruses (OVs) for cancer treatment are in a rapid stage of development, and the direct tumor lysis and activation of a comprehensive host immune response are irreplaceable advantages of cancer immunotherapy. However, excessive antiviral immune responses also restrict the spread of OVs in vivo and the infection of tumor cells. Macrophages are functionally diverse innate immune cells that phagocytose tumor cells and present antigens to activate the immune response, while also limiting the delivery of OVs to tumors. Studies have shown that the functional propensity of macrophages between OVs and tumor cells affects the overall therapeutic effect of oncolytic virotherapy. How to effectively avoid the restrictive effect of macrophages on OVs and reshape the function of tumor-associated macrophages in oncolytic virotherapy is an important challenge we are now facing. Here, we review and summarize the complex dual role of macrophages in oncolytic virotherapy, highlighting how the functional characteristics of macrophage plasticity can be utilized to cooperate with OVs to enhance anti-tumor effects, as well as highlighting the importance of designing and optimizing delivery modalities for OVs in the future.
Collapse
Affiliation(s)
- Zhen Shen
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Guixiang Fan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Qiaoqiao Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Faquan Lin
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Zhikun Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
| | - Liping Zhong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
- Pharmaceutical College, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
177
|
Basak U, Sarkar T, Mukherjee S, Chakraborty S, Dutta A, Dutta S, Nayak D, Kaushik S, Das T, Sa G. Tumor-associated macrophages: an effective player of the tumor microenvironment. Front Immunol 2023; 14:1295257. [PMID: 38035101 PMCID: PMC10687432 DOI: 10.3389/fimmu.2023.1295257] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Cancer progression is primarily caused by interactions between transformed cells and the components of the tumor microenvironment (TME). TAMs (tumor-associated macrophages) make up the majority of the invading immune components, which are further categorized as anti-tumor M1 and pro-tumor M2 subtypes. While M1 is known to have anti-cancer properties, M2 is recognized to extend a protective role to the tumor. As a result, the tumor manipulates the TME in such a way that it induces macrophage infiltration and M1 to M2 switching bias to secure its survival. This M2-TAM bias in the TME promotes cancer cell proliferation, neoangiogenesis, lymphangiogenesis, epithelial-to-mesenchymal transition, matrix remodeling for metastatic support, and TME manipulation to an immunosuppressive state. TAMs additionally promote the emergence of cancer stem cells (CSCs), which are known for their ability to originate, metastasize, and relapse into tumors. CSCs also help M2-TAM by revealing immune escape and survival strategies during the initiation and relapse phases. This review describes the reasons for immunotherapy failure and, thereby, devises better strategies to impair the tumor-TAM crosstalk. This study will shed light on the understudied TAM-mediated tumor progression and address the much-needed holistic approach to anti-cancer therapy, which encompasses targeting cancer cells, CSCs, and TAMs all at the same time.
Collapse
Affiliation(s)
- Udit Basak
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Tania Sarkar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Sumon Mukherjee
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Apratim Dutta
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Saikat Dutta
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Debadatta Nayak
- Central Council for Research in Homeopathy (CCRH), New Delhi, India
| | - Subhash Kaushik
- Central Council for Research in Homeopathy (CCRH), New Delhi, India
| | - Tanya Das
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
178
|
Aizaz M, Khan A, Khan F, Khan M, Musad Saleh EA, Nisar M, Baran N. The cross-talk between macrophages and tumor cells as a target for cancer treatment. Front Oncol 2023; 13:1259034. [PMID: 38033495 PMCID: PMC10682792 DOI: 10.3389/fonc.2023.1259034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
Macrophages represent an important component of the innate immune system. Under physiological conditions, macrophages, which are essential phagocytes, maintain a proinflammatory response and repair damaged tissue. However, these processes are often impaired upon tumorigenesis, in which tumor-associated macrophages (TAMs) protect and support the growth, proliferation, and invasion of tumor cells and promote suppression of antitumor immunity. TAM abundance is closely associated with poor outcome of cancer, with impediment of chemotherapy effectiveness and ultimately a dismal therapy response and inferior overall survival. Thus, cross-talk between cancer cells and TAMs is an important target for immune checkpoint therapies and metabolic interventions, spurring interest in it as a therapeutic vulnerability for both hematological cancers and solid tumors. Furthermore, targeting of this cross-talk has emerged as a promising strategy for cancer treatment with the antibody against CD47 protein, a critical macrophage checkpoint recognized as the "don't eat me" signal, as well as other metabolism-focused strategies. Therapies targeting CD47 constitute an important milestone in the advancement of anticancer research and have had promising effects on not only phagocytosis activation but also innate and adaptive immune system activation, effectively counteracting tumor cells' evasion of therapy as shown in the context of myeloid cancers. Targeting of CD47 signaling is only one of several possibilities to reverse the immunosuppressive and tumor-protective tumor environment with the aim of enhancing the antitumor response. Several preclinical studies identified signaling pathways that regulate the recruitment, polarization, or metabolism of TAMs. In this review, we summarize the current understanding of the role of macrophages in cancer progression and the mechanisms by which they communicate with tumor cells. Additionally, we dissect various therapeutic strategies developed to target macrophage-tumor cell cross-talk, including modulation of macrophage polarization, blockade of signaling pathways, and disruption of physical interactions between leukemia cells and macrophages. Finally, we highlight the challenges associated with tumor hypoxia and acidosis as barriers to effective cancer therapy and discuss opportunities for future research in this field.
