1
|
Liu J, Lu J, Wu L, Zhang T, Wu J, Li L, Tai Z, Chen Z, Zhu Q. Targeting tumor-associated macrophages: Novel insights into immunotherapy of skin cancer. J Adv Res 2025; 67:231-252. [PMID: 38242529 PMCID: PMC11725115 DOI: 10.1016/j.jare.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/19/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND The incidence of skin cancer is currently increasing, and conventional treatment options inadequately address the demands of disease management. Fortunately, the recent rapid advancement of immunotherapy, particularly immune checkpoint inhibitors (ICIs), has ushered in a new era for numerous cancer patients. However, the efficacy of immunotherapy remains suboptimal due to the impact of the tumor microenvironment (TME). Tumor-associated macrophages (TAMs), a major component of the TME, play crucial roles in tumor invasion, metastasis, angiogenesis, and immune evasion, significantly impacting tumor development. Consequently, TAMs have gained considerable attention in recent years, and their roles have been extensively studied in various tumors. However, the specific roles of TAMs and their regulatory mechanisms in skin cancer remain unclear. AIM OF REVIEW This paper aims to elucidate the origin and classification of TAMs, investigate the interactions between TAMs and various immune cells, comprehensively understand the precise mechanisms by which TAMs contribute to the pathogenesis of different types of skin cancer, and finally discuss current strategies for targeting TAMs in the treatment of skin cancer. KEY SCIENTIFIC CONCEPTS OF OVERVIEW With a specific emphasis on the interrelationship between TAMs and skin cancer, this paper posits that therapeutic modalities centered on TAMs hold promise in augmenting and harmonizing with prevailing clinical interventions for skin cancer, thereby charting a novel trajectory for advancing the landscape of immunotherapeutic approaches for skin cancer.
Collapse
Affiliation(s)
- Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Jiaye Lu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Ling Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Tingrui Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Junchao Wu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Lisha Li
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China.
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China.
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China.
| |
Collapse
|
2
|
Li R, Huang J, Wei Y, Wang Y, Lu C, Liu J, Ma X. Nanotherapeutics for Macrophage Network Modulation in Tumor Microenvironments: Targets and Tools. Int J Nanomedicine 2024; 19:13615-13651. [PMID: 39717515 PMCID: PMC11665441 DOI: 10.2147/ijn.s491573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/04/2024] [Indexed: 12/25/2024] Open
Abstract
Macrophage is an important component in the tumor immune microenvironment, which exerts significant influence on tumor development and metastasis. Due to their dual nature of promoting and suppressing inflammation, macrophages can serve as both targets for tumor immunotherapy and tools for treating malignancies. However, the abundant infiltration of tumor-associated macrophages dominated by an immunosuppressive phenotype maintains a pro-tumor microenvironment, and engineering macrophages using nanotechnology to manipulate the tumor immune microenvironment represent a feasible approach for cancer immunotherapy. Additionally, considering the phagocytic and specifically tumor-targeting capabilities of M1 macrophages, macrophages manipulated through cellular engineering and nanotechnology, as well as macrophage-derived exosomes and macrophage membranes, can also become effective tools for cancer treatment. In conclusion, nanotherapeutics targeting macrophages remains immense potential for the development of macrophage-mediated tumor treatment methods and will further enhance our understanding, diagnosis, and treatment of various malignants.
Collapse
Affiliation(s)
- Renwei Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Jing Huang
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Yuhao Wei
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Yusha Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Can Lu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Jifeng Liu
- Department of Otolaryngology Head and Neck Surgery/Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- State Key Laboratory of Intelligent Construction and Healthy Operation and Maintenance of Deep Underground Engineering, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| |
Collapse
|
3
|
Wang B, Cao X, Garcia-Mansfield K, Zhou J, Manousopoulou A, Pirrotte P, Wang Y, Wang LD, Feng M. Phosphoproteomic Profiling Reveals mTOR Signaling in Sustaining Macrophage Phagocytosis of Cancer Cells. Cancers (Basel) 2024; 16:4238. [PMID: 39766137 PMCID: PMC11674635 DOI: 10.3390/cancers16244238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Macrophage-mediated cancer cell phagocytosis has demonstrated considerable therapeutic potential. While the initiation of phagocytosis, facilitated by interactions between cancer cell surface signals and macrophage receptors, has been characterized, the mechanisms underlying its sustentation and attenuation post-initiation remain poorly understood. Methods: Through comprehensive phosphoproteomic profiling, we interrogated the temporal evolution of the phosphorylation profiles within macrophages during cancer cell phagocytosis. Results: Our findings reveal that activation of the mTOR pathway occurs following the initiation of phagocytosis and is crucial in sustaining phagocytosis of cancer cells. mTOR inhibition impaired the phagocytic capacity, but not affinity, of the macrophages toward the cancer cells by delaying phagosome maturation and impeding the transition between non-phagocytic and phagocytic states of macrophages. Conclusions: Our findings delineate the intricate landscape of macrophage phagocytosis and highlight the pivotal role of the mTOR pathway in mediating this process, offering valuable mechanistic insights for therapeutic interventions.
Collapse
Affiliation(s)
- Bixin Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Krystine Garcia-Mansfield
- Cancer and Cell Biology Division, Translational Genomics Institute, Phoenix, AZ 85004, USA
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Jingkai Zhou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Antigoni Manousopoulou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Patrick Pirrotte
- Cancer and Cell Biology Division, Translational Genomics Institute, Phoenix, AZ 85004, USA
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Yingyu Wang
- Center for Informatics, City of Hope, Duarte, CA 91010, USA
| | - Leo D. Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
- Department of Pediatrics, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
4
|
Ma Y, Sun Y, Guo H, Yang R. Tumor-associated macrophages in bladder cancer: roles and targeted therapeutic strategies. Front Immunol 2024; 15:1418131. [PMID: 39606239 PMCID: PMC11599180 DOI: 10.3389/fimmu.2024.1418131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Bladder cancer (BC) is the ninth most common and "expensive" cancer in the world. Despite the availability of various treatment modalities such as chemotherapy, immunotherapy and surgery, the overall survival rate of patients with advanced bladder cancer remains low. As one of the most abundant infiltrating immune cells in bladder cancer, tumor-associated macrophages (TAMs) play an important role in the development of BC and in the standard regimen of intravesical BCG therapy. Targeting TAMs have achieved excellent results in clinical trials for a variety of other cancers, but few studies have been conducted for bladder cancer. Further exploration is still needed to develop TAM-related therapeutic strategies for BC treatment, which are expected to improve the therapeutic efficacy and life quality of patients. This review summarizes the relationship between TAMs in bladder cancer and disease staging, evolution, patient prognosis, and treatment outcome. Several potential TAM targets in BC are also pointed, which may help to inhibit tumor-promoting TAMs and provide new therapeutic approaches for advanced BC.
Collapse
Affiliation(s)
- Yuanchun Ma
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ying Sun
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Urology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University Advanced Institute of Life Sciences (NAILS), Nanjing, China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
5
|
Santerre JP, Yang Y, Du Z, Wang W, Zhang X. Biomaterials' enhancement of immunotherapy for breast cancer by targeting functional cells in the tumor micro-environment. Front Immunol 2024; 15:1492323. [PMID: 39600709 PMCID: PMC11588700 DOI: 10.3389/fimmu.2024.1492323] [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: 09/06/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Immunotherapy for breast cancer is now being considered clinically, and more recently, the number of investigations aimed specifically at nano-biomaterials-assisted immunotherapy for breast cancer treatment is growing. Alterations of the breast cancer micro-environment can play a critical role in anti-tumor immunity and cancer development, progression and metastasis. The improvement and rearrangement of tumor micro-environment (TME) may enhance the permeability of anti-tumor drugs. Therefore, targeting the TME is also an ideal and promising option during the selection of effective nano-biomaterial-based immuno-therapeutic strategies excepted for targeting intrinsic resistant mechanisms of the breast tumor. Although nano-biomaterials designed to specifically release loaded anti-tumor drugs in response to tumor hypoxia and low pH conditions have shown promises and the diversity of the TME components also supports a broad targeting potential for anti-tumor drug designs, yet the applications of nano-biomaterials for targeting immunosuppressive cells/immune cells in the TME for improving the breast cancer treating outcomes, have scarcely been addressed in a scientific review. This review provides a thorough discussion for the application of the different forms of nano-biomaterials, as carrier vehicles for breast cancer immunotherapy, targeting specific types of immune cells in the breast tumor microenvironment. In parallel, the paper provides a critical analysis of current advances/challenges with leading nano-biomaterial-mediated breast cancer immunotherapeutic strategies. The current review is timely and important to the cancer research field and will provide a critical tool for nano-biomaterial design and research groups pushing the clinical translation of new nano-biomaterial-based immuno-strategies targeting breast cancer TME, to further open new avenues for the understanding, prevention, diagnosis and treatment of breast cancer, as well as other cancer types.
Collapse
Affiliation(s)
- J. Paul Santerre
- The School of Basic Medicine, Binzhou Medical University, Yantai, Shandong, China
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Yangyang Yang
- The School of Basic Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Ziwei Du
- The School of Basic Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Wenshuang Wang
- Department of Gynecology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Xiaoqing Zhang
- The School of Basic Medicine, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
6
|
Wang Y, Shi J, Xin M, Kahkoska AR, Wang J, Gu Z. Cell-drug conjugates. Nat Biomed Eng 2024; 8:1347-1365. [PMID: 38951139 PMCID: PMC11646559 DOI: 10.1038/s41551-024-01230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/01/2024] [Indexed: 07/03/2024]
Abstract
By combining living cells with therapeutics, cell-drug conjugates can potentiate the functions of both components, particularly for applications in drug delivery and therapy. The conjugates can be designed to persist in the bloodstream, undergo chemotaxis, evade surveillance by the immune system, proliferate, or maintain or transform their cellular phenotypes. In this Review, we discuss strategies for the design of cell-drug conjugates with specific functions, the techniques for their preparation, and their applications in the treatment of cancers, autoimmune diseases and other pathologies. We also discuss the translational challenges and opportunities of this class of drug-delivery systems and therapeutics.
Collapse
Affiliation(s)
- Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jiaqi Shi
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Minhang Xin
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Anna R Kahkoska
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
7
|
Patni H, Chaudhary R, Kumar A. Unleashing nanotechnology to redefine tumor-associated macrophage dynamics and non-coding RNA crosstalk in breast cancer. NANOSCALE 2024; 16:18274-18294. [PMID: 39292162 DOI: 10.1039/d4nr02795g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Breast cancer is a significant global health issue. Tumor-associated macrophages (TAMs) are crucial in influencing the tumor microenvironment and the progression of the disease. TAMs exhibit remarkable plasticity in adopting distinct phenotypes ranging from pro-inflammatory and anti-tumorigenic (M1-like) to immunosuppressive and tumor-promoting (M2-like). This review elucidates the multifaceted roles of TAMs in driving breast tumor growth, angiogenesis, invasion, and metastatic dissemination. Significantly, it highlights the intricate crosstalk between TAMs and non-coding RNAs (ncRNAs), including microRNAs, long noncoding RNAs, and circular RNAs, as a crucial regulatory mechanism modulating TAM polarization and functional dynamics that present potential therapeutic targets. Nanotechnology-based strategies are explored as a promising approach to reprogramming TAMs toward an anti-tumor phenotype. Various nanoparticle delivery systems have shown potential for modulating TAM polarization and inhibiting tumor-promoting effects. Notably, nanoparticles can deliver ncRNA therapeutics to TAMs, offering unique opportunities to modulate their polarization and activity.
