1
|
You H, Geng S, Li S, Imani M, Brambilla D, Sun T, Jiang C. Recent advances in biomimetic strategies for the immunotherapy of glioblastoma. Biomaterials 2024; 311:122694. [PMID: 38959533 DOI: 10.1016/j.biomaterials.2024.122694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Immunotherapy is regarded as one of the most promising approaches for treating tumors, with a multitude of immunotherapeutic thoughts currently under consideration for the lethal glioblastoma (GBM). However, issues with immunotherapeutic agents, such as limited in vivo stability, poor blood-brain barrier (BBB) penetration, insufficient GBM targeting, and represented monotherapy, have hindered the success of immunotherapeutic interventions. Moreover, even with the aid of conventional drug delivery systems, outcomes remain suboptimal. Biomimetic strategies seek to overcome these formidable drug delivery challenges by emulating nature's intelligent structures and functions. Leveraging the variety of biological structures and functions, biomimetic drug delivery systems afford a versatile platform with enhanced biocompatibility for the co-delivery of diverse immunotherapeutic agents. Moreover, their inherent capacity to traverse the BBB and home in on GBM holds promise for augmenting the efficacy of GBM immunotherapy. Thus, this review begins by revisiting the various thoughts and agents on immunotherapy for GBM. Then, the barriers to successful GBM immunotherapy are analyzed, and the corresponding biomimetic strategies are explored from the perspective of function and structure. Finally, the clinical translation's current state and prospects of biomimetic strategy are addressed. This review aspires to provide fresh perspectives on the advancement of immunotherapy for GBM.
Collapse
Affiliation(s)
- Haoyu You
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuo Geng
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shangkuo Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mohammad Imani
- Department of Science, Iran Polymer and Petrochemical Institute, Tehran 14977-13115, Iran; Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Tehran 14588-89694, Iran
| | - Davide Brambilla
- Faculty of Pharmacy, University of Montreal, Montreal Quebec H3T 1J4, Canada
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
2
|
Wu H, Cao P, Wang H, Wang W, Yu H, You C, Shen T, Yang S, Hu Z, Zhou T, Wang J, Wang Q, Qian X, Zhang J, Wang X, Cao Y, Ning L, Lin F. Postoperative Injection of a Triptolide-Preloaded Hydrogel Prevents the Recurrence of Glioblastoma by Dual-Pathway Activation of Ferroptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2406036. [PMID: 39375977 DOI: 10.1002/smll.202406036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/14/2024] [Indexed: 10/09/2024]
Abstract
Glioblastoma (GBM) recurrence leads to high mortality, which remains a major concern in clinical therapy. Herein, an injectable triptolide (TP)-preloaded hydrogel (TP@DNH) accompanied by a postoperative injection strategy is developed to prevent the recurrence of GBM. With a potential inhibitor of the NRF2/SLC7A11/GPX4 axis, it is demonstrated that TP can deactivate glutathione peroxidase 4 (GPX4) from the source of glutathione (GSH) biosynthesis, thereby activating ferroptosis in GBM cells by blocking the neutralization of intracellular lipid peroxide (LPO). Based on acid-sensitive Fe3+/tannic acid (TA) metal-phenolic networks (MPNs), the TP@DNH hydrogel can induce the effective generation of reactive oxygen species (ROS) through Fe3+/TA-mediated Fenton reaction and achieve controllable release of TP in resected GBM cavity. Due to ROS generation and GPX4 deactivation, postoperative injection of TP@DNH can achieve high-level ferroptosis through dual-pathway LPO accumulation, remarkably suppressing the growth of recurrent GBM and prolonging the overall survival in orthotopic GBM relapse mouse model. This work provides an alternative paradigm for regulating ferroptosis in the postoperative treatment of GBM.
Collapse
Affiliation(s)
- Hongshuai Wu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Key Laboratory for Multidisciplinary Intersection of Radiotherapy and Immunology for Gastrointestinal Tumor, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, Wuxi, Jiangsu, 214499, P. R. China
| | - Pingping Cao
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Haiyang Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Digestive Endoscopy, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Wenhong Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Hangyang Yu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Chaoqun You
- Jiangsu Key Lab for the Chemistry and Utilization of Agro-Forest Biomass, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Tianqi Shen
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Suisui Yang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Ziyi Hu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Tingting Zhou
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Jing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, 211166, P. R. China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Junxia Zhang
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Neurosurgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Xiuxing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yuandong Cao
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Radiation Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Like Ning
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Fan Lin
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Gastroenterology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, 471023, P. R. China
| |
Collapse
|
3
|
Zhang S, Zhong R, Younis MR, He H, Xu H, Li G, Yang R, Lui S, Wang Y, Wu M. Hydrogel Applications in the Diagnosis and Treatment of Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39366948 DOI: 10.1021/acsami.4c11855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Glioblastoma multiforme (GBM), a common malignant neurological tumor, has boundaries indistinguishable from those of normal tissue, making complete surgical removal ineffective. The blood-brain barrier (BBB) further impedes the efficacy of radiotherapy and chemotherapy, leading to suboptimal treatment outcomes and a heightened probability of recurrence. Hydrogels offer multiple advantages for GBM diagnosis and treatment, including overcoming the BBB for improved drug delivery, controlled drug release for long-term efficacy, and enhanced relaxation properties of magnetic resonance imaging (MRI) contrast agents. Hydrogels, with their excellent biocompatibility and customizability, can mimic the in vivo microenvironment, support tumor cell culture, enable drug screening, and facilitate the study of tumor invasion and metastasis. This paper reviews the classification of hydrogels and recent research for the diagnosis and treatment of GBM, including their applications as cell culture platforms and drugs including imaging contrast agents carriers. The mechanisms of drug release from hydrogels and methods to monitor the activity of hydrogel-loaded drugs are also discussed. This review is intended to facilitate a more comprehensive understanding of the current state of GBM research. It offers insights into the design of integrated hydrogel-based GBM diagnosis and treatment with the objective of achieving the desired therapeutic effect and improving the prognosis of GBM.