Collapse
Affiliation(s)
- Muhammad Aizaz
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Aakif Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Faisal Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Maria Khan
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar, Pakistan
| | - Ebraheem Abdu Musad Saleh
- Department of Chemistry, College of Arts & Science, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Maryum Nisar
- School of Interdisciplinary Engineering & Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
179
|
Jiang J, Wang W, Zhu L, Shi B, Chen Y, Xia Y, Feng W, Yao W, Lu A, Zhang H. Unveiling the role of hypoxic macrophage-derived exosomes in driving colorectal cancer progression. Front Immunol 2023; 14:1260638. [PMID: 38022589 PMCID: PMC10666760 DOI: 10.3389/fimmu.2023.1260638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
The crosstalk between tumor cells and macrophages under hypoxic conditions has been acknowledged as a pivotal determinant in the progression of colorectal cancer (CRC). Previous research has underscored the significance of exosomes derived from hypoxic tumor cells in facilitating tumor progression through inducing the polarization of macrophages towards the M2-like phenotype. The precise influence of hypoxic macrophage-derived exosomes (HMDEs) on the progression of CRC has not yet been fully elucidated. The objective of this study was to investigate the role of HMDEs in the progression of CRC. We discovered that there was an elevated release of exosomes derived from macrophages in hypoxic conditions. Additionally, the hypoxia-induced macrophage-derived exosomes played a crucial role in promoting the progression of CRC. We have also demonstrated that HMDEs have the ability to induce cell cycle transition and inhibit cell apoptosis, thereby promoting the growth of CRC cells. Furthermore, the underlying molecular mechanisms of these effects have been identified. The overexpression of Hif-1α results in its direct interaction with distinct regions (-521- -516 bp and -401- -391 bp) of the Hsp90 promoter during hypoxic circumstances. This binding event led to the overexpression of Hsp90 and the subsequent elevation of Hsp90 protein levels within HMDEs. Importantly, the crucial interaction between Hsp90 and Lats1 resulted in the deactivation of Lats1 and the inhibition of Yap phosphorylation. Ultimately, this series of events lead to the deactivation of the Hippo signaling pathway. Our in vivo and in vitro studies presented compelling evidence for the crucial role of hypoxic macrophage-derived exosomal Hsp90 in promoting CRC progression through the inhibition of the Hippo signaling pathway. These findings represented a significant advancement in our comprehension of the complex interplay between macrophages and CRC cells under hypoxic conditions.
Collapse
Affiliation(s)
- Jiang Jiang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenfang Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lan Zhu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bowen Shi
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihan Xia
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiming Feng
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwu Yao
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aiguo Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
180
|
Lee D, Dunn ZS, Guo W, Rosenthal CJ, Penn NE, Yu Y, Zhou K, Li Z, Ma F, Li M, Song TC, Cen X, Li YR, Zhou JJ, Pellegrini M, Wang P, Yang L. Unlocking the potential of allogeneic Vδ2 T cells for ovarian cancer therapy through CD16 biomarker selection and CAR/IL-15 engineering. Nat Commun 2023; 14:6942. [PMID: 37938576 PMCID: PMC10632431 DOI: 10.1038/s41467-023-42619-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 10/17/2023] [Indexed: 11/09/2023] Open
Abstract
Allogeneic Vγ9Vδ2 (Vδ2) T cells have emerged as attractive candidates for developing cancer therapy due to their established safety in allogeneic contexts and inherent tumor-fighting capabilities. Nonetheless, the limited clinical success of Vδ2 T cell-based treatments may be attributed to donor variability, short-lived persistence, and tumor immune evasion. To address these constraints, we engineer Vδ2 T cells with enhanced attributes. By employing CD16 as a donor selection biomarker, we harness Vδ2 T cells characterized by heightened cytotoxicity and potent antibody-dependent cell-mediated cytotoxicity (ADCC) functionality. RNA sequencing analysis supports the augmented effector potential of Vδ2 T cells derived from CD16 high (CD16Hi) donors. Substantial enhancements are further achieved through CAR and IL-15 engineering methodologies. Preclinical investigations in two ovarian cancer models substantiate the effectiveness and safety of engineered CD16Hi Vδ2 T cells. These cells target tumors through multiple mechanisms, exhibit sustained in vivo persistence, and do not elicit graft-versus-host disease. These findings underscore the promise of engineered CD16Hi Vδ2 T cells as a viable therapeutic option for cancer treatment.
Collapse
Affiliation(s)
- Derek Lee
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Zachary Spencer Dunn
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
| | - Wenbin Guo
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Carl J Rosenthal
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Natalie E Penn
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Yanqi Yu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Kuangyi Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Zhe Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Feiyang Ma
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Miao Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Tsun-Ching Song
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Xinjian Cen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Jin J Zhou
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
181
|
Wu H, Wu Z, Ye D, Li H, Dai Y, Wang Z, Bao J, Xu Y, He X, Wang X, Dai X. Prognostic value analysis of cholesterol and cholesterol homeostasis related genes in breast cancer by Mendelian randomization and multi-omics machine learning. Front Oncol 2023; 13:1246880. [PMID: 38023262 PMCID: PMC10661325 DOI: 10.3389/fonc.2023.1246880] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The high incidence of breast cancer (BC) prompted us to explore more factors that might affect its occurrence, development, treatment, and also recurrence. Dysregulation of cholesterol metabolism has been widely observed in BC; however, the detailed role of how cholesterol metabolism affects chemo-sensitivity, and immune response, as well as the clinical outcome of BC is unknown. Methods With Mendelian randomization (MR) analysis, the potential causal relationship between genetic variants of cholesterol and BC risk was assessed first. Then we analyzed 73 cholesterol homeostasis-related genes (CHGs) in BC samples and their expression patterns in the TCGA cohort with consensus clustering analysis, aiming to figure out the relationship between cholesterol homeostasis and BC prognosis. Based on the CHG analysis, we established a CAG_score used for predicting therapeutic response and overall survival (OS) of BC patients. Furthermore, a machine learning method was adopted to accurately predict the prognosis of BC patients by comparing multi-omics differences of different risk groups. Results We observed that the alterations in plasma cholesterol appear to be correlative with the venture of BC (MR Egger, OR: 0.54, 95% CI: 0.35-0.84, p<0.006). The expression patterns of CHGs were classified into two distinct groups(C1 and C2). Notably, the C1 group exhibited a favorable prognosis characterized by a suppressed immune response and enhanced cholesterol metabolism in comparison to the C2 group. In addition, high CHG score were accompanied by high performance of tumor angiogenesis genes. Interestingly, the expression of vascular genes (CDH5, CLDN5, TIE1, JAM2, TEK) is lower in patients with high expression of CHGs, which means that these patients have poorer vascular stability. The CAG_score exhibits robust predictive capability for the immune microenvironment characteristics and prognosis of patients(AUC=0.79). It can also optimize the administration of various first-line drugs, including AKT inhibitors VIII Imatinib, Crizotinib, Saracatinib, Erlotinib, Dasatinib, Rapamycin, Roscovitine and Shikonin in BC patients. Finally, we employed machine learning techniques to construct a multi-omics prediction model(Risklight),with an area under the feature curve (AUC) of up to 0.89. Conclusion With the help of CAG_score and Risklight, we reveal the signature of cholesterol homeostasis-related genes for angiogenesis, immune responses, and the therapeutic response in breast cancer, which contributes to precision medicine and improved prognosis of BC.