Collapse
Affiliation(s)
- Hardik Patni
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Ramesh Chaudhary
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| |
Collapse
|
8
|
Wang R, Kumar P, Reda M, Wallstrum AG, Crumrine NA, Ngamcherdtrakul W, Yantasee W. Nanotechnology Applications in Breast Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308639. [PMID: 38126905 PMCID: PMC11493329 DOI: 10.1002/smll.202308639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Next-generation cancer treatments are expected not only to target cancer cells but also to simultaneously train immune cells to combat cancer while modulating the immune-suppressive environment of tumors and hosts to ensure a robust and lasting response. Achieving this requires carriers that can codeliver multiple therapeutics to the right cancer and/or immune cells while ensuring patient safety. Nanotechnology holds great potential for addressing these challenges. This article highlights the recent advances in nanoimmunotherapeutic development, with a focus on breast cancer. While immune checkpoint inhibitors (ICIs) have achieved remarkable success and lead to cures in some cancers, their response rate in breast cancer is low. The poor response rate in solid tumors is often associated with the low infiltration of anti-cancer T cells and an immunosuppressive tumor microenvironment (TME). To enhance anti-cancer T-cell responses, nanoparticles are employed to deliver ICIs, bispecific antibodies, cytokines, and agents that induce immunogenic cancer cell death (ICD). Additionally, nanoparticles are used to manipulate various components of the TME, such as immunosuppressive myeloid cells, macrophages, dendritic cells, and fibroblasts to improve T-cell activities. Finally, this article discusses the outlook, challenges, and future directions of nanoimmunotherapeutics.
Collapse
Affiliation(s)
- Ruijie Wang
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Pramod Kumar
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Moataz Reda
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Noah A. Crumrine
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| |
Collapse
|
9
|
Fu Z, Feng M, Wu J, Liu B, Fu J, Song W. Photodynamic Therapy Synergizes CD47 Blockade Strategy for Enhanced Antitumor Therapy. Mol Pharm 2024; 21:3897-3908. [PMID: 38959154 DOI: 10.1021/acs.molpharmaceut.4c00254] [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] [Indexed: 07/05/2024]
Abstract
The antitumor strategies based on innate immunity activation have become favored by researchers in recent years. In particular, strategies targeting antiphagocytic signaling blockade to enhance phagocytosis have been widely reported. For example, the addition of prophagocytic signals such as calreticulin could make the strategy significantly more effective. In this study, an antitumor strategy that combines photodynamic therapy (PDT) with CD47 blockade has been reported. This approach promotes the maturation of dendritic cells and the presentation of tumor antigens by PDT-mediated tumor immunogenic cell death, as well as the enhancement of cytotoxic T lymphocyte infiltration in tumor areas and the phagocytic activity of phagocytes. Furthermore, the downregulation and blockage of CD47 protein could further promote phagocytic activity, strengthen the innate immune system, and ultimately elevate the antitumor efficacy and inhibit tumor metastasis.
Collapse
Affiliation(s)
- Zhaoming Fu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Minghui Feng
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Jinxian Wu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Bo Liu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Jiajia Fu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Wen Song
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| |
Collapse
|
10
|
Liu W, Zhou H, Lai W, Hu C, Xu R, Gu P, Luo M, Zhang R, Li G. The immunosuppressive landscape in tumor microenvironment. Immunol Res 2024; 72:566-582. [PMID: 38691319 DOI: 10.1007/s12026-024-09483-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
Recent advances in cancer immunotherapy, especially immune checkpoint inhibitors (ICIs), have revolutionized the clinical outcome of many cancer patients. Despite the fact that impressive progress has been made in recent decades, the response rate remains unsatisfactory, and many patients do not benefit from ICIs. Herein, we summarized advanced studies and the latest insights on immune inhibitory factors in the tumor microenvironment. Our in-depth discussion and updated landscape of tumor immunosuppressive microenvironment may provide new strategies for reversing tumor immune evasion, enhancing the efficacy of ICIs therapy, and ultimately achieving a better clinical outcome.
Collapse
Affiliation(s)
- Wuyi Liu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Huyue Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Changpeng Hu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Rufu Xu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Peng Gu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Menglin Luo
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China.
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 83 Xinqiao Road, Shapingba, Chongqing, China.
| |
Collapse
|
11
|
Di L, Li M, Lei X, Xie W, Liu G, Wang Y, Zhang W, Zhu WG. Caspase-4 in glioma indicates deterioration and unfavorable prognosis by affecting tumor cell proliferation and immune cell recruitment. Sci Rep 2024; 14:17443. [PMID: 39075190 PMCID: PMC11286837 DOI: 10.1038/s41598-024-65018-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/16/2024] [Indexed: 07/31/2024] Open
Abstract
Gliomas are the most common malignant tumors of the central nervous system, accounting for approximately 80% of all malignant brain tumors. Accumulating evidence suggest that pyroptosis plays an essential role in the progression of cancer. Unfortunately, the effect of the pyroptosis-related factor caspase-4 (CASP4) on immunotherapy and drug therapy for tumors has not been comprehensively investigated. In this study, we systematically screened six hub genes by pooling differential pyroptosis-related genes in The Cancer Genome Atlas (TCGA) glioma data and the degree of centrality of index-related genes in the protein-protein interaction network. We performed functional and pathway enrichment analyses of the six hub genes to explore their biological functions and potential molecular mechanisms. We then investigated the importance of CASP4 using Kaplan-Meier survival analysis of glioma patients. TCGA and the Chinese Glioma Genome Atlas (CGGA) databases showed that reduced CASP4 expression leads to the potent clinical deterioration of glioma patients. Computational analysis of the effect of CASP4 on the infiltration level and recruitment of glioma immune cells revealed that CASP4 expression was closely associated with a series of tumor-suppressive immune checkpoint molecules, chemokines, and chemokine receptors. We also found that aberrant CASP4 expression correlated with chemotherapeutic drug sensitivity. Finally, analysis at the cellular and tissue levels indicated an increase in CASP4 expression in glioma, and that CASP4 inhibition significantly inhibited the proliferation of glioma cells. Thus, CASP4 is implicated as a new prognostic biomarker for gliomas with the potential to further guide immunotherapy and chemotherapy strategies for glioma patients.
Collapse
Affiliation(s)
- Longjiang Di
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mengyan Li
- Guangdong Key Laboratory of Genomic Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, 518055, China
| | - Xianli Lei
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Faculty of Medicine, Peking University, Beijing, 100191, China
| | - Wenting Xie
- Department of Clinical Laboratory, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Guoqiang Liu
- Department of Clinical Laboratory, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Yongqing Wang
- Division of Rheumatology and Immunology, University of Toledo Medical Center, Toledo, OH, 43614, USA
| | - Wenjing Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.
| | - Wei-Guo Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Key Laboratory of Genomic Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
- College of Basic Medical Sciences, Wan Nan Medical College, Wuhu, 241006, China.
- International Cancer Center, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
12
|
Zhao Y, Ni Q, Zhang W, Yu S. Progress in reeducating tumor-associated macrophages in tumor microenvironment. Discov Oncol 2024; 15:312. [PMID: 39060648 PMCID: PMC11282027 DOI: 10.1007/s12672-024-01186-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
Malignant tumor, one of the most threatening diseases to human health, has been comprehensively treated with surgery, radiotherapy, chemotherapy and targeted therapy, but the prognosis has not always been ideal. In the past decade, immunotherapy has shown increased efficacy in tumor treatment; however, for immunotherapy to achieve its fullest potential, obstacles are to be conquered, among which tumor microenvironment (TME) has been widely investigated. In remodeling the tumor immune microenvironment to inhibit tumor progression, macrophages, as the most abundant innate immune population, play an irreplaceable role in the immune response. Therefore, how to remodel TME and alter the recruitment and polarization status of tumor-associated macrophages (TAM) has been of wide interest. In this context, nanoparticles, photodynamic therapy and other therapeutic approaches capable of affecting macrophage polarization have emerged. In this paper, we categorize and organize the existing means and methods for reprogramming TAM to provide ideas for clinical application of novel tumor-related therapies.
Collapse
Affiliation(s)
- Yiming Zhao
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China
| | - Qianyang Ni
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China
| | - Weijian Zhang
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China
| | - Suyang Yu
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China.
| |
Collapse
|
13
|
Hao J, Zhao X, Wang C, Cao X, Liu Y. Recent Advances in Nanoimmunotherapy by Modulating Tumor-Associated Macrophages for Cancer Therapy. Bioconjug Chem 2024; 35:867-882. [PMID: 38919067 DOI: 10.1021/acs.bioconjchem.4c00242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Cancer immunotherapy has yielded remarkable results across a variety of tumor types. Nevertheless, the complex and immunosuppressive microenvironment within solid tumors poses significant challenges to established therapies such as immune checkpoint blockade (ICB) and chimeric antigen receptor T-cell (CAR-T) therapy. Within the milieu, tumor-associated macrophages (TAMs) play a significant role by directly suppressing T-cell functionality and fostering an immunosuppressive environment. Effective regulation of TAMs is, therefore, crucial to enhancing the efficacy of immunotherapies. Various therapeutic strategies targeting TAM modulation have emerged, including blocking TAM recruitment, direct elimination, promoting repolarization toward the M1 phenotype, and enhancing phagocytic capacity against tumor cells. The recently introduced CAR macrophage (CAR-M) therapy opens new possibilities for macrophage-based immunotherapy. Compared with CAR-T, CAR-M may demonstrate superior targeting and infiltration capabilities toward solid tumors. This review predominantly delves into the origin and development process of TAMs, their role in promoting tumor growth, and provides a comprehensive overview of immunotherapies targeting TAMs. It underscores the significance of regulating TAMs in bolstering antitumor therapies while discussing the potential and challenges of developing TAMs as targets for immunotherapy.
Collapse
Affiliation(s)
- Jialei Hao
- Key Laboratory of Functional Polymer Materials (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xinzhi Zhao
- Key Laboratory of Functional Polymer Materials (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Chun Wang
- Key Laboratory of Functional Polymer Materials (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xianghui Cao
- Key Laboratory of Functional Polymer Materials (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yang Liu
- Key Laboratory of Functional Polymer Materials (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
14
|
Salmaninejad A, Layeghi SM, Falakian Z, Golestani S, Kobravi S, Talebi S, Yousefi M. An update to experimental and clinical aspects of tumor-associated macrophages in cancer development: hopes and pitfalls. Clin Exp Med 2024; 24:156. [PMID: 39003350 PMCID: PMC11246281 DOI: 10.1007/s10238-024-01417-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Tumor-associated macrophages (TAMs) represent one of the most abundant tumor-infiltrating stromal cells, and their normal function in tumor microenvironment (TME) is to suppress tumor cells by producing cytokines which trigger both direct cell cytotoxicity and antibody-mediated immune response. However, upon prolonged exposure to TME, the classical function of these so-called M1-type TAMs can be converted to another type, "M2-type," which are recruited by tumor cells so that they promote tumor growth and metastasis. This is the reason why the accumulation of TAMs in TME is correlated with poor prognosis in cancer patients. Both M1- and M2-types have high degree of plasticity, and M2-type cells can be reprogrammed to M1-type for therapeutic purposes. This characteristic introduces TAMs as promising target for developing novel cancer treatments. In addition, inhibition of M2-type cells and blocking their recruitment in TME, as well as their depletion by inducing apoptosis, are other approaches for effective immunotherapy of cancer. In this review, we summarize the potential of TAMs to be targeted for cancer immunotherapy and provide an up-to-date about novel strategies for targeting TAMs.