Collapse
Affiliation(s)
- Shuaimei Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Renming Zhong
- Radiotherapy Physics & Technology Center, Cancer Center, West China Hospital, Chengdu, Sichuan 610041, P. R. China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Hualong He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Hong Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Ruiyan Yang
- Department of Biology, Macalester College, Saint Paul, Minnesota 55105, United States
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
4
|
Huang T, Bei C, Hu Z, Li Y. CAR-macrophage: Breaking new ground in cellular immunotherapy. Front Cell Dev Biol 2024; 12:1464218. [PMID: 39421021 PMCID: PMC11484238 DOI: 10.3389/fcell.2024.1464218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Chimeric Antigen Receptor (CAR) technology has revolutionized cellular immunotherapy, particularly with the success of CAR-T cells in treating hematologic malignancies. However, CAR-T cells have the limited efficacy of against solid tumors. To address these limitations, CAR-macrophages (CAR-Ms) leverage the innate properties of macrophages with the specificity and potency of CAR technology, offering a novel and promising approach to cancer immunotherapy. Preclinical studies have shown that CAR-Ms can effectively target and destroy tumor cells, even within challenging microenvironments, by exhibiting direct cytotoxicity and enhancing the recruitment and activation of other immune cells. Additionally, the favorable safety profile of macrophages and their persistence within solid tumors position CAR-Ms as potentially safer and more durable therapeutic options compared to CAR-T cells. This review explores recent advancements in CAR-Ms technology, including engineering strategies to optimize their anti-tumor efficacy and preclinical evidence supporting their use. We also discuss the challenges and future directions in developing CAR-Ms therapies, emphasizing their potential to revolutionize cellular immunotherapy. By harnessing the unique properties of macrophages, CAR-Ms offer a groundbreaking approach to overcoming the current limitations of CAR-T cell therapies, paving the way for more effective and sustainable cancer treatments.
Collapse
Affiliation(s)
- Ting Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenqi Bei
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenhua Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
| | - Yuanyuan Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
5
|
Zhang Y, Fang Z, Liu Z, Xi K, Zhang Y, Zhao D, Feng F, Geng H, Liu M, Lou J, Chen C, Zhang Y, Wu Z, Xu F, Jiang X, Ni S. Implantable Microneedle-Mediated Eradication of Postoperative Tumor Foci Mitigates Glioblastoma Relapse. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409857. [PMID: 39205511 DOI: 10.1002/adma.202409857] [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: 07/09/2024] [Revised: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Glioblastoma multiforme (GBM) remains incurable despite multimodal treatments after surgical debulking. Almost all patients with GBM relapse within a narrow margin (2-3 cm) of the initial resected lesion due to the unreachable residual cancerous cells. Here, a completely biodegradable microneedle for surgical cavity delivery glioblastoma-associated macrophages (GAMs)-activating immune nano-stimulator that mitigates glioblastoma relapse is reported. The residual tumor lesion-directed biocompatible microneedle releases the nano-stimulator and toll-like receptor 9 agonist in a controlled manner until the microneedles completely degrade over 1 week, efferently induce in situ phonotypic shifting of GAMs from anti- to pro-inflammatory and the tumor recurrence is obviously inhibited. The implantable microneedles offer a significant improvement over conventional transdermal ones, as they are 100% degradable, ensuring safe application within surgical cavities. It is also revealed that the T cells are recruited to the tumor niche as the GAMs initiate anti-tumor response and eradicate residual GBM cells. Taken together, this work provides a potential strategy for immunomodulating the postoperative tumor niche to mitigate tumor relapse in GBM patients, which may have broad applications in other malignancies with surgical intervention.
Collapse
Affiliation(s)
- Yulin Zhang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key laboratory of chemical Biology, Ministry of education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Zezheng Fang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Zejuan Liu
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Kaiyan Xi
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Yi Zhang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Dawang Zhao
- Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Fan Feng
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Humin Geng
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Minglu Liu
- Bellastem Biotechnology Limited, High-Tech incubator, Intersection of Liquan Street and Gaoxin Er Road, Gaomi, Shandong, 261500, China
| | - Jingzhao Lou
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key laboratory of chemical Biology, Ministry of education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Chen Chen
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key laboratory of chemical Biology, Ministry of education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Yanmin Zhang
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Zimei Wu
- Faculty of Medicine and Health Sciences, School of Pharmacy, University of Auckland, Auckland, 1023, New Zealand
| | - Feng Xu
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xinyi Jiang
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key laboratory of chemical Biology, Ministry of education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| |
Collapse
|
6
|
Xu F, Ni Q, Gong N, Xia B, Zhang J, Guo W, Hu Z, Li J, Liang XJ. Delivery Systems Developed for Treatment Combinations to Improve Adoptive Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407525. [PMID: 39165065 DOI: 10.1002/adma.202407525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/26/2024] [Indexed: 08/22/2024]
Abstract
Adoptive cell therapy (ACT) has shown great success in the clinic for treating hematologic malignancies. However, solid tumor treatment with ACT monotherapy is still challenging, owing to insufficient expansion and rapid exhaustion of adoptive cells, tumor antigen downregulation/loss, and dense tumor extracellular matrix. Delivery strategies for combination cell therapy have great potential to overcome these hurdles. The delivery of vaccines, immune checkpoint inhibitors, cytokines, chemotherapeutics, and photothermal reagents in combination with adoptive cells, have been shown to improve the expansion/activation, decrease exhaustion, and promote the penetration of adoptive cells in solid tumors. Moreover, the delivery of nucleic acids to engineer immune cells directly in vivo holds promise to overcome many of the hurdles associated with the complex ex vivo cell engineering strategies. Here, these research advance, as well as the opportunities and challenges for integrating delivery technologies into cell therapy s are discussed, and the outlook for these emerging areas are criticlly analyzed.
Collapse
Affiliation(s)
- Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qiankun Ni
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Ningqiang Gong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Weisheng Guo
- College of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
7
|
Liu M, Zhou X, Wang Y, Zhao W, Zhao X, Li L, Xue F, Zhang Q, Yan J, Su Y, Zeng W. A Strategy Involving Microporous Microneedles Integrated with CAR-TREM2-Macrophages for Scar Management by Regulating Fibrotic Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2406153. [PMID: 39313983 DOI: 10.1002/adma.202406153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/23/2024] [Indexed: 09/25/2024]
Abstract
Dipeptidyl peptidase 4 (DPP4) positive fibroblasts play a pivotal role in scar development following skin injury. Heterogeneous vascular endothelial cells (ECs) within scarred areas retain the capacity to drive tissue regeneration and repair. Simultaneously, TREM2 macrophages play a crucial role in the progression and resolution of fibrosis by engaging in mutual regulation with ECs. However, effective strategies to inhibit scar formation through multi-factor regulation of the scar microenvironment remain a challenge. Here, CAR-TREM2-macrophages (CAR-TREM2-Ms) capable of targeting DPP4+ fibroblasts and modulating ECs subtype within the scar microenvironment are engineered to effectively prevent scarring. Hydrogel microporous microneedles (mMNs) are employed to deliver CAR-TREM2-Ms, which can effectively alleviate scar. Single-cell transcriptome sequencing (scRNA-seq) analysis reveals that CAR-TREM2-Ms can modify ECs fibrotic phenotype and regulate fibrosis by suppressing the profibrotic gene leucine-rich-alpha-2-glycoprotein 1 (Lrg1). In vitro experiments further demonstrate that CAR-TREM2-Ms improve the scar microenvironment by phagocytosing DPP4+ fibroblasts and suppressing TGFβ secretion. This, in turn, inhibits the phenotypic conversion of LRG1 ECs and provides multifactorial way of alleviating scars. This study uncovers the evidence that mMNs attached to CAR-TREM2-Ms may exert vital influences on skin scarring through the regulation of the skin scar microenvironment, providing a promising approach for treating posttraumatic scarring.