Collapse
Affiliation(s)
- Haodong Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhixuan Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, Zhejiang, China
| | - Daijiao Ye
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongfeng Li
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yinwei Dai
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ziqiong Wang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingxia Bao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiying Xu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaofei He
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaowu Wang
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, Zhejiang, China
| | - Xuanxuan Dai
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
182
|
Wu HF, Liu H, Zhang ZW, Chen JM. CENPE and LDHA were potential prognostic biomarkers of chromophobe renal cell carcinoma. Eur J Med Res 2023; 28:481. [PMID: 37925501 PMCID: PMC10625266 DOI: 10.1186/s40001-023-01449-0] [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: 08/31/2023] [Accepted: 10/15/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Most sarcomatoid differentiated renal cell carcinoma was differentiated from Chromophobe renal cell carcinoma (KICH) and related to a bad prognosis. Thus, finding biomarkers is important for the therapy of KICH. METHODS The UCSC was used for determining the expression of mRNA and miRNA and clinical data in KICH and normal samples. KEGG and GO were used for predicting potential function of differently expressed genes (DEGs). Optimal prognostic markers were determined by Lasso regression. Kaplan-Meier survival, ROC, and cox regression were used for assessing prognosis value. GSEA was used for predicting potential function of markers. The relations between markers and immune cell infiltration were determined by Pearson method. The upstream miRNA of markers was predicted in TargetScan and DIANA. RESULTS The 6162 upregulated and 13,903 downregulated DEGs were identified in KICH. Further CENPE and LDHA were screened out as optimal prognostic risk signatures. CENPE was highly expressed while LDHA was lowly expressed in KICH samples, and the high expressions of 2 genes contributed to bad prognosis. The functions of CENPE and LDHA were mainly enriched in proliferation related pathways such as cell cycle and DNA replication. In addition, the correlation of 2 genes with immune infiltrates in KICH was also observed. Finally, we found that has-miR-577 was the common upstream of 2 genes and the binding sites can be predicted. CONCLUSION CENPE and LDHA were identified as the important prognostic biomarkers in KICH, and they might be involved in the proliferation of cancer cell.
Collapse
Affiliation(s)
- Hui-Feng Wu
- Department of Urology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China
| | - Hao Liu
- Department of Urology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China.
| | - Zhe-Wei Zhang
- Department of Urology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China
| | - Ji-Min Chen
- Department of Urology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China
| |
Collapse
|
183
|
Chang M, Ellmaker K, Esposito A, Lauricella A, Pa S, Roberts M, Kurt RA. The Impact of Tumor-Associated Macrophages Found at Mammary Carcinoma Sites is Dependent Upon the Tumor Type. Immunol Invest 2023; 52:909-924. [PMID: 37647339 DOI: 10.1080/08820139.2023.2252463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
BACKGROUND Previously, we reported that tumor-associated macrophages (TAM) at early sites of mammary carcinoma showed a decrease in ATP production rate and a higher dependence on oxidative phosphorylation. METHODS Since these changes can result from activation of AMP-activated protein kinase (AMPK) and glucose transporter 1 (Glut1) during metabolic stress, we investigated whether the TAM showed increased expression of ampk and glut1, as well as another indicator of metabolic stress, pkm2. Indeed, the TAM exhibited significant expression of pkm2, glut1, and ampk. RESULTS Bone marrow-derived macrophages (BMDM) co-cultured with 4T1, EMT6, and 168 in vitro similarly showed increased expression of pkm2, glut1, and ampk. Moreover, lactate, which is expressed at significant levels by all three tumors, induced expression of these same genes in BMDM suggesting that lactate may induce a metabolic stress response in these TAM. Yet, the three different mammary carcinoma models benefited from different targeting strategies. Macrophage depletion studies revealed that the TAM contributed to growth of the EMT6 tumor and metastasis of the 4T1 tumor. Targeting the stress response with the Integrated Stress Response Inhibitor (ISRIB), which targets eIF2, impacted 168 tumor progression, and ISRIB as well as FX-11, which targets lactate dehydrogenase, impacted 4T1 tumor progression and metastasis. CONCLUSIONS Collectively, these data demonstrate that targeting TAM or metabolism at early tumor sites can impact tumor progression. However, variability in the responses underscore the fact that the impact of macrophages differs even within three different syngeneic mammary carcinoma models.
Collapse
Affiliation(s)
- Musea Chang
- Department of Biology, Lafayette College, Easton, Pennsylvania, USA
| | | | - Abigail Esposito
- Department of Biology, Lafayette College, Easton, Pennsylvania, USA
| | | | - Sophea Pa
- Department of Biology, Lafayette College, Easton, Pennsylvania, USA
| | - Meghan Roberts
- Department of Biology, Lafayette College, Easton, Pennsylvania, USA
| | - Robert A Kurt
- Department of Biology, Lafayette College, Easton, Pennsylvania, USA
| |
Collapse
|
184
|
Zuo B, Li T, Liu X, Wang S, Cheng J, Liu X, Cui W, Shi H, Ling C. Dipeptidyl peptidase 4 inhibitor reduces tumor-associated macrophages and enhances anti-PD-L1-mediated tumor suppression in non-small cell lung cancer. Clin Transl Oncol 2023; 25:3188-3202. [PMID: 37115489 PMCID: PMC10514125 DOI: 10.1007/s12094-023-03187-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/03/2023] [Indexed: 04/29/2023]
Abstract
PURPOSE The efficacy of immune checkpoint inhibitors such as programmed cell death ligand 1 (PD-L1) antibodies in non-small cell lung cancer (NSCLC) is limited, and combined use with other therapies is recommended. Dipeptidyl peptidase 4 (DPP4) inhibitors, a class of small molecule inhibitors, are highly effective for treating type 2 diabetes. Emerging evidence implicates DPP4 inhibitors as immunomodulators that modify aspects of innate and adaptive immunity. We evaluated the combination of a DPP4 inhibitor (anagliptin) and PD-L1 blockade in an NSCLC mouse model. METHODS The effect of the combination of anti-PD-L1 and anagliptin was evaluated in subcutaneous mouse models of NSCLC. Tumor-infiltrating immune cells were analyzed by flow cytometry. Bone marrow-derived monocytes of C57BL/6 mice were isolated in vitro to examine the underlying mechanism of anagliptin on the differentiation and polarization of macrophage. RESULTS Anagliptin dramatically improved the efficacy of PD-L1 antibody monotherapy by inhibiting macrophage formation and M2 polarization in the tumor microenvironment. Mechanistically, anagliptin suppressed the production of reactive oxygen species in bone marrow monocytes by inhibiting NOX1 and NOX2 expression induced by macrophage colony-stimulating factor, reduced late ERK signaling pathway activation, and inhibited monocyte-macrophage differentiation. However, the inhibitory effect was reactivated by lipopolysaccharide and interferon-gamma interacting with corresponding receptors during M1 macrophage polarization, but not M2. CONCLUSIONS Anagliptin can enhance PD-L1 blockade efficacy in NSCLC by inhibiting macrophage differentiation and M2 macrophage polarization, and combination therapy may be a promising strategy for treating PD-L1 blockade therapy-resistant patients with NSCLC.