Collapse
Affiliation(s)
- Arash Salmaninejad
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Sepideh Mehrpour Layeghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Falakian
- Department of Laboratory Science, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Shahin Golestani
- Department of Ophthalmology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Kobravi
- Department of Oral and Maxillofacial Surgery, Tehran Azad University, Tehran, Iran
| | - Samaneh Talebi
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Yousefi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
15
|
Yang L, Hu Q, Huang T. Breast Cancer Treatment Strategies Targeting the Tumor Microenvironment: How to Convert "Cold" Tumors to "Hot" Tumors. Int J Mol Sci 2024; 25:7208. [PMID: 39000314 PMCID: PMC11241188 DOI: 10.3390/ijms25137208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Breast cancer characterized as "cold tumors" exhibit low levels of immune cell infiltration, which limits the efficacy of conventional immunotherapy. Recent studies have focused on strategies using nanotechnology combined with tumor microenvironment modulation to transform "cold tumors" into "hot tumors". This approach involves the use of functionalized nanoparticles that target and modify the tumor microenvironment to promote the infiltration and activation of antitumor immune cells. By delivering immune activators or blocking immunosuppressive signals, these nanoparticles activate otherwise dormant immune responses, enhancing tumor immunogenicity and the therapeutic response. These strategies not only promise to increase the response rate of breast cancer patients to existing immunotherapies but also may pave new therapeutic avenues, providing a new direction for the immunotherapy of breast cancer.
Collapse
Affiliation(s)
- Liucui Yang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qingyi Hu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
16
|
Gong Y, Gao W, Zhang J, Dong X, Zhu D, Ma G. Engineering nanoparticles-enabled tumor-associated macrophages repolarization and phagocytosis restoration for enhanced cancer immunotherapy. J Nanobiotechnology 2024; 22:341. [PMID: 38890636 PMCID: PMC11184870 DOI: 10.1186/s12951-024-02622-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are pivotal within the immunosuppressive tumor microenvironment (TME), and recently, have attracted intensive attention for cancer treatment. However, concurrently to promote TAMs repolarization and phagocytosis of cancer cells remains challenging. Here, a TAMs-targeted albumin nanoparticles-based delivery system (M@SINPs) was constructed for the co-delivery of photosensitizer IR820 and SHP2 inhibitor SHP099 to potentiate macrophage-mediated cancer immunotherapy. M@SINPs under laser irradiation can generate the intracellular reactive oxygen species (ROS) and facilitate M2-TAMs to an M1 phenotype. Meanwhile, inhibition of SHP2 could block the CD47-SIRPa pathway to restore M1 macrophage phagocytic activity. M@SINPs-mediated TAMs remodeling resulted in the immunostimulatory TME by repolarizing TAMs to an M1 phenotype, restoring its phagocytic function and facilitating intratumoral CTLs infiltration, which significantly inhibited tumor growth. Furthermore, M@SINPs in combination with anti-PD-1 antibody could also improve the treatment outcomes of PD-1 blockade and exert the synergistic anticancer effects. Thus, the macrophage repolarization/phagocytosis restoration combination through M@SINPs holds promise as a strategy to concurrently remodel TAMs in TME for improving the antitumor efficiency of immune checkpoint block and conventional therapy.
Collapse
Affiliation(s)
- Yonghua Gong
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Wenyue Gao
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Jinyang Zhang
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Xia Dong
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China.
| | - Dunwan Zhu
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China.
| | - Guilei Ma
- Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China.
| |
Collapse
|
17
|
Zhao M, Shuai W, Su Z, Xu P, Wang A, Sun Q, Wang G. Protein tyrosine phosphatases: emerging role in cancer therapy resistance. Cancer Commun (Lond) 2024; 44:637-653. [PMID: 38741380 PMCID: PMC11194456 DOI: 10.1002/cac2.12548] [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: 10/31/2023] [Revised: 04/14/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Tyrosine phosphorylation of intracellular proteins is a post-translational modification that plays a regulatory role in signal transduction during cellular events. Dephosphorylation of signal transduction proteins caused by protein tyrosine phosphatases (PTPs) contributed their role as a convergent node to mediate cross-talk between signaling pathways. In the context of cancer, PTP-mediated pathways have been identified as signaling hubs that enabled cancer cells to mitigate stress induced by clinical therapy. This is achieved by the promotion of constitutive activation of growth-stimulatory signaling pathways or modulation of the immune-suppressive tumor microenvironment. Preclinical evidences suggested that anticancer drugs will release their greatest therapeutic potency when combined with PTP inhibitors, reversing drug resistance that was responsible for clinical failures during cancer therapy. AREAS COVERED This review aimed to elaborate recent insights that supported the involvement of PTP-mediated pathways in the development of resistance to targeted therapy and immune-checkpoint therapy. EXPERT OPINION This review proposed the notion of PTP inhibition in anticancer combination therapy as a potential strategy in clinic to achieve long-term tumor regression. Ongoing clinical trials are currently underway to assess the safety and efficacy of combination therapy in advanced-stage tumors.
Collapse
Affiliation(s)
- Min Zhao
- Innovation Center of Nursing ResearchNursing Key Laboratory of Sichuan ProvinceDepartment of BiotherapyCancer Center and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, West China School of Nursing, Sichuan UniversityChengduSichuanP. R. China
| | - Wen Shuai
- Innovation Center of Nursing ResearchNursing Key Laboratory of Sichuan ProvinceDepartment of BiotherapyCancer Center and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, West China School of Nursing, Sichuan UniversityChengduSichuanP. R. China
| | - Zehao Su
- Innovation Center of Nursing ResearchNursing Key Laboratory of Sichuan ProvinceDepartment of BiotherapyCancer Center and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, West China School of Nursing, Sichuan UniversityChengduSichuanP. R. China
- West China Biomedical Big Data CenterMed‐X Center for InformaticsSichuan UniversityChengduSichuanP. R. China
| | - Ping Xu
- Emergency DepartmentZigong Fourth People's HospitalChengduSichuanP. R. China
| | - Aoxue Wang
- Innovation Center of Nursing ResearchNursing Key Laboratory of Sichuan ProvinceDepartment of BiotherapyCancer Center and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, West China School of Nursing, Sichuan UniversityChengduSichuanP. R. China
| | - Qiu Sun
- Innovation Center of Nursing ResearchNursing Key Laboratory of Sichuan ProvinceDepartment of BiotherapyCancer Center and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, West China School of Nursing, Sichuan UniversityChengduSichuanP. R. China
| | - Guan Wang
- Innovation Center of Nursing ResearchNursing Key Laboratory of Sichuan ProvinceDepartment of BiotherapyCancer Center and State Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China Hospital, West China School of Nursing, Sichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
18
|
Kuznetsova AB, Kolesova EP, Parodi A, Zamyatnin AA, Egorova VS. Reprogramming Tumor-Associated Macrophage Using Nanocarriers: New Perspectives to Halt Cancer Progression. Pharmaceutics 2024; 16:636. [PMID: 38794298 PMCID: PMC11124960 DOI: 10.3390/pharmaceutics16050636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer remains a significant challenge for public healthcare systems worldwide. Within the realm of cancer treatment, considerable attention is focused on understanding the tumor microenvironment (TME)-the complex network of non-cancerous elements surrounding the tumor. Among the cells in TME, tumor-associated macrophages (TAMs) play a central role, traditionally categorized as pro-inflammatory M1 macrophages or anti-inflammatory M2 macrophages. Within the TME, M2-like TAMs can create a protective environment conducive to tumor growth and progression. These TAMs secrete a range of factors and molecules that facilitate tumor angiogenesis, increased vascular permeability, chemoresistance, and metastasis. In response to this challenge, efforts are underway to develop adjuvant therapy options aimed at reprogramming TAMs from the M2 to the anti-tumor M1 phenotype. Such reprogramming holds promise for suppressing tumor growth, alleviating chemoresistance, and impeding metastasis. Nanotechnology has enabled the development of nanoformulations that may soon offer healthcare providers the tools to achieve targeted drug delivery, controlled drug release within the TME for TAM reprogramming and reduce drug-related adverse events. In this review, we have synthesized the latest data on TAM polarization in response to TME factors, highlighted the pathological effects of TAMs, and provided insights into existing nanotechnologies aimed at TAM reprogramming and depletion.
Collapse
Affiliation(s)
- Alyona B. Kuznetsova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Ekaterina P. Kolesova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Andrey A. Zamyatnin
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Vera S. Egorova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| |
Collapse
|
19
|
Sun W, Xie S, Liu SF, Hu X, Xing D. Evolving Tumor Characteristics and Smart Nanodrugs for Tumor Immunotherapy. Int J Nanomedicine 2024; 19:3919-3942. [PMID: 38708176 PMCID: PMC11070166 DOI: 10.2147/ijn.s453265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Typical physiological characteristics of tumors, such as weak acidity, low oxygen content, and upregulation of certain enzymes in the tumor microenvironment (TME), provide survival advantages when exposed to targeted attacks by drugs and responsive nanomedicines. Consequently, cancer treatment has significantly progressed in recent years. However, the evolution and adaptation of tumor characteristics still pose many challenges for current treatment methods. Therefore, efficient and precise cancer treatments require an understanding of the heterogeneity degree of various factors in cancer cells during tumor evolution to exploit the typical TME characteristics and manage the mutation process. The highly heterogeneous tumor and infiltrating stromal cells, immune cells, and extracellular components collectively form a unique TME, which plays a crucial role in tumor malignancy, including proliferation, invasion, metastasis, and immune escape. Therefore, the development of new treatment methods that can adapt to the evolutionary characteristics of tumors has become an intense focus in current cancer treatment research. This paper explores the latest understanding of cancer evolution, focusing on how tumors use new antigens to shape their "new faces"; how immune system cells, such as cytotoxic T cells, regulatory T cells, macrophages, and natural killer cells, help tumors become "invisible", that is, immune escape; whether the diverse cancer-associated fibroblasts provide support and coordination for tumors; and whether it is possible to attack tumors in reverse. This paper discusses the limitations of targeted therapy driven by tumor evolution factors and explores future strategies and the potential of intelligent nanomedicines, including the systematic coordination of tumor evolution factors and adaptive methods, to meet this therapeutic challenge.
Collapse
Affiliation(s)
- Wenshe Sun
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Shaowei Xie
- Department of Ultrasound, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Shi Feng Liu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Xiaokun Hu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
| |
Collapse
|
20
|
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, Han P. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy. J Hematol Oncol 2024; 17:16. [PMID: 38566199 PMCID: PMC10986145 DOI: 10.1186/s13045-024-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Dongquan Kou
- Department of Rehabilitation Medicine, Chongqing Public Health Medical Center, Chongqing, China
| | - Shenghan Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Israel Canadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore
| | - Gautam Sethi
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Republic of Singapore
| | - Franklin Tay
- The Graduate School, Augusta University, 30912, Augusta, GA, USA
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Peng Han
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
21
|
Li SL, Hou HY, Chu X, Zhu YY, Zhang YJ, Duan MD, Liu J, Liu Y. Nanomaterials-Involved Tumor-Associated Macrophages' Reprogramming for Antitumor Therapy. ACS NANO 2024; 18:7769-7795. [PMID: 38420949 DOI: 10.1021/acsnano.3c12387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Tumor-associated macrophages (TAMs) play pivotal roles in tumor development. As primary contents of tumor environment (TME), TAMs secrete inflammation-related substances to regulate tumoral occurrence and development. There are two kinds of TAMs: the tumoricidal M1-like TAMs and protumoral M2-like TAMs. Reprogramming TAMs from immunosuppressive M2 to immunocompetent M1 phenotype is considered a feasible way to improve immunotherapeutic efficiency. Notably, nanomaterials show great potential for biomedical fields due to their controllable structures and properties. There are many types of nanomaterials that exhibit great regulatory activities for TAMs' reprogramming. In this review, the recent progress of nanomaterials-involved TAMs' reprogramming is comprehensively discussed. The various nanomaterials for TAMs' reprogramming and the reprogramming strategies are summarized and introduced. Additionally, the challenges and perspectives of TAMs' reprogramming for efficient therapy are discussed, aiming to provide inspiration for TAMs' regulator design and promote the development of TAMs-mediated immunotherapy.