Collapse
Affiliation(s)
- Min Liu
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| | - Xin Zhou
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| | - Yu Wang
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
- Department of Plastic & Cosmetic Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Wenyan Zhao
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| | - Xingli Zhao
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| | - Lang Li
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| | - Fangchao Xue
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| | - Qiao Zhang
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
- Department of Pain and Rehabilitation, Xinqiao Hospital, Army Medical University, Chongqing, 400038, China
| | - Juan Yan
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Yang Su
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| | - Wen Zeng
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
- Jinfeng Laboratory, Chongqing, 401329, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, 400038, China
| |
Collapse
|
8
|
Tataranu LG, Turliuc S, Kamel A, Rizea RE, Dricu A, Staicu GA, Baloi SC, Rodriguez SMB, Manole AIM. Glioblastoma Tumor Microenvironment: An Important Modulator for Tumoral Progression and Therapy Resistance. Curr Issues Mol Biol 2024; 46:9881-9894. [PMID: 39329940 PMCID: PMC11430601 DOI: 10.3390/cimb46090588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/23/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The race to find an effective treatment for glioblastoma (GBM) remains a critical topic, because of its high aggressivity and impact on survival and the quality of life. Currently, due to GBM's high heterogeneity, the conventional treatment success rate and response to therapy are relatively low, with a median survival rate of less than 20 months. A new point of view can be provided by the comprehension of the tumor microenvironment (TME) in pursuance of the development of new therapeutic strategies to aim for a longer survival rate with an improved quality of life and longer disease-free interval (DFI). The main components of the GBM TME are represented by the extracellular matrix (ECM), glioma cells and glioma stem cells (GSCs), immune cells (microglia, macrophages, neutrophils, lymphocytes), neuronal cells, all of them having dynamic interactions and being able to influence the tumoral growth, progression, and drug resistance thus being a potential therapeutic target. This paper will review the latest research on the GBM TME and the potential therapeutic targets to form an up-to-date strategy.
Collapse
Affiliation(s)
- Ligia Gabriela Tataranu
- Neurosurgical Department, University of Medicine and Pharmacy "Carol Davila", 020022 Bucharest, Romania
- Neurosurgical Department, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy "G. T. Popa", 700115 Iasi, Romania
| | - Amira Kamel
- Neurosurgical Department, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Radu Eugen Rizea
- Neurosurgical Department, University of Medicine and Pharmacy "Carol Davila", 020022 Bucharest, Romania
- Neurosurgical Department, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Anica Dricu
- Biochemistry Department, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | | | - Stefania Carina Baloi
- Biochemistry Department, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | | | | |
Collapse
|
9
|
Sun X, Zhou X, Shi X, Abed OA, An X, Lei YL, Moon JJ. Strategies for the development of metalloimmunotherapies. Nat Biomed Eng 2024; 8:1073-1091. [PMID: 38914800 PMCID: PMC11410547 DOI: 10.1038/s41551-024-01221-7] [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: 10/21/2022] [Accepted: 03/30/2024] [Indexed: 06/26/2024]
Abstract
Metal ions play crucial roles in the regulation of immune pathways. In fact, metallodrugs have a long record of accomplishment as effective treatments for a wide range of diseases. Here we argue that the modulation of interactions of metal ions with molecules and cells involved in the immune system forms the basis of a new class of immunotherapies. By examining how metal ions modulate the innate and adaptive immune systems, as well as host-microbiota interactions, we discuss strategies for the development of such metalloimmunotherapies for the treatment of cancer and other immune-related diseases.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Editas Medicine, Cambridge, MA, USA.
| | - Xingwu Zhou
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoyue Shi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Omar A Abed
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Xinran An
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Yu Leo Lei
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Sadri M, Heidari S, Faridzadeh A, Roozbehani M, Toosi S, Mahmoudian RA, Hoseinzadeh A, Salmani Fard MT, Arab FL, Fard SR, Faraji F. Potential applications of macrophages in cancer immunotherapy. Biomed Pharmacother 2024; 178:117161. [PMID: 39047419 DOI: 10.1016/j.biopha.2024.117161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
Immunotherapy has improved cancer treatment based on investigations of tumor immune escape. Manipulation of the immune system stimulates antitumor immune responses and blocks tumor immune escape routes. Genetically adoptive cell therapy, such as T cells, has yielded promising results for hematologic malignancies, but their application to solid tumors has been challenging. Macrophages have a wide broad of capabilities in regulating immune responses, homeostasis, and tissue development, as well as the ability to phagocyte, present antigens, and infiltrate the tumor microenvironment (TME). Given the importance of macrophages in cancer development, they could serve as novel tool for tumor treatment. Therefore, macrophages are used in different formats for direct and indirect targeting of tumor cells. This review summarized the available data on the various applications of macrophages in cancer immunotherapy.
Collapse
Affiliation(s)
- Maryam Sadri
- Department of Immunology, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O. Box: 1449614535, Tehran, Iran.
| | - Sahel Heidari
- Department of Immunology, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O. Box: 1449614535, Tehran, Iran.
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad 1313199137, Iran.
| | - Mona Roozbehani
- Vaccine Research Center, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O. Box: 1449614535, Tehran, Iran.
| | - Shirin Toosi
- Stem Cell and Regenerative Medicine Center, Mashhad University of Medical Science, Mashhad 1313199137, Iran.
| | | | - Akram Hoseinzadeh
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 3513119111, Iran.
| | - Mohammad Taha Salmani Fard
- School of Biology, College of Science, University of Tehran, Faculty of Sciences, Enqelab Square, Tehran 1417614411, Iran.
| | - Fahimeh Lavi Arab
- Immunology Research center, Mashhad University of Medical Sciences, Mashhad 1313199137, Iran.
| | - Soheil Rahmani Fard
- Antimicrobial Resistance Research Center, Institute of Immunology and Infection Diseases Iran University of Medical Sciences, Floor 3, Building no. 3, Hazrat-e Rasool General Hospital, Niyayesh St, Sattar Khan St, P.O. Box: 1445613131, Tehran, Iran.
| | - Fatemeh Faraji
- Antimicrobial Resistance Research Center, Institute of Immunology and Infection Diseases Iran University of Medical Sciences, Floor 3, Building no. 3, Hazrat-e Rasool General Hospital, Niyayesh St, Sattar Khan St, P.O. Box: 1445613131, Tehran, Iran.