Collapse
Affiliation(s)
- Bei Zuo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Tao Li
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Xiaoyun Liu
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Shuling Wang
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Jianxiang Cheng
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Xiangqun Liu
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Wenjie Cui
- Department of Respiratory and Critical Care Medicine, The Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221116, China
| | - Hengliang Shi
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, China.
| | - Chunhua Ling
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China.
| |
Collapse
|
185
|
Kohn DB, Chen YY, Spencer MJ. Successes and challenges in clinical gene therapy. Gene Ther 2023; 30:738-746. [PMID: 37935854 PMCID: PMC10678346 DOI: 10.1038/s41434-023-00390-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/20/2023] [Accepted: 02/07/2023] [Indexed: 11/09/2023]
Abstract
Despite the ups and downs in the field over three decades, the science of gene therapy has continued to advance and provide enduring treatments for increasing number of diseases. There are active clinical trials approaching a variety of inherited and acquired disorders of different organ systems. Approaches include ex vivo modification of hematologic stem cells (HSC), T lymphocytes and other immune cells, as well as in vivo delivery of genes or gene editing reagents to the relevant target cells by either local or systemic administration. In this article, we highlight success and ongoing challenges in three areas of high activity in gene therapy: inherited blood cell diseases by targeting hematopoietic stem cells, malignant disorders using immune effector cells genetically modified with chimeric antigen receptors, and ophthalmologic, neurologic, and coagulation disorders using in vivo administration of adeno-associated virus (AAV) vectors. In recent years, there have been true cures for many of these diseases, with sustained clinical benefit that exceed those from other medical approaches. Each of these treatments faces ongoing challenges, namely their high one-time costs and the complexity of manufacturing the therapeutic agents, which are biological viruses and cell products, at pharmacologic standards of quality and consistency. New models of reimbursement are needed to make these innovative treatments widely available to patients in need.
Collapse
Affiliation(s)
- Donald B Kohn
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Yvonne Y Chen
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at UCLA, University of California, Los Angeles, Los Angeles, CA, USA
| | - Melissa J Spencer
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
186
|
Zhong W, Lu Y, Han X, Yang J, Qin Z, Zhang W, Yu Z, Wu B, Liu S, Xu W, Zheng C, Schuchter LM, Karakousis GC, Mitchell TC, Amaravadi R, Flowers AJ, Gimotty PA, Xiao M, Mills G, Herlyn M, Dong H, Mitchell MJ, Kim J, Xu X, Guo W. Upregulation of exosome secretion from tumor-associated macrophages plays a key role in the suppression of anti-tumor immunity. Cell Rep 2023; 42:113224. [PMID: 37805922 PMCID: PMC10697782 DOI: 10.1016/j.celrep.2023.113224] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 06/15/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023] Open
Abstract
Macrophages play a pivotal role in tumor immunity. We report that reprogramming of macrophages to tumor-associated macrophages (TAMs) promotes the secretion of exosomes. Mechanistically, increased exosome secretion is driven by MADD, which is phosphorylated by Akt upon TAM induction and activates Rab27a. TAM exosomes carry high levels of programmed death-ligand 1 (PD-L1) and potently suppress the proliferation and function of CD8+ T cells. Analysis of patient melanoma tissues indicates that TAM exosomes contribute significantly to CD8+ T cell suppression. Single-cell RNA sequencing analysis showed that exosome-related genes are highly expressed in macrophages in melanoma; TAM-specific RAB27A expression inversely correlates with CD8+ T cell infiltration. In a murine melanoma model, lipid nanoparticle delivery of small interfering RNAs (siRNAs) targeting macrophage RAB27A led to better T cell activation and sensitized tumors to anti-programmed cell death protein 1 (PD-1) treatment. Our study demonstrates tumors use TAM exosomes to combat CD8 T cells and suggests targeting TAM exosomes as a potential strategy to improve immunotherapies.
Collapse
Affiliation(s)
- Wenqun Zhong
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Youtao Lu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingbo Yang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhiyuan Qin
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Zhang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ziyan Yu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bin Wu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shujing Liu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cathy Zheng
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lynn M Schuchter
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giorgos C Karakousis
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Mitchell
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi Amaravadi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ahron J Flowers
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phyllis A Gimotty
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Xiao
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Gordon Mills
- Division of Oncological Science, School of Medicine, and Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Haidong Dong
- Departments of Urology and Immunology, Mayo College of Medicine and Science, Rochester, MN 55905, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junhyong Kim
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
187
|
Jia L, Liao L, Jiang Y, Hu X, Lu G, Xiao W, Gong W, Jia X. Low-dose adropin stimulates inflammasome activation of macrophage via mitochondrial ROS involved in colorectal cancer progression. BMC Cancer 2023; 23:1042. [PMID: 37904094 PMCID: PMC10614368 DOI: 10.1186/s12885-023-11519-5] [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: 04/17/2023] [Accepted: 10/11/2023] [Indexed: 11/01/2023] Open
Abstract
Adropin is encoded by the energy homeostasis-associated (ENHO) gene and widely present in liver, pancreas, heart, kidney, brain, and vascular tissues. Abnormal adropin is associated with metabolic, inflammatory, immune, and central nervous disorders. Whether adropin is involved in the development of colorectal cancer (CRC) is still unclear. Here, decreased adropin expression of tumor-nest cells in advanced-stage CRC was demonstrated. Adropin expressed by carcinoma cells was negatively correlated with macrophage infiltration in the matrix of CRC tissues. However, tumor macrophages enhanced adropin expression and were positively correlated with tumor invasion and metastasis. ENHO gene transfection into colon cancer (MC38) cells inhibited tumor growth in vivo, accompanying the increase of M1 macrophages. Treatment with low-dose adropin (< 100 ng/mL) on macrophages ex vivo directly increased mitochondrial reactive oxygen species for inflammasome activation. Furthermore, ENHO-/- mice had less M1 macrophages in vivo, and ENHO-/- macrophages were inert to be induced into the M1 subset ex vivo. Finally, low-dose adropin promoted glucose utilization, and high-dose adropin enhanced the expression of CPT1α in macrophages. Therefore, variations of adropin level in carcinoma cells or macrophages in tumor tissues are differently involved in CRC progression. Low-dose adropin stimulates the antitumor activity of macrophages, but high-dose adropin facilitates the pro-tumor activity of macrophages. Increasing or decreasing the adropin level can inhibit tumor progression at different CRC stages.