Collapse
Affiliation(s)
- Shu-Lan Li
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Hua-Ying Hou
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Xu Chu
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Ying Zhu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Juan Zhang
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Meng-Die Duan
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Junyi Liu
- Albany Medical College, New York 12208, United States
| | - Yi Liu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, P. R. China
| |
Collapse
|
22
|
Xue Y, Yan X, Li D, Dong S, Ping Y. Proinflammatory polarization of engineered heat-inducible macrophages reprogram the tumor immune microenvironment during cancer immunotherapy. Nat Commun 2024; 15:2270. [PMID: 38491004 PMCID: PMC10943244 DOI: 10.1038/s41467-024-46210-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/16/2024] [Indexed: 03/18/2024] Open
Abstract
The success of macrophage-based adoptive cell therapy is largely constrained by poor polarization from alternatively activated (M2-like) to classically activated (M1-like) phenotype in the immunosuppressive tumor microenvironment (TME). Here, we show that the engineered macrophage (eMac) with a heat-inducible genetic switch can induce both self-polarization of adoptively transferred eMac and re-polarization of tumour-associated macrophages in response to mild temperature elevation in a mouse model. The locoregional production of proinflammatory cytokines by eMac in the TME dose not only induces the strong polarization of macrophages into a classically activated phenotype, but also ensures that the side effects typical for systemically administrate proinflammatory cytokines are avoided. We also present a wearable warming device which is adaptable for human patients and can be remotely controlled by a smartphone. In summary, our work represents a safe and efficient adoptive transfer immunotherapy method with potential for human translation.
Collapse
Affiliation(s)
- Yanan Xue
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaojie Yan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Da Li
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Shurong Dong
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuan Ping
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China.
| |
Collapse
|
23
|
Nandi D, Debnath M, Forster J, Pandey A, Bharadwaj H, Patel R, Kulkarni A. Nanoparticle-mediated co-delivery of inflammasome inhibitors provides protection against sepsis. NANOSCALE 2024; 16:4678-4690. [PMID: 38317511 DOI: 10.1039/d3nr05570a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The NLRP3 inflammasome, a multiprotein complex responsible for triggering the release of pro-inflammatory cytokines, plays a crucial role in inducing the inflammatory response associated with sepsis. While small molecule inhibitors of the NLRP3 inflammasome have been investigated for sepsis management, delivering NLRP3 inhibitors has been accompanied by several challenges, primarily related to the drug formulation, delivery route, stability, and toxicity. Many existing inflammasome inhibitors either show higher liver toxicity or require a high dosage to efficiently impede the inflammasome complex assembly. Moreover, the potential synergistic effects of combining multiple inflammasome inhibitors in sepsis therapy remain largely unexplored. Therefore, a rational approach is essential for presenting the potential administration of NLRP3 small molecule inhibitors to inhibit NLRP3 inflammasome activation effectively. In this context, we present a lipid nanoparticle-based dual-drug delivery system loaded with MCC 950 and disulfiram, demonstrating markedly higher efficiency compared to an equivalent amount of free-drug combinations and individual drug nanoparticles in vitro. This combination therapy substantially improved the in vivo survival rate of mice for LPS-induced septic peritonitis. Additionally, the synergistic approach illustrated a significant reduction in the expression of active caspase-1 as well as IL-1β inhibition integral components in the NLRP3 pathway. This study underscores the importance of integrating combination therapies facilitated by nanoparticle delivery to address the limitations of small molecule inflammasome inhibitors.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical Engineering, University of Massachusetts Amherst, MA, USA.
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, MA, USA
| | - Maharshi Debnath
- Department of Chemical Engineering, University of Massachusetts Amherst, MA, USA.
| | - James Forster
- Department of Chemical Engineering, University of Massachusetts Amherst, MA, USA.
| | - Ankit Pandey
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, MA, USA
| | - Hariharan Bharadwaj
- Department of Pathology, UMass Chan Medical School-Baystate, Springfield, Massachusetts 01107, United States.
| | - Ruchi Patel
- Department of Pathology, UMass Chan Medical School-Baystate, Springfield, Massachusetts 01107, United States.
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts Amherst, MA, USA.
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, MA, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, MA, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
24
|
Zhang Y, Liu L, Pei J, Ren Z, Deng Y, Yu K. Tissue factor overexpression promotes resistance to KRAS-G12C inhibition in non-small cell lung cancer. Oncogene 2024; 43:668-681. [PMID: 38191673 PMCID: PMC10890931 DOI: 10.1038/s41388-023-02924-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 01/10/2024]
Abstract
The recently approved KRASG12C mutation-specific inhibitors sotorasib and adagrasib (KRASG12C-I) represent a promising therapy for KRASG12C-driven non-small cell lung cancer (NSCLC). However, many eligible patients do not benefit due to intrinsic or acquired drug resistance. Tissue factor (TF) is overexpressed in KRAS-mutated (KRASmut) NSCLC and is the target of the FDA-approved ADC Tivdak. Here, we employed HuSC1-39, the parent antibody of a clinical stage TF-ADC (NCT04843709), to investigate the role of TF in KRASmut NSCLC. We found that patients with TF-overexpression had poor survival, elevated P-ERK/P-AKT activity levels and low immune effector cell infiltration in the tumor. In a panel of KRASG12C cell lines, KRASG12C-I response correlated with suppression of TF mRNA, which was not observed in resistant cells. In the drug resistant cells, TF-overexpression relied on an mTORC2-mediated and proteasome-dependent pathway. Combination treatment of HuSC1-39 or mTORC1/2 inhibitor MTI-31 with KRASG12C-I each produced synergistic antitumor efficacy in cell culture and in an orthotopic lung tumor model. TF-depletion in the resistant cells diminished epithelial mesenchymal transition, reduced tumor growth and greatly sensitized KRASG12C-I response. Moreover, employing immunohistochemistry and coculture studies, we demonstrated that HuSC1-39 or MTI-31 reset the tumor microenvironment and restore KRASG12C-I sensitivity by reshaping an M1-like macrophage profile with greatly enhanced phagocytic capacity toward tumor cell killing. Thus, we have identified the TF/mTORC2 axis as a critical new mechanism for triggering immunosuppression and KRASG12C-I resistance. We propose that targeting this axis with HuSC1-39 or MTI-31 will improve KRASG12C-I response in KRAS-driven NSCLC.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Liang Liu
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Jinpeng Pei
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Zhiqiang Ren
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Yan Deng
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Ker Yu
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China.
| |
Collapse
|
25
|
Hu Y, Nie W, Lyu L, Zhang X, Wang W, Zhang Y, He S, Guo A, Liu F, Wang B, Qian Z, Gao X. Tumor-Microenvironment-Activatable Nanoparticle Mediating Immunogene Therapy and M2 Macrophage-Targeted Inhibitor for Synergistic Cancer Immunotherapy. ACS NANO 2024; 18:3295-3312. [PMID: 38252684 DOI: 10.1021/acsnano.3c10037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Immunotherapy has achieved prominent clinical efficacy in combating cancer and has recently become a mainstream treatment strategy. However, achieving broad efficacy with a single modality is challenging, and the heterogeneity of the tumor microenvironment (TME) restricts the accuracy and effectiveness of immunotherapy strategies for tumors. Herein, a TME-responsive targeted nanoparticle to enhance antitumor immunity and reverse immune escape by codelivering interleukin-12 (IL-12) expressing gene and colony-stimulating factor-1 receptor (CSF-1R) inhibitor PLX3397 (PLX) is presented. The introduction of disulfide bonds and cyclo(Arg-Gly-Asp-d-Phe-Lys) (cRGD) peptides conferred reduction reactivity and tumor targeting to the nanoparticles, respectively. It is hypothesized that activating host immunity by the local expression of IL-12, while modulating the tumor-associated macrophages (TAM) function through blocking CSF-1/CSF-1R signaling, could constitute a feasible approach for cancer immunotherapy. The fabricated functional nanoparticle successfully ameliorated the TME by stimulating the proliferation and activation of T lymphocytes, promoting the repolarization of TAMs, reducing myeloid-derived suppressor cells (MDSCs), and promoting the maturation of dendritic cells (DC) as well as the secretion of antitumor cytokines, which efficiently suppressed tumor growth and metastasis. Finally, substantial changes in the TME were deciphered by single-cell analysis including infiltration of different cells, transcriptional states, secretory signaling and cell-cell communications. These findings provide a promising combinatorial immunotherapy strategy through immunomodulatory nanoparticles.
Collapse
Affiliation(s)
- Yuzhu Hu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
- Department of Radiation Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Wen Nie
- Department of Radiation Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Liang Lyu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Xifeng Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Wanyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Yunchu Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Anjie Guo
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Fei Liu
- Department of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| |
Collapse
|
26
|
Zhou B, Yang Y, Kang Y, Hou J, Yang Y. Targeting the macrophage immunocheckpoint: a novel insight into solid tumor immunotherapy. Cell Commun Signal 2024; 22:66. [PMID: 38273373 PMCID: PMC10809660 DOI: 10.1186/s12964-023-01384-x] [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: 09/04/2023] [Accepted: 11/04/2023] [Indexed: 01/27/2024] Open
Abstract
Tumor immunotherapy, which targets immune checkpoints, presents a promising strategy for the treatment of various cancer types. However, current clinical data indicate challenges in its application to solid tumors. Recent studies have revealed a significant correlation between the degree of immune response in immunotherapy and the tumor microenvironment, particularly with regard to tumor-infiltrating immune cells. Among these immune cells, macrophages, a critical component, are playing an increasingly vital role in tumor immunotherapy. This review focuses on elucidating the role of macrophages within solid tumors and provides an overview of the progress in immunotherapy approaches centered around modulating macrophage responses through various immune factors. Video Abstract.
Collapse
Affiliation(s)
- Bei Zhou
- Department of Biochemistry and molecular biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Yan Yang
- Department of Biochemistry and molecular biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Yan Kang
- Department of Biochemistry and molecular biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Jingjing Hou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, 361004, China.
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen, Fujian, 361004, China.
| | - Yun Yang
- Department of Biochemistry and molecular biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
| |
Collapse
|
27
|
Kang X, Huang Y, Wang H, Jadhav S, Yue Z, Tiwari AK, Babu RJ. Tumor-Associated Macrophage Targeting of Nanomedicines in Cancer Therapy. Pharmaceutics 2023; 16:61. [PMID: 38258072 PMCID: PMC10819517 DOI: 10.3390/pharmaceutics16010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/24/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
The tumor microenvironment (TME) is pivotal in tumor growth and metastasis, aligning with the "Seed and Soil" theory. Within the TME, tumor-associated macrophages (TAMs) play a central role, profoundly influencing tumor progression. Strategies targeting TAMs have surfaced as potential therapeutic avenues, encompassing interventions to block TAM recruitment, eliminate TAMs, reprogram M2 TAMs, or bolster their phagocytic capabilities via specific pathways. Nanomaterials including inorganic materials, organic materials for small molecules and large molecules stand at the forefront, presenting significant opportunities for precise targeting and modulation of TAMs to enhance therapeutic efficacy in cancer treatment. This review provides an overview of the progress in designing nanoparticles for interacting with and influencing the TAMs as a significant strategy in cancer therapy. This comprehensive review presents the role of TAMs in the TME and various targeting strategies as a promising frontier in the ever-evolving field of cancer therapy. The current trends and challenges associated with TAM-based therapy in cancer are presented.