| |
Collapse
|
11
|
Li C, Niu C, Chen L, Yu B, Luo F, Qie J, Yang H, Qian J, Chu Y. Classical Monocytes Shuttling for Precise Delivery of Nanotherapeutics to Glioblastoma. Adv Healthc Mater 2024:e2400925. [PMID: 39212635 DOI: 10.1002/adhm.202400925] [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: 03/11/2024] [Revised: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor for which current therapies have limited efficacy. Immunosuppression and difficulties in accessing tumors with therapeutic agents are major obstacles for GBM treatments. Classical monocytes (CMs) possess the strongest infiltration among myeloid cells recruited into tumors during tumorigenesis. In this study, CMs are utilized to deliver the small-molecule CUDC-907 encapsulated in nanoparticles (907-NPs@CMs) for GBM therapy. Hitchhiking on CMs enables more 907-NPs to successfully penetrate the blood-brain barrier (BBB) and reach the interior of tumors. Results demonstrate that 907-NPs@CMs significantly improve the survival rates by suppressing tumor growth and reversing the immunosuppression of tumor microenvironment (TME). Furthermore, the high delivery efficiency of CMs reduces the amount of CUDC-907 required for treatments, reducing the physiological toxicity and off-target effects caused by high doses. 907-NPs@CMs is a safe and versatile therapeutic system that provides a platform for targeted drug delivery to tumors and the ability to treat GBM through a combination of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Congwen Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Congyi Niu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lin Chen
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Jingbo Qie
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Hui Yang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jiawen Qian
- Shanghai Fifth People's Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Science, Fudan University, Shanghai, 200030, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, 200032, China
| |
Collapse
|
12
|
Mousavi R, Soltani M, Souri M. Microneedle patch capable of dual drug release for drug delivery to brain tumors. Drug Deliv Transl Res 2024:10.1007/s13346-024-01696-6. [PMID: 39186235 DOI: 10.1007/s13346-024-01696-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 08/27/2024]
Abstract
Primary brain tumors are mostly managed using surgical resection procedures. Nevertheless, in certain cases, a thin layer of tumors may remain outside of the resection process due to the possibility of permanent injury; these residual tumors expose patients to the risk of tumor recurrence. This study has introduced the use of microneedle patches implanted after surgery with a dual-release mechanism for the administration of doxorubicin. The proposed patches possess the capability to administer drugs directly to the residual tumors and initiate chemotherapy immediately following surgical procedures. Three-dimensional simulation of drug delivery to residual tumors in the brain has been performed based on a finite element method. The impact of four important parameters on drug delivery has been investigated, involving the fraction of drug released in the burst phase, the density of microneedles on the patch, the length of microneedles, and the microvascular density of the tumor. The simulation findings indicate that lowering the fraction of drug released in the initial burst phase reduces the maximum average concentration, but the sustained release that continues for a longer period, increasing the bioavailability of free drug. However, the area under curve (AUC) for different release rates remains unchanged due to the fact that an identical dose of drug is supplied in each instance. By increasing the density of microneedles on the patch, concentration accumulation is provided over an extensive region of tumor, which in turn induces more cancer cell death. A comparative analysis of various lengths reveals that longer microneedles facilitate profound penetration into the tumor layers and present better therapeutic response due to extensive area of the tumor which is exposure to chemotherapeutic drugs. Furthermore, high microvascular density, as a characteristic of the tumor microenvironment, is shown to have a significant impact on the blood microvessels drainage of drugs and consequently lower therapeutic response outcome. Our approach offers a computational framework for creating localized drug delivery systems and addressing the challenges related to residual brain tumors.
Collapse
Affiliation(s)
- Robab Mousavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada.
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada.
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada.
- Centre for Sustainable Business, International Business University, Toronto, Canada.
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
13
|
Liu P, Hu Q. Engineering Cells for Cancer Therapy. Acc Chem Res 2024; 57:2358-2371. [PMID: 39093824 DOI: 10.1021/acs.accounts.4c00293] [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: 08/04/2024]
Abstract
ConspectusCells, particularly living cells, serve as natural carriers of bioactive substances. Their inherent low immunogenicity and multifunctionality have garnered significant attention in the realm of disease treatment applications, specifically within the domains of cancer immunotherapy and regenerative tissue repair. Nevertheless, several prominent challenges impede their swift translation into clinical applications, including obstacles related to large-scale production feasibility and high utilization costs. To address these issues comprehensively, researchers have proposed the notion of bionic cells that are synthetically generated through chemical or biosynthetic means to emulate cellular functions and behaviors. However, artificial cell strategies encounter difficulties in fully replicating the intricate functionalities exhibited by living cells while also grappling with the complexities associated with design implementation for clinical translation purposes. The convergence of disciplines has facilitated the reform of living cells through a range of approaches, including chemical-, biological-, genetic-, and materials-based methods. These techniques can be employed to impart specific functions to cells or enhance the efficacy of therapy. For example, cells are engineered through gene transduction, surface modifications, endocytosis of drugs as delivery systems, and membrane fusion. The concept of engineered cells presents a promising avenue for enhancing control over living cells, thereby enhancing therapeutic efficacy while concurrently mitigating toxic side effects and ultimately facilitating the realization of precision medicine.In this Account, we present a comprehensive overview of our recent research advancements in the field of engineered cells. Our work involves the application of biological or chemical engineering techniques to manipulate endogenous cells for therapeutics or drug delivery purposes. For instance, to avoid the laborious process of isolating, modifying, and expanding engineered cells in vitro, we proposed the concept of in situ engineered cells. By applying a hydrogel loaded with nanoparticles carrying edited chimeric antigen receptor (CAR) plasmids within the postoperative cavity of glioma, we successfully targeted tumor-associated macrophages for gene editing, leading to effective tumor recurrence inhibition. Furthermore, leveraging platelet's ability to release microparticles upon activation at injury sites, we modified antiprogrammed death 1 (PD-1) antibodies on their surface to suppress postoperative tumor recurrence and provide immunotherapy for inoperable tumors. Similarly, by exploiting bacteria's active tropism toward sites of inflammation and hypoxia, we delivered protein drugs by engineered bacteria to induce cancer cell death through pyroptosis initiation and immunotherapy strategies. In the final section, we summarize our aforementioned research progress while providing an outlook on cancer therapy and the hurdles for clinical translation with potential solutions or future directions based on the concept of engineered cells.
Collapse
Affiliation(s)
- Peixin Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
14
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
15
|
Xin Q, Chen Y, Sun X, Li R, Wu Y, Huang X. CAR-T therapy for ovarian cancer: Recent advances and future directions. Biochem Pharmacol 2024; 226:116349. [PMID: 38852648 DOI: 10.1016/j.bcp.2024.116349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Ovarian cancer (OC) is a common gynecological tumor with high mortality, which is difficult to control its progression with conventional treatments and is prone to recurrence. Recent studies have identified OC as an immunogenic tumor that can be recognized by the host immune system. Immunotherapy for OC is being evaluated, but approaches such as immune checkpoint inhibitors have limited efficacy, adoptive cell therapy is an alternative therapy, in which CAR(chimeric antigen receptor)-T therapy has been applied to the clinical treatment of hematological malignancies. In addition, CAR-NK and CAR-macrophage (CAR-M) have also shown great potential in the treatment of solid tumors. Here, we discuss recent advances in preclinical and clinical studies of CAR-T for OC treatment, introduce the efforts made by researchers to modify the structure of CAR in order to achieve effective OC immunotherapy, as well as the research status of CAR-NK and CAR-M, and highlight emerging therapeutic opportunities that can be utilized to improve the survival of patients with OC using CAR-based adoptive cell therapy.