Collapse
Affiliation(s)
- Linghui Jia
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Liting Liao
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Yongshuai Jiang
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Xiangyu Hu
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Guotao Lu
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou, 225001, P. R. China
| | - Weiming Xiao
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou, 225001, P. R. China
| | - Weijuan Gong
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, P. R. China
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou, 225001, P. R. China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou, 225001, P. R. China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225001, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225001, P. R. China
| | - Xiaoqin Jia
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, P. R. China.
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou, 225001, P. R. China.
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou, 225001, P. R. China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225001, P. R. China.
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225001, P. R. China.
| |
Collapse
|
188
|
Chen W, Zou F, Song T, Xia Y, Xing J, Rao T, Zhou X, Ning J, Zhao S, Yu W, Cheng F. Comprehensive analysis reveals XCL2 as a cancer prognosis and immune infiltration-related biomarker. Aging (Albany NY) 2023; 15:11891-11917. [PMID: 37905956 PMCID: PMC10683633 DOI: 10.18632/aging.205156] [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: 05/15/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND X-C Motif Chemokine Ligand 2 (XCL2) is a 114 amino acid, structurally conserved chemokine involved in activating cytotoxic T cells. However, the pathophysiological mechanisms of XCL2 protein in various disease conditions, particularly cancer, remain poorly understood. METHODS Bioinformatics was used to detect the expression of XCL2, the relationship between survival time and XCL2 in BLCA patients, the mutational status of XCL2, the role of XCL2 in the tumor immune microenvironment, and the sensitivity of XCL2-targeted drugs in 33 cancers. In vitro experiments were conducted to investigate the chemotactic effects of XCL2 expression on M1-type macrophages in human specimens and in isolated cancer cells. RESULTS XCL2 expression was downregulated in tumor tissues and closely associated with the prognosis of human cancers. Furthermore, XCL2 affects DNA methylation, tumor mutation burden (TMB), microsatellite instability (MSI), and mismatch repair (MMR) in human cancers. The expression level of XCL2 significantly correlated with infiltrated immune cells, immunological pathways, and other immune markers. More importantly, we found that XCL2 was positively associated with T lymphocytes and macrophages in the transcriptome and single-cell sequencing data. Using multiple immunofluorescence staining, we found that the expression level of XCL2 was upregulated in many cells in pan-cancer samples, and the number of M1 macrophage marker CD68 and INOS-positive cells increased. 786O, U251, and MDA-MB-231 cells could recruit more M1 macrophages in vitro after overexpressing XCL2. CONCLUSIONS Our results reveal that XCL2 could act as a vital chemokine in pan-cancer and provide new targets and concepts for cancer treatment.
Collapse
Affiliation(s)
- Wu Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Zou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianbao Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuqi Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ji Xing
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jinzhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sheng Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
189
|
Chen Y, Wu B, Shang H, Sun Y, Tian H, Yang H, Wang C, Wang X, Cheng W. Sono-Immunotherapy Mediated Controllable Composite Nano Fluorescent Probes Reprogram the Immune Microenvironment of Hepatocellular Carcinoma. Int J Nanomedicine 2023; 18:6059-6073. [PMID: 37908671 PMCID: PMC10615103 DOI: 10.2147/ijn.s426297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/18/2023] [Indexed: 11/02/2023] Open
Abstract
Background Despite the clinical efficacy of immunotherapy in treating malignant tumors, its effectiveness is often hampered by the immunosuppressive nature of the tumor microenvironment (TME). In this study, we propose the design of a nanoscale ultrasound contrast agent capable of triggering macrophage polarization and immunogenic cell death (ICD) for the treatment of hepatocellular carcinoma (HCC) through sonodynamic treatment (SDT) and immunotherapy. Methods The re-educator (designated as ICG@C3F8-R848 NBs) is composed of the Toll-like receptor agonist resiquimod (R848) and the sonosensitizer Indocyanine green (ICG), utilizing nanobubbles (NBs) as carriers. The technique known as ultrasound-targeted nanobubble destruction (UTND) employs nanosized microbubbles and low-frequency ultrasound (LFUS) to ensure accurate drug delivery and enhance safety. Results Following intravenous delivery, ICG@C3F8-R848 NBs have the potential to selectively target and treat primary tumors using SDT in conjunction with ultrasonography. Importantly, R848 can enhance antitumor immunity by inducing the polarization of macrophages from an M2 to an M1 phenotype. Conclusion The SDT-initiated immunotherapy utilizing ICG@C3F8-R848 NBs demonstrates significant tumor suppression effects with minimal risk of systemic toxicity. The utilization of this self-delivery re-education technique would contribute to advancing the development of nanomedicine for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yichi Chen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin, 150081, People’s Republic of China
| | - Bolin Wu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin, 150081, People’s Republic of China
| | - Haitao Shang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin, 150081, People’s Republic of China
| | - Yucao Sun
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin, 150081, People’s Republic of China
| | - Huimin Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin, 150081, People’s Republic of China
| | - Huajing Yang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin, 150081, People’s Republic of China
| | - Chunyue Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin, 150081, People’s Republic of China
| | - Xiaodong Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin, 150081, People’s Republic of China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People’s Republic of China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin, 150081, People’s Republic of China
| |
Collapse
|
190
|
Tang Q, Zhang F, Luo L, Duan Y, Zhu T, Ni Y, Wang Y, Qi H, Jiang S, Zhou J, Ma X, Zhang Y. Ultrasound-Induced Gold Nanoparticle United with Acoustic Reprogramming of Macrophages for Enhanced Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50926-50939. [PMID: 37877885 DOI: 10.1021/acsami.3c12599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Sonodynamic therapy (SDT) has considerable potential in cancer treatment and exhibits high tissue penetration with minimal damage to healthy tissues. The efficiency of SDT is constrained by the complex immunological environment and tumor treatment resistance. Herein, a specific acoustic-actuated tumor-targeted nanomachine is proposed to generate mechanical damage to lysosomes for cancer SDT. The hybrid nanomachine was assembled with gold nanoparticles (GNPs) as the core and encapsulated with macrophage exosomes modified by AS1411 aptamers (GNP@EXO-APs) to optimize the pharmacokinetics and tumor aggregation. GNP@EXO-APs could be specifically transferred to the lysosomes of tumor cells. After induction with ultrasound, GNP@EXO-APs generated strong mechanical stress to produce lysosomal-dependent cell death in cancer cells. Notably, tumor-associated macrophages were reprogrammed in the ultrasound environment to an antitumor phenotype. Enhanced mechanical destruction via GNP@EXO-APs and immunotherapy of cancer cells were verified both in vitro and in vivo under SDT. This study provides a new direction for inside-out killing effects on tumor cells for cancer treatment.