Collapse
Affiliation(s)
- Xuejia Kang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA;
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Yongzhuo Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangzhou 528400, China;
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Huiyuan Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Sanika Jadhav
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA;
| | - Zongliang Yue
- Department of Health Outcome and Research Policy, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA;
| | - Amit K. Tiwari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas of Medical Sciences, Little Rock, AR 72205, USA;
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA;
| |
Collapse
|
28
|
Nandi D, Forster J, Ramesh A, Nguyen A, Bharadwaj H, Kulkarni A. Caspase-1 Responsive Nanoreporter for In Vivo Monitoring of Inflammasome Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:55545-55558. [PMID: 37990965 PMCID: PMC11056827 DOI: 10.1021/acsami.3c15733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Inflammasomes are multimeric protein signaling complexes that are assembled in innate immune cells in response to a multitude of pathogen and damage-associated signals. They are essential for generating robust inflammatory responses to prevent pathogenic insults. However, inflammasome dysregulation can induce cascading immune responses, resulting in systemic toxicities and inflammatory disease. In this sense, there is a strong need to develop potent inflammasome inhibiting therapies as well as technologies to monitor their efficacy, yet current systems lack the ability to effectively image inflammasome activation and track therapy response early. To overcome these limitations, we report a novel nanoparticle system delivering both a caspase-1 cleavable inflammasome detecting probe and the NLRP3 inhibitor drug MCC-950, providing dual capabilities of monitoring and regulation of inflammasome activation in a biocompatible, tissue penetrating, and sustained release liposomal formulation. We observed this liposomal nanoreporter's ability to reduce and detect inflammasome activation both in vitro in immortalized bone marrow-derived macrophages and in vivo in a DSS-induced ulcerative colitis mouse model. Our results exhibited the nanoreporter's ability to penetrate inflammatory tissues and detect inflammasome activation early and in real-time for multiple days while alleviating inflammation in the groups coencapsulating imaging reporter and inflammasome inhibitor. Overall, the developed liposomal nanoreporter platform enables spatiotemporal delivery of imaging probe and inhibitor, captures early and sustained inflammasome detection, and induces inflammasome amelioration, thus establishing a novel tool for the real-time monitoring and treatment of inflammasome-mediated disease with high potential for clinical application.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical Engineering and Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - James Forster
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Anujan Ramesh
- Department of Chemical Engineering and Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Anh Nguyen
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Hariharan Bharadwaj
- Department of Pathology, UMass Chan, Medical School-Baystate, Springfield, Massachusetts 01107, United States
| | - Ashish Kulkarni
- Department of Chemical Engineering, Department of Veterinary and Animal Sciences, Department of Biomedical Engineering, and Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
29
|
Gong X, Liu Y, Liang K, Chen Z, Ding K, Qiu L, Wei J, Du H. Cucurbitacin I Reverses Tumor-Associated Macrophage Polarization to Affect Cancer Cell Metastasis. Int J Mol Sci 2023; 24:15920. [PMID: 37958903 PMCID: PMC10650020 DOI: 10.3390/ijms242115920] [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: 09/28/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
The tumor microenvironment plays a critical role in tumor progression and immune regulation. As one of the most important components of the tumor microenvironment, macrophages have become a new therapeutic target for inhibiting tumor progression. Despite the well-documented anticancer activity of cucurbitacin I, its effect on macrophages remains unclear. In this study, we established a coculture system of macrophages and cancer cells under hypoxic conditions to simulate the tumor-promoting environment mediated by M2-like macrophages. We determined whether cucurbitacin I modulates M2-like polarization in macrophages in vitro and conducted RNA sequencing to identify gene expression changes induced by cucurbitacin I in macrophages. The results indicated a remarkable inhibition of the M2-like polarization phenotype in macrophages following treatment with cucurbitacin I, which was accompanied by the significant downregulation of heme oxygenase-1. Moreover, we found that cucurbitacin I-treated macrophages reduced the migration of cancer cells by inhibiting the M2 polarization in vitro. These findings highlight the potential of cucurbitacin I as a therapeutic agent that targets M2-like macrophages to inhibit cancer cell metastasis. Our study provides novel insights into the intricate interplay among macrophage polarization, cucurbitacin I, and heme oxygenase-1, thereby opening new avenues for cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinfen Wei
- School of Biology and Biological Engineering, South China University of Technology, University Town Campus, Guangzhou Higher Education Mega Centre, Panyu District, Guangzhou 510006, China; (X.G.); (Y.L.); (K.L.); (Z.C.); (K.D.); (L.Q.)
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, University Town Campus, Guangzhou Higher Education Mega Centre, Panyu District, Guangzhou 510006, China; (X.G.); (Y.L.); (K.L.); (Z.C.); (K.D.); (L.Q.)
| |
Collapse
|
30
|
Sodir NM, Pathria G, Adamkewicz JI, Kelley EH, Sudhamsu J, Merchant M, Chiarle R, Maddalo D. SHP2: A Pleiotropic Target at the Interface of Cancer and Its Microenvironment. Cancer Discov 2023; 13:2339-2355. [PMID: 37682219 PMCID: PMC10618746 DOI: 10.1158/2159-8290.cd-23-0383] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/20/2023] [Accepted: 07/27/2023] [Indexed: 09/09/2023]
Abstract
The protein phosphatase SHP2/PTPN11 has been reported to be a key modulator of proliferative pathways in a wide range of malignancies. Intriguingly, SHP2 has also been described as a critical regulator of the tumor microenvironment. Based on this evidence SHP2 is considered a multifaceted target in cancer, spurring the notion that the development of direct inhibitors of SHP2 would provide the twofold benefit of tumor intrinsic and extrinsic inhibition. In this review, we will discuss the role of SHP2 in cancer and the tumor microenvironment, and the clinical strategies in which SHP2 inhibitors are leveraged as combination agents to improve therapeutic response. SIGNIFICANCE The SHP2 phosphatase functions as a pleiotropic factor, and its inhibition not only hinders tumor growth but also reshapes the tumor microenvironment. Although their single-agent activity may be limited, SHP2 inhibitors hold the potential of being key combination agents to enhance the depth and the durability of tumor response to therapy.
Collapse
Affiliation(s)
- Nicole M. Sodir
- Department of Translational Oncology, Genentech, South San Francisco, California
| | - Gaurav Pathria
- Department of Oncology Biomarker Development, Genentech, South San Francisco, California
| | | | - Elizabeth H. Kelley
- Department of Discovery Chemistry, Genentech, South San Francisco, California
| | - Jawahar Sudhamsu
- Department of Structural Biology, Genentech, South San Francisco, California
| | - Mark Merchant
- Department of Translational Oncology, Genentech, South San Francisco, California
| | - Roberto Chiarle
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Danilo Maddalo
- Department of Translational Oncology, Genentech, South San Francisco, California
| |
Collapse
|
31
|
Ashrafizadeh M, Zarrabi A, Bigham A, Taheriazam A, Saghari Y, Mirzaei S, Hashemi M, Hushmandi K, Karimi-Maleh H, Nazarzadeh Zare E, Sharifi E, Ertas YN, Rabiee N, Sethi G, Shen M. (Nano)platforms in breast cancer therapy: Drug/gene delivery, advanced nanocarriers and immunotherapy. Med Res Rev 2023; 43:2115-2176. [PMID: 37165896 DOI: 10.1002/med.21971] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/09/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023]
Abstract
Breast cancer is the most malignant tumor in women, and there is no absolute cure for it. Although treatment modalities including surgery, chemotherapy, and radiotherapy are utilized for breast cancer, it is still a life-threatening disease for humans. Nanomedicine has provided a new opportunity in breast cancer treatment, which is the focus of the current study. The nanocarriers deliver chemotherapeutic agents and natural products, both of which increase cytotoxicity against breast tumor cells and prevent the development of drug resistance. The efficacy of gene therapy is boosted by nanoparticles and the delivery of CRISPR/Cas9, Noncoding RNAs, and RNAi, promoting their potential for gene expression regulation. The drug and gene codelivery by nanoparticles can exert a synergistic impact on breast tumors and enhance cellular uptake via endocytosis. Nanostructures are able to induce photothermal and photodynamic therapy for breast tumor ablation via cell death induction. The nanoparticles can provide tumor microenvironment remodeling and repolarization of macrophages for antitumor immunity. The stimuli-responsive nanocarriers, including pH-, redox-, and light-sensitive, can mediate targeted suppression of breast tumors. Besides, nanoparticles can provide a diagnosis of breast cancer and detect biomarkers. Various kinds of nanoparticles have been employed for breast cancer therapy, including carbon-, lipid-, polymeric- and metal-based nanostructures, which are different in terms of biocompatibility and delivery efficiency.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Turkey
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials - National Research Council (IPCB-CNR), Naples, Italy
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yalda Saghari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hassan Karimi-Maleh
- School of Resources and Environment, University of Electronic Science and Technology of China, Chengdu, PR China
| | | | - Esmaeel Sharifi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Türkiye
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, New South Wales, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mingzhi Shen
- Department of Cardiology, Hainan Hospital of PLA General Hospital, Sanya, China
| |
Collapse
|
32
|
Ramesh A, Deshpande N, Malik V, Nguyen A, Malhotra M, Debnath M, Brouillard A, Kulkarni A. Activatable Nanoreporters for Real-Time Tracking of Macrophage Phenotypic States Associated with Disease Progression. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300978. [PMID: 37317008 DOI: 10.1002/smll.202300978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/05/2023] [Indexed: 06/16/2023]
Abstract
Diagnosis of inflammatory diseases is characterized by identifying symptoms, biomarkers, and imaging. However, conventional techniques lack the sensitivities and specificities to detect disease early. Here, it is demonstrated that the detection of macrophage phenotypes, from inflammatory M1 to alternatively activated M2 macrophages, corresponding to the disease state can be used to predict the prognosis of various diseases. Activatable nanoreporters that can longitudinally detect the presence of the enzyme Arginase 1, a hallmark of M2 macrophages, and nitric oxide, a hallmark of M1 macrophages are engineered, in real-time. Specifically, an M2 nanoreporter enables the early imaging of the progression of breast cancer as predicted by selectively detecting M2 macrophages in tumors. The M1 nanoreporter enables real-time imaging of the subcutaneous inflammatory response that rises from a local lipopolysccharide (LPS) administration. Finally, the M1-M2 dual nanoreporter is evaluated in a muscle injury model, where an initial inflammatory response is monitored by imaging M1 macrophages at the site of inflammation, followed by a resolution phase monitored by the imaging of infiltrated M2 macrophages involved in matrix regeneration and wound healing. It is anticipated that this set of macrophage nanoreporters may be utilized for early diagnosis and longitudinal monitoring of inflammatory responses in various disease models.
Collapse
Affiliation(s)
- Anujan Ramesh
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Nilesh Deshpande
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Vaishali Malik
- Department of Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Anh Nguyen
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Mehak Malhotra
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Maharshi Debnath
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Anthony Brouillard
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Ashish Kulkarni
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
33
|
Wang J, Zhang Z, Chen Y. Supramolecular immunotherapy on diversiform immune cells. J Mater Chem B 2023; 11:8347-8367. [PMID: 37563947 DOI: 10.1039/d3tb00924f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Supramolecular immunotherapy employs supramolecular materials to stimulate the immune system for inhibiting tumor cell growth and metastasis, reducing the cancer recurrence rate, and improving the quality of the patient's life. Additionally, it can lessen patient suffering and the deterioration of their illness, as well as increase their survival rate. This paper will outline the fundamentals of tumor immunotherapy based on supramolecular materials as well as its current state of development and potential applications. To be more specific, we will first introduce the basic principles of supramolecular immunotherapy, including the processes, advantages and limitations of immunotherapy, the construction of supramolecular material structures, and its benefits in treatment. Second, considering the targeting of supramolecular drugs to immune cells, we comprehensively discuss the unique advantages of applying supramolecular drugs with different types of immune cells in tumor immunotherapy. The current research advances in supramolecular immunotherapy, including laboratory research and clinical applications, are also described in detail. Finally, we reveal the tremendous promise of supramolecular materials in tumor immunotherapy, as well as discuss the opportunities and challenges that may be faced in future development.