Collapse
Affiliation(s)
- Qianling Xin
- Anhui Women and Children's Medical Center, Hefei Maternal and Child Health Hospital, Hefei, China
| | - Yizhao Chen
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Xiaojing Sun
- Anhui Women and Children's Medical Center, Hefei Maternal and Child Health Hospital, Hefei, China
| | - Ruilin Li
- Department of Pharmacy, The Third Affiliated Hospital of Anhui Medical University, Hefei First People's Hospital, Hefei, China.
| | - Yujing Wu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| | - Xuegui Huang
- Anhui Women and Children's Medical Center, Hefei Maternal and Child Health Hospital, Hefei, China.
| |
Collapse
|
16
|
Wei F, Liu H, Wang Y, Li Y, Han S. Engineering macrophages and their derivatives: A new hope for antitumor therapy. Biomed Pharmacother 2024; 177:116925. [PMID: 38878637 DOI: 10.1016/j.biopha.2024.116925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 07/28/2024] Open
Abstract
Macrophages are central to the immune system and are found in nearly all tissues. Recently, the development of therapies based on macrophages has attracted significant interest. These therapies utilize macrophages' key roles in immunity, their ability to navigate biological barriers, and their tendency to accumulate in tumors. This review explores the advancement of macrophage-based treatments. We discuss the bioengineering of macrophages for improved anti-tumor effects, the use of CAR macrophage therapy for targeting cancer cells, and macrophages as vehicles for therapeutic delivery. Additionally, we examine engineered macrophage products, like extracellular vesicles and membrane-coated nanoparticles, for their potential in precise and less toxic tumor therapy. Challenges in moving these therapies from research to clinical practice are also highlighted. The aim is to succinctly summarize the current status, challenges, and future directions of engineered macrophages in cancer therapy.
Collapse
Affiliation(s)
- Fang Wei
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China
| | - Haiyang Liu
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China
| | - Yuxiao Wang
- Anesthesia Department, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China
| | - Yan Li
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China.
| | - Shuo Han
- Department of Cardiology, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China.
| |
Collapse
|
17
|
Qiu H, Shao Z, Wen X, Qu D, Liu Z, Chen Z, Zhang X, Ding X, Zhang L. HMGB1/TREM2 positive feedback loop drives the development of radioresistance and immune escape of glioblastoma by regulating TLR4/Akt signaling. J Transl Med 2024; 22:688. [PMID: 39075517 PMCID: PMC11287841 DOI: 10.1186/s12967-024-05489-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/04/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Radioresistance and immune escape are crucial reasons for unsatisfactory therapeutic effects of glioblastoma (GBM). Although triggering receptor expressed on myeloid cells-2 (TREM2) involved in forming immunosuppressive microenvironment, but the underlying mechanism and its roles in mediating cancer radioresistance remain unclear, moreover, the efficient delivery of drugs targeting TREM2 to GBM encounters serious challenges. Hence, this study aimed to elucidate the effect and mechanisms of targeted TREM2 silencing on reversing the radioresistance and immune escape of GBM aided by a glutathione-responsive biomimetic nanoparticle (NP) platform. METHODS Radioresistant GBM cell lines and TREM2 stable knockdown GBM cell lines were firstly established. RNA sequencing, colony formation assay, western blot, enzyme-linked immunosorbent assay and co-immunoprecipitation assay were used to detect the molecular mechanisms of TREM2 in regulating the radioresistance and immune escape of GBM. The glutathione-responsive biomimetic NP, angiopep-2 (A2)- cell membrane (CM)-NP/siTREM2/spam1, was then constructed to triply and targeted inhibit TREM2 for in vivo study. Orthotopic GBM-bearing mouse models were established to evaluate the anti-GBM effect of TREM2 inhibition, multiplex immunofluorescence assay was conducted to detect the infiltration of immune cells. RESULTS TREM2 was a regulator in accelerating the radioresistance and immune escape of GBM through participating in DNA damage repair and forming a positive feedback loop with high mobility group box 1 (HMGB1) to cascade the activation of Toll-like receptor 4 (TLR4)/protein kinase B (Akt) signaling. A2-CM-NP/siTREM2/spam1 was successfully synthesized with excellent passive targeting, active targeting and homologous targeting, and the in vivo results exhibited its remarkable anti-GBM therapeutic effect through promoting the infiltration of type 1 helper T cells and CD8+T cells, reducing the infiltration of type 2 helper T cells and regulatory T cells, repolarizing macrophages to M1-type, and decreasing the secretion of pro-tumor and immunosuppressive cytokines. CONCLUSIONS Targeting TREM2 therapy is a promising avenue for optimizing radiotherapy and immunotherapy to improve the prognosis of GBM patients.
Collapse
Affiliation(s)
- Hui Qiu
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China
| | - Zhiying Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Xin Wen
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China
| | - Debao Qu
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China
| | - Zhengyang Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Ziqin Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Xinyan Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Xin Ding
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China.
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China.
| | - Longzhen Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China.
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China.
| |
Collapse
|
18
|
Tsiverioti CA, Gottschlich A, Trefny M, Theurich S, Anders HJ, Kroiss M, Kobold S. Beyond CAR T cells: exploring alternative cell sources for CAR-like cellular therapies. Biol Chem 2024; 405:485-515. [PMID: 38766710 DOI: 10.1515/hsz-2023-0317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/18/2024] [Indexed: 05/22/2024]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has led to remarkable clinical outcomes in the treatment of hematological malignancies. However, challenges remain, such as limited infiltration into solid tumors, inadequate persistence, systemic toxicities, and manufacturing insufficiencies. The use of alternative cell sources for CAR-based therapies, such as natural killer cells (NK), macrophages (MΦ), invariant Natural Killer T (iNKT) cells, γδT cells, neutrophils, and induced pluripotent stem cells (iPSC), has emerged as a promising avenue. By harnessing these cells' inherent cytotoxic mechanisms and incorporating CAR technology, common CAR-T cell-related limitations can be effectively mitigated. We herein present an overview of the tumoricidal mechanisms, CAR designs, and manufacturing processes of CAR-NK cells, CAR-MΦ, CAR-iNKT cells, CAR-γδT cells, CAR-neutrophils, and iPSC-derived CAR-cells, outlining the advantages, limitations, and potential solutions of these therapeutic strategies.