Collapse
Affiliation(s)
- Qinchao Tang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Fanyu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Licheng Luo
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education and School of Physics and Technology, Wuhan University, Wuhan 430079, China
| | - Yiling Duan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Taomin Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Yueqi Ni
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Yang Wang
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education and School of Physics and Technology, Wuhan University, Wuhan 430079, China
| | - Haoning Qi
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Shuting Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Jingxuan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Xiaoxin Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| |
Collapse
|
191
|
Li H, Guo H, Huang Q, Wang S, Li X, Qiu M. Circular RNA P4HB promotes glycolysis and tumor progression by binding with PKM2 in lung adenocarcinoma. Respir Res 2023; 24:252. [PMID: 37880717 PMCID: PMC10601333 DOI: 10.1186/s12931-023-02563-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Emerging evidence indicates that circular RNAs (circRNAs) play vital roles in tumor progression, including lung adenocarcinomas (LUAD). However, the mechanisms by which circRNAs promote the progression of LUAD still require further investigation. METHODS Quantitative real-time PCR was performed to detect the expression of circP4HB in LUAD tissues and cells. Then, Kaplan-Meier analysis was used to determine the prognostic value of circP4HB expression. We employed RNA pull-down, RNA immunoprecipitation, mass spectrometry, cells fraction, glucose consumption, lactate production, pyruvate kinase M2 (PKM2) activity, and macrophage polarization assays to uncover the underlying mechanisms of circP4HB in LUAD. RESULTS We found that circP4HB is upregulated in LUAD tissues and correlated with advanced TNM stages and lymph node metastasis. LUAD patients with high circP4HB expression had poor prognoses. Functionally, circP4HB promoted LUAD progression in vivo and in vitro. Upregulated circP4HB increased glucose consumption, lactate production and accelerated aerobic glycolysis in LUAD cells. Mechanically, circP4HB mainly accumulated in the cytoplasm of LUAD cells and bound with PKM2 and subsequently upregulating PKM2 enzymatic activity by increasing its tetramer formation. Additionally, circP4HB promoted M2 macrophage phenotype shift via targeting PKM2. Finally, rescue assays further confirmed that circP4HB could promote LUAD cell progression through its interaction with PKM2. CONCLUSION These results demonstrate that circP4HB could promote LUAD progression, indicating circP4HB might be a potential therapeutic target of LUAD.
Collapse
Affiliation(s)
- Haoran Li
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
| | - Haifa Guo
- The First Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Qi Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Shaodong Wang
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
| | - Xiao Li
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
| | - Mantang Qiu
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, China.
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
192
|
Tian L, Li H, Zhao P, Liu Y, Lu Y, Zhong R, Jin Y, Tan T, Cheng Y. C-Myc-induced hypersialylation of small cell lung cancer facilitates pro-tumoral phenotypes of macrophages. iScience 2023; 26:107771. [PMID: 37731607 PMCID: PMC10507237 DOI: 10.1016/j.isci.2023.107771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/03/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
Immunosuppressive myeloid cell populations have been documented in small cell lung cancer (SCLC) subtypes, playing a key role in remolding the tumor microenvironment (TME). However, the cancer-associated transcriptional features of monocytes and tumor-associated macrophages (TAMs) in SCLC remain poorly understood. Herein, we analyzed the molecular features and functions of monocyte/macrophage subsets aiming to inhibit monocyte recruitment and pro-tumor behavior of macrophages. We observe that NEUROD1-high SCLC subtype (SCLC-N) exhibits subtype-specific hypersialylation induced by the unique target c-Myc (MYC) of NEUROD1. The hypersialylation can alter macrophage phenotypes and pro-tumor behavior by regulating the expression of the immune-inhibiting lectin receptors on monocyte-derived macrophages (MDMs) in SCLC-N. Inhibiting the aberrant sialic acid metabolic pathways in SCLC can significantly enhance the phagocytosis of macrophages. This study provides a comprehensive overview of the cancer-specific immune signature of monocytes and macrophages and reveals tumor-associated biomarkers as potential therapeutic targets for SCLC.