Collapse
Affiliation(s)
- Jiahui Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, P. R. China
| | - Ziyi Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, P. R. China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, P. R. China
| |
Collapse
|
34
|
Kinskey JC, Huda TI, Gozlan EC, Quach JU, Arturo JF, Chobrutskiy A, Chobrutskiy BI, Blanck G. The presence of intratumoral Porphyromonas gingivalis correlates with a previously defined pancreatic adenocarcinoma, immune cell expression phenotype and with tumor resident, adaptive immune receptor features. Carcinogenesis 2023; 44:411-417. [PMID: 37195907 DOI: 10.1093/carcin/bgad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/20/2023] [Accepted: 05/15/2023] [Indexed: 05/19/2023] Open
Abstract
The association between pancreatic adenocarcinoma (PAAD) and the pancreatic microbiome is not fully understood, although bacteria may decrease the effectiveness of chemotherapy and lead to anti-apoptotic, pro-inflammatory microenvironments. To better understand the relationship between the PAAD microbiome and the microenvironment, we identified Porphyromonas gingivalis-positive PAAD samples and found a strong association between intratumoral P. gingivalis and: (i) an immune cell gene expression phenotype previously defined by others as gene program 7; and (ii) recovery of immunoglobulin recombination, sequencing reads. We applied a novel chemical complementarity scoring algorithm, suitable for a big data setting, and determined that the previously established P. gingivalis antigen, rpgB had a reduced chemical complementarity with T-cell receptor (TCR) complementarity-determining region-3 amino acid sequences recovered from PAAD samples with P. gingivalis in comparison to TCR-rpgB chemical complementarity represented by the PAAD samples that lacked P. gingivalis. This finding strengthens the existing body of evidence correlating P. gingivalis with PAAD, which may have implications for the treatment and prognosis of patients. Furthermore, demonstrating the correlation of P. gingivalis and gene program 7 raises the question of whether P. gingivalis infection is responsible for the gene program 7 subdivision of PAAD?
Collapse
Affiliation(s)
- Jacob C Kinskey
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Taha I Huda
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Etienne C Gozlan
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Jessica U Quach
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Juan F Arturo
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Andrea Chobrutskiy
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Boris I Chobrutskiy
- Department of Internal Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - George Blanck
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
35
|
Zheng C, Zhong Q, Yi K, Kong H, Cao F, Zhuo C, Xu Y, Shi R, Ju E, Song W, Tao Y, Chen X, Li M. Anti-phagocytosis-blocking repolarization-resistant membrane-fusogenic liposome (ARMFUL) for adoptive cell immunotherapy. SCIENCE ADVANCES 2023; 9:eadh2413. [PMID: 37556535 PMCID: PMC10411906 DOI: 10.1126/sciadv.adh2413] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/07/2023] [Indexed: 08/11/2023]
Abstract
Equipping multiple functionalities on adoptive effector cells is essential to overcome the complex immunological barriers in solid tumors for superior antitumor efficacy. However, current cell engineering technologies cannot endow these functionalities to cells within a single step because of the different spatial distributions of targets in one cell. Here, we present a core-shell anti-phagocytosis-blocking repolarization-resistant membrane-fusogenic liposome (ARMFUL) to achieve one-step multiplexing cell engineering for multifunctional cell construction. Through fusing with the M1 macrophage membrane, ARMFUL inserts an anti-CD47 (aCD47)-modified lipid shell onto the surface and simultaneously delivers colony-stimulating factor 1 receptor inhibitor BLZ945-loaded core into the cytoplasm. The surface-presenting aCD47 boosts macrophage's phagocytosis against the tumor by blocking CD47. The cytoplasm-located BLZ945 prompts its polarization resistance to M2 phenotype in the immunosuppressive microenvironment via inactivating the intracellular M2 polarization signaling pathway. This ARMFUL provides a versatile cell engineering platform to customize multimodal cellular functions for enhanced adoptive cell therapy.
Collapse
Affiliation(s)
- Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Chenya Zhuo
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Enguo Ju
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
36
|
Lucianò AM, Di Martile M, Pérez-Oliva AB, Di Caprio M, Foddai ML, Buglioni S, Mulero V, Del Bufalo D. Exploring association of melanoma-specific Bcl-xL with tumor immune microenvironment. J Exp Clin Cancer Res 2023; 42:178. [PMID: 37488586 PMCID: PMC10364435 DOI: 10.1186/s13046-023-02735-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/16/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Macrophages take center stage in the tumor microenvironment, a niche composed of extracellular matrix and a heterogeneous group of cells, including immune ones. They can evolve during tumor progression and acquire Tumor-Associated Macrophage (TAMs) phenotype. The release of cytokines by tumor and stromal cells, influence the secretion of cytokines by TAMs, which can guarantee tumor progression and influence the response to therapy. Among all factors able to recruit and polarize macrophages, we focused our attention on Bcl-xL, a multifaceted member of the Bcl-2 family, whose expression is deregulated in melanoma. It acts not only as a canonical pro-survival and anti-apoptotic protein, but also as a promoter of tumor progression. METHODS Human melanoma cells silencing or overexpressing Bcl-xL protein, THP-1 monocytic cells and monocyte-derived macrophages were used in this study. Protein array and specific neutralizing antibodies were used to analyze cytokines and chemokines secreted by melanoma cells. qRT-PCR, ELISA and Western Blot analyses were used to evaluate macrophage polarization markers and protein expression levels. Transwell chambers were used to evaluate migration of THP-1 and monocyte-derived macrophages. Mouse and zebrafish models were used to evaluate the ability of melanoma cells to recruit and polarize macrophages in vivo. RESULTS We demonstrated that melanoma cells overexpressing Bcl-xL recruit macrophages at the tumor site and induce a M2 phenotype. In addition, we identified that interleukin-8 and interleukin-1β cytokines are involved in macrophage polarization, and the chemokine CCL5/RANTES in the macrophages recruitment at the tumor site. We also found that all these Bcl-xL-induced factors are regulated in a NF-kB dependent manner in human and zebrafish melanoma models. CONCLUSIONS Our findings confirmed the pro-tumoral function of Bcl-xL in melanoma through its effects on macrophage phenotype.
Collapse
Affiliation(s)
- Anna Maria Lucianò
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, 30100, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Murcia, 30120, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Ana B Pérez-Oliva
- Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Murcia, 30120, Spain
| | - Marica Di Caprio
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Laura Foddai
- Immunohematology and Transfusional Medicine Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Simonetta Buglioni
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Victoriano Mulero
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, 30100, Spain.
- Instituto Murciano de Investigación Biosanitaria (IMIB) Pascual Parrilla, Murcia, 30120, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, 28029, Spain.
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
37
|
Wang F, Cheng F, Zheng F. Bioinformatic-based genetic characterizations of neural regulation in skin cutaneous melanoma. Front Oncol 2023; 13:1166373. [PMID: 37404751 PMCID: PMC10315675 DOI: 10.3389/fonc.2023.1166373] [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: 02/15/2023] [Accepted: 05/31/2023] [Indexed: 07/06/2023] Open
Abstract
Background Recent discoveries uncovered the complex cancer-nerve interactions in several cancer types including skin cutaneous melanoma (SKCM). However, the genetic characterization of neural regulation in SKCM is unclear. Methods Transcriptomic expression data were collected from the TCGA and GTEx portal, and the differences in cancer-nerve crosstalk-associated gene expressions between normal skin and SKCM tissues were analyzed. The cBioPortal dataset was utilized to implement the gene mutation analysis. PPI analysis was performed using the STRING database. Functional enrichment analysis was analyzed by the R package clusterProfiler. K-M plotter, univariate, multivariate, and LASSO regression were used for prognostic analysis and verification. The GEPIA dataset was performed to analyze the association of gene expression with SKCM clinical stage. ssGSEA and GSCA datasets were used for immune cell infiltration analysis. GSEA was used to elucidate the significant function and pathway differences. Results A total of 66 cancer-nerve crosstalk-associated genes were identified, 60 of which were up- or downregulated in SKCM and KEGG analysis suggested that they are mainly enriched in the calcium signaling pathway, Ras signaling pathway, PI3K-Akt signaling pathway, and so on. A gene prognostic model including eight genes (GRIN3A, CCR2, CHRNA4, CSF1, NTN1, ADRB1, CHRNB4, and CHRNG) was built and verified by independent cohorts GSE59455 and GSE19234. A nomogram was constructed containing clinical characteristics and the above eight genes, and the AUCs of the 1-, 3-, and 5-year ROC were 0.850, 0.811, and 0.792, respectively. Expression of CCR2, GRIN3A, and CSF1 was associated with SKCM clinical stages. There existed broad and strong correlations of the prognostic gene set with immune infiltration and immune checkpoint genes. CHRNA4 and CHRNG were independent poor prognostic genes, and multiple metabolic pathways were enriched in high CHRNA4 expression cells. Conclusion Comprehensive bioinformatics analysis of cancer-nerve crosstalk-associated genes in SKCM was performed, and an effective prognostic model was constructed based on clinical characteristics and eight genes (GRIN3A, CCR2, CHRNA4, CSF1, NTN1, ADRB1, CHRNB4, and CHRNG), which were widely related to clinical stages and immunological features. Our work may be helpful for further investigation in the molecular mechanisms correlated with neural regulation in SKCM, and in searching new therapeutic targets.
Collapse
Affiliation(s)
- Fengdi Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fanjun Cheng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fang Zheng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
38
|
Fan S, Han H, Yan Z, Lu Y, He B, Zhang Q. Lipid-based nanoparticles for cancer immunotherapy. MEDICAL REVIEW (2021) 2023; 3:230-269. [PMID: 37789955 PMCID: PMC10542882 DOI: 10.1515/mr-2023-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/07/2023] [Indexed: 10/05/2023]
Abstract
As the fourth most important cancer management strategy except surgery, chemotherapy and radiotherapy, cancer immunotherapy has been confirmed to elicit durable antitumor effects in the clinic by leveraging the patient's own immune system to eradicate the cancer cells. However, the limited population of patients who benefit from the current immunotherapies and the immune related adverse events hinder its development. The immunosuppressive microenvironment is the main cause of the failure, which leads to cancer immune evasion and immunity cycle blockade. Encouragingly, nanotechnology has been engineered to enhance the efficacy and reduce off-target toxicity of their therapeutic cargos by spatiotemporally controlling the biodistribution and release kinetics. Among them, lipid-based nanoparticles are the first nanomedicines to make clinical translation, which are now established platforms for diverse areas. In this perspective, we discuss the available lipid-based nanoparticles in research and market here, then describe their application in cancer immunotherapy, with special emphasis on the T cells-activated and macrophages-targeted delivery system. Through perpetuating each step of cancer immunity cycle, lipid-based nanoparticles can reduce immunosuppression and promote drug delivery to trigger robust antitumor response.