Collapse
Affiliation(s)
| | - Adrian Gottschlich
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Lindwurmstr. 2a, 80337 Munich, Germany
- Department of Medicine III, University Hospital, LMU Munich, Marchioninstr. 15, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), LMU Munich, Pettenkoferstr. 8a, 80336 Munich, Germany
| | - Marcel Trefny
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Lindwurmstr. 2a, 80337 Munich, Germany
| | - Sebastian Theurich
- Department of Medicine III, University Hospital, LMU Munich, Marchioninstr. 15, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), LMU Munich, Pettenkoferstr. 8a, 80336 Munich, Germany
- 74939 German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University Hospital of the LMU , Marchioninstr. 15, 81377 Munich, Germany
- Cancer and Immunometabolism Research Group, 74939 Gene Center LMU , Feodor-Lynen Str. 25, 81377 Munich, Germany
| | - Hans-Joachim Anders
- Department of Medicine IV, University Hospital, LMU Munich, Ziemssenstr. 5, 80336 Munich, Germany
| | - Matthias Kroiss
- Department of Medicine IV, University Hospital, LMU Munich, Ziemssenstr. 5, 80336 Munich, Germany
- Division of Endocrinology and Diabetes, Department of Medicine, University Hospital, University of Würzburg, Josef-Schneider-Str, 9780 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Josef-Schneider-Str. 6, 9780 Würzburg, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Lindwurmstr. 2a, 80337 Munich, Germany
- 74939 German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University Hospital of the LMU , Marchioninstr. 15, 81377 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| |
Collapse
|
19
|
Afzal A, Afzal Z, Bizink S, Davis A, Makahleh S, Mohamed Y, Coniglio SJ. Phagocytosis Checkpoints in Glioblastoma: CD47 and Beyond. Curr Issues Mol Biol 2024; 46:7795-7811. [PMID: 39194679 DOI: 10.3390/cimb46080462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/06/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the deadliest human cancers with very limited treatment options available. The malignant behavior of GBM is manifested in a tumor which is highly invasive, resistant to standard cytotoxic chemotherapy, and strongly immunosuppressive. Immune checkpoint inhibitors have recently been introduced in the clinic and have yielded promising results in certain cancers. GBM, however, is largely refractory to these treatments. The immune checkpoint CD47 has recently gained attention as a potential target for intervention as it conveys a "don't eat me" signal to tumor-associated macrophages (TAMs) via the inhibitory SIRP alpha protein. In preclinical models, the administration of anti-CD47 monoclonal antibodies has shown impressive results with GBM and other tumor models. Several well-characterized oncogenic pathways have recently been shown to regulate CD47 expression in GBM cells and glioma stem cells (GSCs) including Epidermal Growth Factor Receptor (EGFR) beta catenin. Other macrophage pathways involved in regulating phagocytosis including TREM2 and glycan binding proteins are discussed as well. Finally, chimeric antigen receptor macrophages (CAR-Ms) could be leveraged for greatly enhancing the phagocytosis of GBM and repolarization of the microenvironment in general. Here, we comprehensively review the mechanisms that regulate the macrophage phagocytosis of GBM cells.
Collapse
Affiliation(s)
- Amber Afzal
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Zobia Afzal
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Sophia Bizink
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Amanda Davis
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Sara Makahleh
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Yara Mohamed
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Salvatore J Coniglio
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
- Department of Biological Sciences, Kean University, Union, NJ 07083, USA
| |
Collapse
|
20
|
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
|
21
|
Tang Y, Wang Y, Gao Z, Li J, Zhang L, Shi H, Dong J, Song S, Qian C. sAPPα Peptide Promotes Damaged Microglia to Clear Alzheimer's Amyloid-β via Restoring Mitochondrial Function. Chemistry 2024; 30:e202400870. [PMID: 38736169 DOI: 10.1002/chem.202400870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease with amyloid-β (Aβ) deposition as the main pathological feature. It's an important challenge to find new ways to clear Aβ from the brain. The soluble amyloid precursor protein α (sAPPα) is a neuroprotective protein and can attenuate neuronal damage, including toxic Aβ. However, the regulatory role of sAPPα in non-neuronal cells, such as microglia, is less reported and controversial. Here, we showed that sAPPα promoted the phagocytosis and degradation of Aβ in both normal and damaged microglia. Moreover, the function of damaged microglia was improved by the sAPPα through normalizing mitochondrial function. Furthermore, the results of molecular docking simulation showed that sAPPα had a good affinity with Aβ. We preliminarily reveal that sAPPα is similar to antibodies and can participate in the regulation of microglia phagocytosis and degradation of Aβ after binding to Aβ. sAPPα is expected to be a mild and safe peptide drug or drug carrier for AD.
Collapse
Affiliation(s)
- Yingqi Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Yangang Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Ziran Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Jiayi Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Lijia Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Haoting Shi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Jingwen Dong
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Shipeng Song
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Chenggen Qian
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| |
Collapse
|
22
|
Shen J, Lyu S, Xu Y, Zhang S, Li L, Li J, Mou J, Xie L, Tang K, Wen W, Peng X, Yang Y, Shi Y, Li X, Wang M, Li X, Wang J, Cheng T. Activating innate immune responses repolarizes hPSC-derived CAR macrophages to improve anti-tumor activity. Cell Stem Cell 2024; 31:1003-1019.e9. [PMID: 38723634 DOI: 10.1016/j.stem.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 03/18/2024] [Accepted: 04/16/2024] [Indexed: 07/08/2024]
Abstract
Generation of chimeric antigen receptor macrophages (CAR-Ms) from human pluripotent stem cells (hPSCs) offers new prospects for cancer immunotherapy but is currently challenged by low differentiation efficiency and limited function. Here, we develop a highly efficient monolayer-based system that can produce around 6,000 macrophages from a single hPSC within 3 weeks. Based on CAR structure screening, we generate hPSC-CAR-Ms with stable CAR expression and potent tumoricidal activity in vitro. To overcome the loss of tumoricidal activity of hPSC-CAR-Ms in vivo, we use interferon-γ and monophosphoryl lipid A to activate an innate immune response that repolarizes the hPSC-CAR-Ms to tumoricidal macrophages. Moreover, through combined activation of T cells by hPSC-CAR-Ms, we demonstrate that activating a collaborative innate-adaptive immune response can further enhance the anti-tumor effect of hPSC-CAR-Ms in vivo. Collectively, our study provides feasible methodologies that significantly improve the production and function of hPSC-CAR-Ms to support their translation into clinical applications.
Collapse
Affiliation(s)
- Jun Shen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China.
| | - Shuzhen Lyu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China
| | - Shuo Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Li Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China; School of Medicine, Nankai University, Tianjin 300071, China
| | - Jinze Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China; School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Junli Mou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China
| | - Leling Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China
| | - Wei Wen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China
| | - Xuemei Peng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China
| | - Ying Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Xinjie Li
- School of Medicine, Sun Yat-sen University, Guangzhou 510006, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China
| | - Xin Li
- School of Medicine, Sun Yat-sen University, Guangzhou 510006, China.