Collapse
Affiliation(s)
- Lin Tian
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun 130012, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun 130012, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun 130012, China
| | - Hui Li
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun 130012, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun 130012, China
| | - Peiyan Zhao
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun 130012, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun 130012, China
| | - Yan Liu
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun 130012, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun 130012, China
| | - Yuanhua Lu
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun 130012, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun 130012, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun 130012, China
| | - Rui Zhong
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun 130012, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun 130012, China
| | - Yulong Jin
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun 130012, China
| | - Tianyu Tan
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and The First Affliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ying Cheng
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun 130012, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun 130012, China
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun 130012, China
| |
Collapse
|
193
|
Huang M, Xia Y, Li K, Shao F, Feng Z, Li T, Azin M, Demehri S. Carcinogen exposure enhances cancer immunogenicity by blocking the development of an immunosuppressive tumor microenvironment. J Clin Invest 2023; 133:e166494. [PMID: 37843274 PMCID: PMC10575722 DOI: 10.1172/jci166494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 08/15/2023] [Indexed: 10/17/2023] Open
Abstract
Carcinogen exposure is strongly associated with enhanced cancer immunogenicity. Increased tumor mutational burden and resulting neoantigen generation have been proposed to link carcinogen exposure and cancer immunogenicity. However, the neoantigen-independent immunological impact of carcinogen exposure on cancer is unknown. Here, we demonstrate that chemical carcinogen-exposed cancer cells fail to establish an immunosuppressive tumor microenvironment (TME), resulting in their T cell-mediated rejection in vivo. A chemical carcinogen-treated breast cancer cell clone that lacked any additional coding region mutations (i.e., neoantigen) was rejected in mice in a T cell-dependent manner. Strikingly, the coinjection of carcinogen- and control-treated cancer cells prevented this rejection, suggesting that the loss of immunosuppressive TME was the dominant cause of rejection. Reduced M-CSF expression by carcinogen-treated cancer cells significantly suppressed tumor-associated macrophages (TAMs) and resulted in the loss of an immunosuppressive TME. Single-cell analysis of human lung cancers revealed a significant reduction in the immunosuppressive TAMs in former smokers compared with individuals who had never smoked. These findings demonstrate that carcinogen exposure impairs the development of an immunosuppressive TME and indicate a novel link between carcinogens and cancer immunogenicity.
Collapse
Affiliation(s)
- Mei Huang
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yun Xia
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kaiwen Li
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Feng Shao
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhaoyi Feng
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tiancheng Li
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Marjan Azin
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
194
|
Singh N, Maus MV. Synthetic manipulation of the cancer-immunity cycle: CAR-T cell therapy. Immunity 2023; 56:2296-2310. [PMID: 37820585 DOI: 10.1016/j.immuni.2023.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023]
Abstract
Synthetic immunity to cancer has been pioneered by the application of chimeric antigen receptor (CAR) engineering into autologous T cells. CAR T cell therapy is highly amenable to molecular engineering to bypass barriers of the cancer immunity cycle, such as endogenous antigen presentation, immune priming, and natural checkpoints that constrain immune responses. Here, we review CAR T cell design and the mechanisms that drive sustained CAR T cell effector activity and anti-tumor function. We discuss engineering approaches aimed at improving anti-tumor function through a variety of mechanistic interventions for both hematologic and solid tumors. The ability to engineer T cells in such a variety of ways, including by modifying their trafficking, antigen recognition, costimulation, and addition of synthetic genes, circuits, knockouts and base edits to finely tune complex functions, is arguably the most powerful way to manipulate the cancer immunity cycle in patients.
Collapse
Affiliation(s)
- Nathan Singh
- Division of Oncology, Washington University in St Louis School of Medicine, St. Louis, MO 63110, USA.
| | - Marcela V Maus
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
195
|
Liu Y, Liu N, Zhou X, Zhao L, Wei W, Hu J, Luo Z. Constructing a prognostic model for head and neck squamous cell carcinoma based on glucose metabolism related genes. Front Endocrinol (Lausanne) 2023; 14:1245629. [PMID: 37876534 PMCID: PMC10591078 DOI: 10.3389/fendo.2023.1245629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Background Glucose metabolism (GM) plays a crucial role in cancer cell proliferation, tumor growth, and survival. However, the identification of glucose metabolism-related genes (GMRGs) for effective prediction of prognosis in head and neck squamous cell carcinoma (HNSC) is still lacking. Methods We conducted differential analysis between HNSC and Normal groups to identify differentially expressed genes (DEGs). Key module genes were obtained using weighted gene co-expression network analysis (WGCNA). Intersection analysis of DEGs, GMRGs, and key module genes identified GMRG-DEGs. Univariate and multivariate Cox regression analyses were performed to screen prognostic-associated genes. Independent prognostic analysis of clinical traits and risk scores was implemented using Cox regression. Gene set enrichment analysis (GSEA) was used to explore functional pathways and genes between high- and low-risk groups. Immune infiltration analysis compared immune cells between the two groups in HNSC samples. Drug prediction was performed using the Genomics of Drug Sensitivity in Cancer (GDSC) database. Quantitative real-time fluorescence PCR (qRT-PCR) validated the expression levels of prognosis-related genes in HNSC patients. Results We identified 4973 DEGs between HNSC and Normal samples. Key gene modules, represented by black and brown module genes, were identified. Intersection analysis revealed 76 GMRG-DEGs. Five prognosis-related genes (MTHFD2, CDKN2A, TPM2, MPZ, and DNMT1) were identified. A nomogram incorporating age, lymph node status (N), and risk score was constructed for survival prediction in HNSC patients. Immune infiltration analysis showed significant differences in five immune cell types (Macrophages M0, memory B cells, Monocytes, Macrophages M2, and Dendritic resting cells) between the high- and low-risk groups. GDSC database analysis identified 53 drugs with remarkable differences between the groups, including A.443654 and AG.014699. DNMT1 and MTHFD2 were up-regulated, while MPZ was down-regulated in HNSC. Conclusion Our study highlights the significant association of five prognosis-related genes (MTHFD2, CDKN2A, TPM2, MPZ, and DNMT1) with HNSC. These findings provide further evidence of the crucial role of GMRGs in HNSC.
Collapse
Affiliation(s)
- Yu Liu
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Nana Liu
- Department of Onclogy, People’s Hospital of Chongqing Hechuan, Chongqing, China
| | - Xue Zhou
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lingqiong Zhao
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Wei Wei
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Jie Hu
- Department of Otolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, China
| | - Zhibin Luo
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| |
Collapse
|
196
|
Malainou C, Abdin SM, Lachmann N, Matt U, Herold S. Alveolar macrophages in tissue homeostasis, inflammation, and infection: evolving concepts of therapeutic targeting. J Clin Invest 2023; 133:e170501. [PMID: 37781922 PMCID: PMC10541196 DOI: 10.1172/jci170501] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
Alveolar macrophages (AMs) are the sentinel cells of the alveolar space, maintaining homeostasis, fending off pathogens, and controlling lung inflammation. During acute lung injury, AMs orchestrate the initiation and resolution of inflammation in order to ultimately restore homeostasis. This central role in acute lung inflammation makes AMs attractive targets for therapeutic interventions. Single-cell RNA-Seq and spatial omics approaches, together with methodological advances such as the generation of human macrophages from pluripotent stem cells, have increased understanding of the ontogeny, function, and plasticity of AMs during infectious and sterile lung inflammation, which could move the field closer to clinical application. However, proresolution phenotypes might conflict with proinflammatory and antibacterial responses. Therefore, therapeutic targeting of AMs at vulnerable time points over the course of infectious lung injury might harbor the risk of serious side effects, such as loss of antibacterial host defense capacity. Thus, the identification of key signaling hubs that determine functional fate decisions in AMs is of the utmost importance to harness their therapeutic potential.