Collapse
Affiliation(s)
- Shumin Fan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Huize Han
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhicheng Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yao Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo, Zhejiang Province, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo, Zhejiang Province, China
| |
Collapse
|
39
|
Xu D, Liu Z, Liang MX, Chen WQ, Fei YJ, Yang SJ, Wu Y, Zhang W, Tang JH. Hyperthermia promotes M1 polarization of macrophages via exosome-mediated HSPB8 transfer in triple negative breast cancer. Discov Oncol 2023; 14:81. [PMID: 37233869 DOI: 10.1007/s12672-023-00697-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/19/2023] [Indexed: 05/27/2023] Open
Abstract
PURPOSE To investigate the mechanism underlying the modulation of M1 macrophage polarization by exosomes released from hyperthermia-treated triple-negative breast cancer (TNBC) cells. MATERIALS AND METHODS In this study, the effects of hyperthermia on TNBC cells were examined using cell counting kit-8, apoptosis, and cell cycle assays. Transmission electron microscopy was used to identify the structure of exosomes, while bicinchoninic acid and nanoparticle tracking analysis were used to detect particle size and amounts of exosomes released after hyperthermia. The polarization of macrophages incubated with exosomes derived by hyperthermia-pretreated TNBC cells were assessed by RT-qPCR and flow cytometry analysis. Next, RNA sequencing was performed to determine the targeting molecules changed in hyperthermia-treated TNBC cells in vitro. Finally, the mechanism underlying the modulation of macrophage polarization by exosomes derived from hyperthermia-treated TNBC cells was examined by using RT-qPCR, immunofluorescence and flow cytometry analysis. RESULTS Hyperthermia markedly reduced cell viability in TNBC cells and promoted the secretion of TNBC cell-derived exosomes. The hub genes of hyperthermia-treated TNBC cells were significantly correlated with macrophage infiltration. Additionally, hyperthermia-treated TNBC cell-derived exosomes promoted M1 macrophage polarization. Furthermore, the expression levels of heat shock proteins, including HSPA1A, HSPA1B, HSPA6, and HSPB8, were significantly upregulated upon hyperthermia treatment, with HSPB8 exhibiting the highest upregulation. Moreover, hyperthermia can induce M1 macrophage polarization by promoting exosome-mediated HSPB8 transfer. CONCLUSION This study demonstrated a novel mechanism that hyperthermia can induce M1 polarization of macrophages via exosome-mediated HSPB8 transfer. These results will help with future development of an optimized hyperthermia treatment regime for clinical application, especially for combination treatment with immunotherapy.
Collapse
Affiliation(s)
- Di Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Zhen Liu
- Department of General Surgery, Taixing People's Hospital, Taixing, 225400, People's Republic of China
| | - Ming-Xing Liang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Wen-Quan Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Yin-Jiao Fei
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Su-Jin Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Yang Wu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
- Department of Biobank, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Wei Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China.
| | - Jin-Hai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China.
| |
Collapse
|
40
|
Aarhus TI, Bjørnstad F, Wolowczyk C, Larsen KU, Rognstad L, Leithaug T, Unger A, Habenberger P, Wolf A, Bjørkøy G, Pridans C, Eickhoff J, Klebl B, Hoff BH, Sundby E. Synthesis and Development of Highly Selective Pyrrolo[2,3- d]pyrimidine CSF1R Inhibitors Targeting the Autoinhibited Form. J Med Chem 2023; 66:6959-6980. [PMID: 37191268 DOI: 10.1021/acs.jmedchem.3c00428] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Colony-stimulating factor-1 receptor (CSF1R) is a receptor tyrosine kinase that controls the differentiation and maintenance of most tissue-resident macrophages, and the inhibition of CSF1R has been suggested as a possible therapy for a range of human disorders. Herein, we present the synthesis, development, and structure-activity relationship of a series of highly selective pyrrolo[2,3-d]pyrimidines, showing subnanomolar enzymatic inhibition of this receptor and with excellent selectivity toward other kinases in the platelet-derived growth factor receptor (PDGFR) family. The crystal structure of the protein and 23 revealed that the binding conformation of the protein is DFG-out-like. The most promising compounds in this series were profiled for cellular potency and subjected to pharmacokinetic profiling and in vivo stability, indicating that this compound class could be relevant in a potential disease setting. Additionally, these compounds inhibited primarily the autoinhibited form of the receptor, contrasting the behavior of pexidartinib, which could explain the exquisite selectivity of these structures.
Collapse
Affiliation(s)
- Thomas Ihle Aarhus
- Department of Materials Science & Engineering, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Frithjof Bjørnstad
- Department of Materials Science & Engineering, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Camilla Wolowczyk
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | | | - Line Rognstad
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Trygve Leithaug
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Anke Unger
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Peter Habenberger
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Alexander Wolf
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Geir Bjørkøy
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Clare Pridans
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, U.K
| | - Jan Eickhoff
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Bert Klebl
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Bård H Hoff
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Eirik Sundby
- Department of Materials Science & Engineering, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| |
Collapse
|
41
|
Hao Y, Zhou X, Li Y, Li B, Cheng L. The CD47-SIRPα axis is a promising target for cancer immunotherapies. Int Immunopharmacol 2023; 120:110255. [PMID: 37187126 DOI: 10.1016/j.intimp.2023.110255] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/17/2023]
Abstract
Cluster of differentiation 47(CD47) is a transmembrane protein that is ubiquitously found on the surface of many cells in the body and uniquely overexpressed by both solid and hematologic malignant cells. CD47 interacts with signal-regulatory protein α (SIRPα), to trigger a "don't eat me" signal and thereby achieve cancer immune escape by inhibiting macrophage-mediated phagocytosis. Thus, blocking the CD47-SIRPα phagocytosis checkpoint, for release of the innate immune system, is a current research focus. Indeed, targeting the CD47-SIRPα axis as a cancer immunotherapy has shown promising efficacies in pre-clinical outcomes. Here, we first reviewed the origin, structure, and function of the CD47-SIRPα axis. Then, we reviewed its role as a target for cancer immunotherapies, as well as the factors regulating CD47-SIRPα axis-based immunotherapies. We specifically focused on the mechanism and progress of CD47-SIRPα axis-based immunotherapies and their combination with other treatment strategies. Finally, we discussed the challenges and directions for future research and identified potential CD47-SIRPα axis-based therapies that are suitable for clinical application.
Collapse
Affiliation(s)
- Yu Hao
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China
| | - Yiling Li
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bolei Li
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
42
|
Li SY, Guo YL, Tian JW, Zhang HJ, Li RF, Gong P, Yu ZL. Anti-Tumor Strategies by Harnessing the Phagocytosis of Macrophages. Cancers (Basel) 2023; 15:2717. [PMID: 37345054 DOI: 10.3390/cancers15102717] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Macrophages are essential for the human body in both physiological and pathological conditions, engulfing undesirable substances and participating in several processes, such as organism growth, immune regulation, and maintenance of homeostasis. Macrophages play an important role in anti-bacterial and anti-tumoral responses. Aberrance in the phagocytosis of macrophages may lead to the development of several diseases, including tumors. Tumor cells can evade the phagocytosis of macrophages, and "educate" macrophages to become pro-tumoral, resulting in the reduced phagocytosis of macrophages. Hence, harnessing the phagocytosis of macrophages is an important approach to bolster the efficacy of anti-tumor treatment. In this review, we elucidated the underlying phagocytosis mechanisms, such as the equilibrium among phagocytic signals, receptors and their respective signaling pathways, macrophage activation, as well as mitochondrial fission. We also reviewed the recent progress in the area of application strategies on the basis of the phagocytosis mechanism, including strategies targeting the phagocytic signals, antibody-dependent cellular phagocytosis (ADCP), and macrophage activators. We also covered recent studies of Chimeric Antigen Receptor Macrophage (CAR-M)-based anti-tumor therapy. Furthermore, we summarized the shortcomings and future applications of each strategy and look into their prospects with the hope of providing future research directions for developing the application of macrophage phagocytosis-promoting therapy.
Collapse
Affiliation(s)
- Si-Yuan Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yong-Lin Guo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jia-Wen Tian
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - He-Jing Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Rui-Fang Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Ping Gong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Anesthesiology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Li Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
43
|
Liu T, Zhu M, Chang X, Tang X, Yuan P, Tian R, Zhu Z, Zhang Y, Chen X. Tumor-Specific Photothermal-Therapy-Assisted Immunomodulation via Multiresponsive Adjuvant Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300086. [PMID: 36782382 DOI: 10.1002/adma.202300086] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/05/2023] [Indexed: 05/05/2023]
Abstract
Multiresponsive adjuvant nanoparticles (RMmAGL) are fabricated to perform tumor-specific photothermal therapy while regulating the behavior of tumor-associated immune cells for primary tumor eradication and metastasis inhibition. Core-satellite-like RMmAGL have a core of mannose-functionalized mesoporous silica nanoparticles loaded with the TLR7 agonist imiquimod (R837@MSN-mannose) connected via hydrazone bonds to satellites of glutamine (Glu)- and lysine (Lys)-comodified gold nanoparticles (AuNPs-Glu/Lys). During therapy, the acidic environment in tumor tissue cleaves the hydrazone bonds to release AuNPs-Glu/Lys, which further accumulate in tumor cells. After internalization, photothermal agents (aggregated AuNPs-Glu/Lys) are generated in situ through the intratumoral enzyme-catalyzed reaction between Glu and Lys, resulting in tumor-specific photothermal therapy. The detachment of AuNPs-Glu/Lys also triggers the release of R837, which matured dendritic cells (DCs) via a vaccine-like mechanism along with the tumor-associated antigens generated by photothermal therapy. These matured DCs further activates surrounding T cells for immunotherapy. Moreover, the resulting free MSN-mannose serves as an artificial glycocalyx to continuously induce the polarization of tumor-associated macrophages from an immunosuppressive phenotype to an inflammatory phenotype, thus further enhancing immunotherapy. Both in vivo and in vitro experiments demonstrate significant inhibition of malignant tumors after therapy.
Collapse
Affiliation(s)
- Tao Liu
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xiaowei Chang
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xiaoyu Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Pingyun Yuan
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Ran Tian
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
44
|
Wang F, Pu K, Li J. Activating Nanomedicines with Electromagnetic Energy for Deep-Tissue Induction of Immunogenic Cell Death in Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201083. [PMID: 36316270 DOI: 10.1002/smtd.202201083] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Indexed: 05/17/2023]
Abstract
Immunotherapy is an attractive approach for cancer therapy, while its antitumor efficacy is still limited, especially for non-immunogenic tumors. Nanomedicines can be utilized to convert the non-immunogenic "cold" tumors to immunogenic "hot" tumors via inducing immunogenic cell death (ICD), thereby promoting the antitumor immune response. Some nanomedicines that can produce local heat and reactive oxygen species upon the stimulation of electromagnetic energy are the main candidates for inducing the ICD effect. However, their applications are often restricted due to the poor tissue penetration depths of electromagnetic energy, such as light. By contrast, ultrasound, X-ray, alternating magnetic field, and microwave show excellent tissue penetration depths and thereby can be used for sonodynamic therapy, radiotherapy, magnetic hyperthermia therapy, and microwave ablation therapy, all of which can effectively induce ICD. Herein, the combination of deep-tissue electromagnetic energy with nanomedicines for inducing ICD and cancer immunotherapy are summarized. In particular, the designs of nanomedicines to amplify ICD effect in the presence of deep-tissue electromagnetic energy and sensitize tumors to various immunotherapies will be discussed. At the end of this review, a brief conclusion and discussion of current challenges and further perspectives in this subfield are provided.