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China.
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China; Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin 300020, China; Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin 300020, China.
| |
Collapse
|
23
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
24
|
Ye S, Yang B, Yang L, Wei W, Fu M, Yan Y, Wang B, Li X, Liang C, Zhao W. Stemness subtypes in lower-grade glioma with prognostic biomarkers, tumor microenvironment, and treatment response. Sci Rep 2024; 14:14758. [PMID: 38926605 PMCID: PMC11208487 DOI: 10.1038/s41598-024-65717-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024] Open
Abstract
Our research endeavors are directed towards unraveling the stem cell characteristics of lower-grade glioma patients, with the ultimate goal of formulating personalized treatment strategies. We computed enrichment stemness scores and performed consensus clustering to categorize phenotypes. Subsequently, we constructed a prognostic risk model using weighted gene correlation network analysis (WGCNA), random survival forest regression analysis as well as full subset regression analysis. To validate the expression differences of key genes, we employed experimental methods such as quantitative Polymerase Chain Reaction (qPCR) and assessed cell line proliferation, migration, and invasion. Three subtypes were assigned to patients diagnosed with LGG. Notably, Cluster 2 (C2), exhibiting the poorest survival outcomes, manifested characteristics indicative of the subtype characterized by immunosuppression. This was marked by elevated levels of M1 macrophages, activated mast cells, along with higher immune and stromal scores. Four hub genes-CDCA8, ORC1, DLGAP5, and SMC4-were identified and validated through cell experiments and qPCR. Subsequently, these validated genes were utilized to construct a stemness risk signature. Which revealed that Lower-Grade Glioma (LGG) patients with lower scores were more inclined to demonstrate favorable responses to immune therapy. Our study illuminates the stemness characteristics of gliomas, which lays the foundation for developing therapeutic approaches targeting CSCs and enhancing the efficacy of current immunotherapies. By identifying the stemness subtype and its correlation with prognosis and TME patterns in glioma patients, we aim to advance the development of personalized treatments, enhancing the ability to predict and improve overall patient prognosis.
Collapse
Affiliation(s)
- Shengda Ye
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bin Yang
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Liu Yang
- Department of Neurosurgery, Central Theater General Hospital of the Chinese People's Liberation Army, Wuhan, China
| | - Wei Wei
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mingyue Fu
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu Yan
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bo Wang
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiang Li
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Frontier Science Center for Immunology and Metabolism, Wuhan, China.
- Medical Research Institute, Wuhan University, Wuhan, China.
- Sino-Italian Ascula Brain Science Joint Laboratory, Wuhan, China.
| | - Chen Liang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Cancer Hospital of Zhongnan Hospital of Wuhan University, Wuhan, China.
- Cancer Clinical Study Center of Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Tumor Biological Behavior, Wuhan, China.
| | - Wenyuan Zhao
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
25
|
Zhang L, Teng F, Xin H, Xu W, Wu W, Yao C, Wang Z. A Big Prospect for Hydrogel Nano-System in Glioma. Int J Nanomedicine 2024; 19:5605-5618. [PMID: 38882547 PMCID: PMC11179662 DOI: 10.2147/ijn.s470315] [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: 04/17/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024] Open
Abstract
Patients diagnosed with glioma typically face a limited life expectancy (around 15 months on average), a bleak prognosis, and a high likelihood of recurrence. As such, glioma is recognized as a significant form of malignancy. Presently, the treatment options for glioma include traditional approaches such as surgery, chemotherapy, and radiotherapy. Regrettably, the efficacy of these treatments has been less than optimal. Nevertheless, a promising development in glioma treatment lies in the use of hydrogel nano-systems as sophisticated delivery systems. These nano-systems have demonstrated exceptional therapeutic effects in the treatment of glioma by various responsive ways, including temperature-response, pH-response, liposome-response, ROS-response, light-response, and enzyme-response. This study seeks to provide a comprehensive summary of both the therapeutic application of hydrogel nano-systems in managing glioma and the underlying immune action mechanisms.
Collapse
Affiliation(s)
- Lu Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- The State Key Laboratory of Power Transmission Equipment and System Security and New Technology, College of Electrical Engineering, Chongqing University, Chongqing, 400044, People’s Republic of China
| | - Fei Teng
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Huajie Xin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Wei Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Wei Wu
- College of Biological Engineering, Chongqing University, Chongqing, 400030, People’s Republic of China
| | - Chenguo Yao
- The State Key Laboratory of Power Transmission Equipment and System Security and New Technology, College of Electrical Engineering, Chongqing University, Chongqing, 400044, People’s Republic of China
| | - Zhiqiang Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Center of Thoracic Cancer, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| |
Collapse
|
26
|
Greiner D, Xue Q, Waddell TQ, Kurudza E, Belote RL, Dotti G, Judson-Torres RL, Reeves MQ, Cheshier SH, Roh-Johnson M. CSPG4-targeting CAR-macrophages inhibit melanoma growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597413. [PMID: 38895447 PMCID: PMC11185669 DOI: 10.1101/2024.06.04.597413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of hematological malignancies but has been clinically less effective in solid tumors. Engineering macrophages with CARs has emerged as a promising approach to overcome some of the challenges faced by CAR-T cells due to the macrophage's ability to easily infiltrate tumors, phagocytose their targets, and reprogram the immune response. We engineered CAR-macrophages (CAR-Ms) to target chondroitin sulfate proteoglycan 4 (CSPG4), an antigen expressed in melanoma, and several other solid tumors. CSPG4-targeting CAR-Ms exhibited specific phagocytosis of CSPG4-expressing melanoma cells. Combining CSPG4-targeting CAR-Ms with CD47 blocking antibodies synergistically enhanced CAR-M-mediated phagocytosis and effectively inhibited melanoma spheroid growth in 3D. Furthermore, CSPG4-targeting CAR-Ms inhibited melanoma tumor growth in mouse models. These results suggest that CSPG4-targeting CAR-M immunotherapy is a promising solid tumor immunotherapy approach for treating melanoma. STATEMENT OF SIGNIFICANCE We engineered macrophages with CARs as an alternative approach for solid tumor treatment. CAR-macrophages (CAR-Ms) targeting CSPG4, an antigen expressed in melanoma and other solid tumors, phagocytosed melanoma cells and inhibited melanoma growth in vivo . Thus, CSPG4-targeting CAR-Ms may be a promising strategy to treat patients with CSPG4-expressing tumors.