Collapse
Affiliation(s)
- Christina Malainou
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| | - Shifaa M. Abdin
- German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology and
- REBIRTH Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Nico Lachmann
- German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology and
- REBIRTH Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- RESIST (Resolving Infection Susceptibility), Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Ulrich Matt
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| | - Susanne Herold
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
197
|
Duan Z, Li Z, Wang Z, Chen C, Luo Y. Chimeric antigen receptor macrophages activated through TLR4 or IFN-γ receptors suppress breast cancer growth by targeting VEGFR2. Cancer Immunol Immunother 2023; 72:3243-3257. [PMID: 37438548 PMCID: PMC10992605 DOI: 10.1007/s00262-023-03490-8] [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: 01/22/2023] [Accepted: 06/28/2023] [Indexed: 07/14/2023]
Abstract
Chimeric antigen receptor macrophage (CAR-M) is a promising immunotherapy strategy of anti-tumor due to its high infiltration, direct phagocytosis of tumor cells, immunomodulation of tumor microenvironment (TME) and linkage of innate and adaptive immunity. Here a series of novelly designed CAR-Ms by targeting vascular endothelial growth factor receptor-2 (VEGFR2), which highly expressed in tumor cells and TME, were evaluated. Their activation signals were transduced by Tlr4 or Ifn-γ receptors either alone or in combination, which were designed to mediate M1 polarization of macrophages as the downstream of lipopolysaccharide or Ifn-γ that had been widely reported. Our results showed that VEGFR2-targeting CAR-Ms could be activated under the stimulation of VEGFR2-expressing cells. They exhibited higher expression of CD86, MHCII and TNF-α in vitro and enhanced tumor suppressive abilities in vivo. Implantation of these CAR-Ms into 4T1 breast cancer-bearing mice could obviously inhibit the progression of tumor without significant toxic side effects, especially the group of mmC in which constructed with Tlr4 as the intracellular domain of CAR. In conclusion, this research provides a promising design of CAR that induce macrophages activation by Tlr4 and/or Ifn-γ receptors, and these CAR-Ms could effectively inhibit tumor growth through targeting VEGFR2.
Collapse
Affiliation(s)
- Zhaojun Duan
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Zhen Li
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Ziyuan Wang
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Chong Chen
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Yunping Luo
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
198
|
Zheng J, Bu X, Wei X, Ma X, Zhao P. The role of FoxM1 in immune cells. Clin Exp Med 2023; 23:1973-1979. [PMID: 36913035 DOI: 10.1007/s10238-023-01037-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/01/2023] [Indexed: 03/14/2023]
Abstract
Forkhead box M1 (FoxM1), a proliferation specific transcriptional modulator, plays a principal role in many physiological and pathological processes. FoxM1-mediated oncogenic processes have been well addressed. However, functions of FoxM1 in immune cells are less summarized. The literatures about the expression of FoxM1 and its regulation on immune cells were searched on PubMed and Google Scholar. In this review, we provide an overview on the roles of FoxM1 in regulating functions of immune cells, including T cells, B cells, monocytes, macrophages, and dendritic cells, and discuss their contributions to diseases.
Collapse
Affiliation(s)
- Jinju Zheng
- Biotherapy Center, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Xiaocui Bu
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| | - Xiaofang Wei
- Biotherapy Center, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Xuezhen Ma
- Department of Oncology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China.
| | - Peng Zhao
- Biotherapy Center, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
199
|
Wang Q, Zhi Y, Zi M, Mo Y, Wang Y, Liao Q, Zhang S, Gong Z, Wang F, Zeng Z, Guo C, Xiong W. Spatially Resolved Transcriptomics Technology Facilitates Cancer Research. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302558. [PMID: 37632718 PMCID: PMC10602551 DOI: 10.1002/advs.202302558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/16/2023] [Indexed: 08/28/2023]
Abstract
Single cell RNA sequencing (scRNA-seq) provides a great convenience for studying tumor occurrence and development for its ability to study gene expression at the individual cell level. However, patient-derived tumor tissues are composed of multiple types of cells including tumor cells and adjacent non-malignant cells such as stromal cells and immune cells. The spatial locations of various cells in situ tissues plays a pivotal role in the occurrence and development of tumors, which cannot be elucidated by scRNA-seq alone. Spatially resolved transcriptomics (SRT) technology emerges timely to explore the unrecognized relationship between the spatial background of a particular cell and its functions, and is increasingly used in cancer research. This review provides a systematic overview of the SRT technologies that are developed, in particular the more widely used cutting-edge SRT technologies based on next-generation sequencing (NGS). In addition, the main achievements by SRT technologies in precisely unveiling the underappreciated spatial locations on gene expression and cell function with unprecedented high-resolution in cancer research are emphasized, with the aim of developing more effective clinical therapeutics oriented to a deeper understanding of the interaction between tumor cells and surrounding non-malignant cells.
Collapse
Affiliation(s)
- Qian Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410008P. R. China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research InstituteCentral South UniversityChangshaHunan410008P. R. China
| | - Yuan Zhi
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410012P. R. China
| | - Moxin Zi
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410012P. R. China
| | - Yongzhen Mo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research InstituteCentral South UniversityChangshaHunan410008P. R. China
- Department of Otolaryngology Head and Neck SurgeryXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Yumin Wang
- Department of Otolaryngology Head and Neck SurgeryXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410008P. R. China
| | - Shanshan Zhang
- Department of Otolaryngology Head and Neck SurgeryXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410012P. R. China
| | - Fuyan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research InstituteCentral South UniversityChangshaHunan410008P. R. China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410008P. R. China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research InstituteCentral South UniversityChangshaHunan410008P. R. China
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410008P. R. China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research InstituteCentral South UniversityChangshaHunan410008P. R. China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410008P. R. China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research InstituteCentral South UniversityChangshaHunan410008P. R. China
| |
Collapse
|
200
|
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
|