Collapse
Affiliation(s)
- Fengshuo Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
45
|
Du L, Tang L, Xiao L, Tang K, Zeng Z, Liang Y, Guo Y. Increased expression of CSF1 in patients with eosinophilic asthma. Immun Inflamm Dis 2023; 11:e847. [PMID: 37249291 PMCID: PMC10170305 DOI: 10.1002/iid3.847] [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: 12/30/2022] [Revised: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND The link between colony-stimulating factor 1 (CSF1) and asthma was reported recently. However, the role and mechanism of CSF1 in asthma remain poorly understood. In this study, we aimed to explore the expression and its potential mechanism of CSF1 in asthma. METHODS CSF1 expression in the airway samples from asthmatics and healthy controls were examined, then the correlations between CSF1 and eosinophilic indicators were analyzed. Subsequently, bronchial epithelial cells (BEAS-2B) with CSF1 overexpression and knockdown were constructed to investigate the potential molecular mechanism of CSF1. Finally, the effect of CSF1R inhibitor on STAT1 was investigated. RESULTS The expression of CSF1 was significantly increased in patients with asthma compared to healthy controls, especially in patients with severe and eosinophilic asthma. Upregulated CSF1 positively correlated with airway-increased eosinophil inflammation. In vitro, cytokines interleukin 13 (IL-13) and IL-33 can stimulate the upregulation of CSF1 expression. CSF1 overexpression enhanced p-CSF1R/CSF1R and p-STAT1/STAT1 expression, while knockdown CSF1 using anti-CSF1 siRNAs decreased p-CSF1R/CSF1R and p-STAT1/STAT1 expression. Furthermore, the inhibitor of CSF1R significantly decreased p-STAT1/STAT1 expression. CONCLUSIONS Sputum CSF1 may be involved in asthmatic airway eosinophil inflammation by interacting with CSF1R and further activating the STAT1 signaling. Interfering this potential pathway could serve as an anti-inflammatory therapy for asthma.
Collapse
Affiliation(s)
- Lijuan Du
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Lu Tang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Lisha Xiao
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Kun Tang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Zhimin Zeng
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yuxia Liang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yubiao Guo
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
46
|
Zhao C, Cheng Y, Huang P, Wang C, Wang W, Wang M, Shan W, Deng H. X-ray-Guided In Situ Genetic Engineering of Macrophages for Sustained Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208059. [PMID: 36527738 DOI: 10.1002/adma.202208059] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/22/2022] [Indexed: 06/17/2023]
Abstract
Effective repolarization of macrophages has emerged as a promising approach for anticancer therapy. However, there are very few studies on the effect of reprogramming macrophages from M2 phenotype to M1 phenotype without reconversion while maintaining an activated M1 phenotype. Moreover, these immunomodulatory methods have serious drawbacks due to the activation of normal monocytic cells. Therefore, it remains a challenge to selectively reprogram tumor-associated macrophages (TAMs) without systemic toxicities. Here, X-ray-guided and triggered remote control of a CRISPR/Cas9 genome editing system (X-CC9) that exclusively activates therapeutic agents at tumor sites is established. Under X-ray irradiation, X-CC9 selectively enhances M2-to-M1 repolarization within the tumor microenvironment, and significantly improves antitumor efficacy with robust immune responses in two animal models. This strategy provides an ideal method for improving the safety of macrophage polarization and may constitute a promising immunotherapy strategy.
Collapse
Affiliation(s)
- Caiyan Zhao
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Yaya Cheng
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Pei Huang
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Changrong Wang
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Weipeng Wang
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Mengjiao Wang
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Wenbo Shan
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Hongzhang Deng
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
47
|
Szegvari G, Dora D, Lohinai Z. Effective Reversal of Macrophage Polarization by Inhibitory Combinations Predicted by a Boolean Protein–Protein Interaction Model. BIOLOGY 2023; 12:biology12030376. [PMID: 36979068 PMCID: PMC10045914 DOI: 10.3390/biology12030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Background: The function and polarization of macrophages has a significant impact on the outcome of many diseases. Targeting tumor-associated macrophages (TAMs) is among the greatest challenges to solve because of the low in vitro reproducibility of the heterogeneous tumor microenvironment (TME). To create a more comprehensive model and to understand the inner workings of the macrophage and its dependence on extracellular signals driving polarization, we propose an in silico approach. Methods: A Boolean control network was built based on systematic manual curation of the scientific literature to model the early response events of macrophages by connecting extracellular signals (input) with gene transcription (output). The network consists of 106 nodes, classified as 9 input, 75 inner and 22 output nodes, that are connected by 217 edges. The direction and polarity of edges were manually verified and only included in the model if the literature plainly supported these parameters. Single or combinatory inhibitions were simulated mimicking therapeutic interventions, and output patterns were analyzed to interpret changes in polarization and cell function. Results: We show that inhibiting a single target is inadequate to modify an established polarization, and that in combination therapy, inhibiting numerous targets with individually small effects is frequently required. Our findings show the importance of JAK1, JAK3 and STAT6, and to a lesser extent STK4, Sp1 and Tyk2, in establishing an M1-like pro-inflammatory polarization, and NFAT5 in creating an anti-inflammatory M2-like phenotype. Conclusions: Here, we demonstrate a protein–protein interaction (PPI) network modeling the intracellular signalization driving macrophage polarization, offering the possibility of therapeutic repolarization and demonstrating evidence for multi-target methods.
Collapse
Affiliation(s)
- Gabor Szegvari
- Translational Medicine Institute, Semmelweis University, 1094 Budapest, Hungary
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1094 Budapest, Hungary
- Correspondence: (D.D.); (Z.L.); Tel.: +36-1-2156920 (D.D.)
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, 1094 Budapest, Hungary
- Pulmonary Hospital Torokbalint, 2045 Torokbalint, Hungary
- Correspondence: (D.D.); (Z.L.); Tel.: +36-1-2156920 (D.D.)
| |
Collapse
|
48
|
Zhan C, Jin Y, Xu X, Shao J, Jin C. Antitumor therapy for breast cancer: Focus on tumor-associated macrophages and nanosized drug delivery systems. Cancer Med 2023. [PMID: 36794651 DOI: 10.1002/cam4.5489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/15/2022] [Accepted: 11/17/2022] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND In breast cancer (BC), tumor-associated macrophages (TAMs) are an important component of the tumor microenvironment and are closely related to poor prognosis. A growing number of studies have focused on the role of TAMs in BC progression and therapeutic strategies targeting TAMs. As an emerging treatment, the application of nanosized drug delivery systems (NDDSs) in the treatment of BC by targeting TAMs has attracted much attention. AIMS This review is to summarize the characteristics and treatment strategies targeting TAMs in BC and to clarify the applications of NDDSs targeting TAMs in the treatment of BC by targeting TAMs. MATERIALS & METHODS The existing results related to characteristics of TAMs in BC, BC treatment strategies by targeting TAMs, and the applications of NDDSs in these strategies are described. Through analyzing these results, the advantages and disadvantages of the treatment strategies using NDDSs are discussed, which could provide advices on designing NDDSs for BC treatment. RESULTS TAMs are one of the most prominent noncancer cell types in BC. TAMs not only promote angiogenesis, tumor growth and metastasis but also lead to therapeutic resistance and immunosuppression. Mainly four strategies have been used to target TAMs for BC therapy, which include depleting macrophages, blocking recruitment, reprogramming to attain an anti-tumor phenotype, and increasing phagocytosis. Since NDDSs can efficiently deliver drugs to TAMs with low toxicity, they are promising approaches for targeting TAMs in tumor therapy. NDDSs with various structures can deliver immunotherapeutic agents and nucleic acid therapeutics to TAMs. In addition, NDDSs can realize combination therapies. DISCUSSION TAMs play a critical role in the progression of BC. An increasing number of strategies have been proposed to regulate TAMs. Compared with free drugs, NDDSs targeting TAMs improve drug concentration, reduce toxicity and realize combination therapies. However, in order to achieve better therapeutic efficacy, there are still some disadvantages that need to be considered in the design of NDDSs. CONCLUSION TAMs play an important role in the progression of BC, and targeting TAMs is a promising strategy for BC therapy. In particular, NDDSs targeting TAMs have unique advantages and are potential treatments for BC.
Collapse
Affiliation(s)
- Cuiping Zhan
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ying Jin
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xinzhi Xu
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China.,Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing, China
| | - Jiangbo Shao
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chunxiang Jin
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
49
|
Zhang C, Xu M, He S, Huang J, Xu C, Pu K. Checkpoint Nano-PROTACs for Activatable Cancer Photo-Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208553. [PMID: 36427459 DOI: 10.1002/adma.202208553] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/04/2022] [Indexed: 06/16/2023]
Abstract
Checkpoint immunotherapy holds great potential to treat malignancies via blocking the immunosuppressive signaling pathways, which however suffers from inefficiency and off-target adverse effects. Herein, checkpoint nano-proteolysis targeting chimeras (nano-PROTACs) in combination with photodynamic tumor regression and immunosuppressive protein degradation to block checkpoint signaling pathways for activatable cancer photo-immunotherapy are reported. These nano-PROTACs are composed of a photosensitizer (protoporphyrin IX, PpIX) and an Src homology 2 domain-containing phosphatase 2 (SHP2)-targeting PROTAC peptide (aPRO) via a caspase 3-cleavable segment. aPRO is activated by the increased expression of caspase 3 in tumor cells after phototherapeutic treatment and induces targeted degradation of SHP2 via the ubiquitin-proteasome system. The persistent depletion of SHP2 blocks the immunosuppressive checkpoint signaling pathways (CD47/SIRPα and PD-1/PD-L1), thus reinvigorating antitumor macrophages and T cells. Such a checkpoint PROTAC strategy synergizes immunogenic phototherapy to boost antitumor immune response. Thus, this study represents a generalized PROTAC platform to modulate immune-related signaling pathways for improved anticancer therapy.
Collapse
Affiliation(s)
- Chi Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Shasha He
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Jingsheng Huang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| |
Collapse
|
50
|
Nandi D, Forster J, Ramesh A, Nguyen A, Bharadwaj H, Kulkarni A. Nanoreporter for Real-Time Monitoring of Inflammasome Activity and Targeted Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204900. [PMID: 36603165 PMCID: PMC9951342 DOI: 10.1002/advs.202204900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/18/2022] [Indexed: 06/17/2023]
Abstract
Inflammasome activation is associated with a myriad of inflammatory diseases. However, existing methods provides a limited understanding of spatiotemporal kinetics of inflammasome activation, with restricted scope for early detection of associated treatment efficacy. This limitation offers an opportunity for the development of biocompatible in-vivo inflammasome monitoring tools with translational prospects. To achieve this, they report developing a pair of lipid-based nanoparticle systems, a reporter nanoparticle consisting of a caspase-1 activatable probe alone, and a theranostic nanoparticle combining the probe with an inflammasome-inhibiting drug. This biocompatible platform enhances the probe's residence time in circulation by preventing its opsonization and allowing its sustained release over time. Their results demonstrate the specificity of reporter nanoparticles towards caspase-1 activity and provides early-on monitoring of inflammasome activation both in-vitro as well as in-vivo. Additionally, the delivery of disulfiram, an inflammasome-inhibiting drug, along with reporter probe using theranostic nanoparticles enables real-time tracking of treatment efficacy in the gouty-arthritis inflammatory model. In summary, they report an unparalleled pair of the inflammasome-associated reporter and theranostic platforms suited not only for diagnostic applications but can also detect inflammasome-targeted treatment efficiency in real-time. These findings establish two novel, sensitive nanotools for non-invasive evaluation of inflammasome-targeted immunotherapy.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Department of Veterinary and Animal SciencesUniversity of MassachusettsAmherstMA01003USA
| | - James Forster
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
| | - Anujan Ramesh
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Department of Biomedical EngineeringUniversity of MassachusettsAmherstMA01003USA
| | - Anh Nguyen
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
| | - Hariharan Bharadwaj
- Department of PathologyUMass ChanMedical School‐BaystateSpringfieldMA01107USA
| | - Ashish Kulkarni
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Department of Veterinary and Animal SciencesUniversity of MassachusettsAmherstMA01003USA
- Department of Biomedical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Center for Bioactive DeliveryInstitute for Applied Life SciencesUniversity of MassachusettsAmherstMA01003USA
| |
Collapse
|