Collapse
|
27
|
Li N, Geng S, Dong ZZ, Jin Y, Ying H, Li HW, Shi L. A new era of cancer immunotherapy: combining revolutionary technologies for enhanced CAR-M therapy. Mol Cancer 2024; 23:117. [PMID: 38824567 PMCID: PMC11143597 DOI: 10.1186/s12943-024-02032-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/24/2024] [Indexed: 06/03/2024] Open
Abstract
Significant advancements have been made in the application of chimeric antigen receptor (CAR)-T treatment for blood cancers during the previous ten years. However, its effectiveness in treating solid tumors is still lacking, necessitating the exploration of alternative immunotherapies that can overcome the significant challenges faced by current CAR-T cells. CAR-based immunotherapy against solid tumors shows promise with the emergence of macrophages, which possess robust phagocytic abilities, antigen-presenting functions, and the ability to modify the tumor microenvironment and stimulate adaptive responses. This paper presents a thorough examination of the latest progress in CAR-M therapy, covering both basic scientific studies and clinical trials. This study examines the primary obstacles hindering the realization of the complete potential of CAR-M therapy, as well as the potential strategies that can be employed to overcome these hurdles. With the emergence of revolutionary technologies like in situ genetic modification, synthetic biology techniques, and biomaterial-supported gene transfer, which provide a wider array of resources for manipulating tumor-associated macrophages, we suggest that combining these advanced methods will result in the creation of a new era of CAR-M therapy that demonstrates improved efficacy, safety, and availability.
Collapse
Affiliation(s)
- Na Li
- Key lab of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China
- Department of Immunology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Shinan Geng
- Key lab of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China
| | - Zhen-Zhen Dong
- Key lab of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ying Jin
- Hangzhou Institute of Medicine (HIM), Zhejiang Caner Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Hangjie Ying
- Hangzhou Institute of Medicine (HIM), Zhejiang Caner Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Hung-Wing Li
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Liyun Shi
- Key lab of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China.
| |
Collapse
|
28
|
Li X, Zhao L, Li W, Gao P, Zhang N. HER2-targeting CAR-T cells show highly efficient anti-tumor activity against glioblastoma both in vitro and in vivo. Genes Immun 2024; 25:201-208. [PMID: 38702509 PMCID: PMC11178492 DOI: 10.1038/s41435-024-00275-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/16/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant primary brain tumor in adults. Current treatment options for GBM include surgical resection, radiation, and chemotherapy, which predominantly slow cancer growth and reduce symptoms, resulting in a 5-year survival rate of no more than 10%. Chimeric antigen receptor (CAR) T-cell therapy is a new class of cellular immunotherapy that has made great progress in treating malignant tumors. Human epidermal growth factor receptor 2 (HER2) is overexpressed in GBM and may provide a potential therapeutic target for GBM treatment. In this study, we constructed third-generation CAR-T cells targeting the HER2 antigen in GBM. HER2-CAR-T cells showed effective anti-tumor activity both in vitro and in vivo. Furthermore, HER2-specific CAR-T cells exhibited strong cytotoxicity and cytokine-secreting abilities against GBM cells in vitro. Anti-HER2 CAR-T cells also exhibited increased cytotoxicity with increasing effector-to-target ratios. Anti-HER2 CAR-T cells delivered via peritumoral injection successfully stunted tumor progression in vivo. Moreover, peritumoral intravenous administration of anti-HER2 CAR-T cells resulted in therapeutic improvement against GBM cells compared with intravenous administration. In conclusion, our study shows that HER2 CAR-T cells represent an emerging immunotherapy for treating GBM.
Collapse
Affiliation(s)
- Xueying Li
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, China
- Research Institute for Microbial Diseases and World Premier International Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Lifen Zhao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, China
| | - Wenzhe Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Peng Gao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, China.
| | - Nianzhu Zhang
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, China.
| |
Collapse
|
29
|
Dai H, Zhu C, Huai Q, Xu W, Zhu J, Zhang X, Zhang X, Sun B, Xu H, Zheng M, Li X, Wang H. Chimeric antigen receptor-modified macrophages ameliorate liver fibrosis in preclinical models. J Hepatol 2024; 80:913-927. [PMID: 38340812 DOI: 10.1016/j.jhep.2024.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND & AIMS Treatments directly targeting fibrosis remain limited. Given the unique intrinsic features of macrophages and their capacity to engraft in the liver, we genetically engineered bone marrow-derived macrophages with a chimeric antigen receptor (CAR) to direct their phagocytic activity against hepatic stellate cells (HSCs) in multiple mouse models. This study aimed to demonstrate the therapeutic efficacy of CAR macrophages (CAR-Ms) in mouse models of fibrosis and cirrhosis and to elucidate the underlying mechanisms. METHODS uPAR expression was studied in patients with fibrosis/cirrhosis and in murine models of liver fibrosis, including mice treated with carbon tetrachloride, a 5-diethoxycarbonyl-1, 4-dihydrocollidine diet, or a high-fat/cholesterol/fructose diet. The safety and efficacy of CAR-Ms were evaluated in vitro and in vivo. RESULTS Adoptive transfer of CAR-Ms resulted in a significant reduction in liver fibrosis and the restoration of function in murine models of liver fibrosis. CAR-Ms modulated the hepatic immune microenvironment to recruit and modify the activation of endogenous immune cells to drive fibrosis regression. These CAR-Ms were able to recruit and present antigens to T cells and mount specific antifibrotic T-cell responses to reduce fibroblasts and liver fibrosis in mice. CONCLUSION Collectively, our findings demonstrate the potential of using macrophages as a platform for CAR technology to provide an effective treatment option for liver fibrosis. CAR-Ms might be developed for treatment of patients with liver fibrosis. IMPACT AND IMPLICATIONS Liver fibrosis is an incurable condition that afflicts millions of people globally. Despite the clear clinical need, therapies for liver fibrosis are limited. Our findings provide the first preclinical evidence that chimeric antigen receptor (CAR)-macrophages (CAR-Ms) targeting uPAR can attenuate liver fibrosis and cirrhosis. We show that macrophages expressing this uPAR CAR exert a direct antifibrotic effect and elicit a specific T-cell response that augments the immune response against liver fibrosis. These findings demonstrate the potential of using CAR-Ms as an effective cell-based therapy for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Hanren Dai
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Cheng Zhu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Qian Huai
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Wentao Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Jiejie Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xu Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Xianzheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Honghai Xu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Minghua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaolei Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China; Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.
| |
Collapse
|
30
|
Bao P, Gu HY, Jiang YC, Wang JW, Wu M, Yu A, Zhong Z, Zhang XZ. In Situ Sprayed Exosome-Cross-Linked Gel as Artificial Lymph Nodes for Postoperative Glioblastoma Immunotherapy. ACS NANO 2024; 18:13266-13276. [PMID: 38709874 DOI: 10.1021/acsnano.4c02425] [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: 05/08/2024]
Abstract
One key challenge in postoperative glioblastoma immunotherapy is to guarantee a potent and durable T-cell response, which is restricted by the immunosuppressive microenvironment within the lymph nodes (LNs). Here, we develop an in situ sprayed exosome-cross-linked gel that acts as an artificial LN structure to directly activate the tumor-infiltrating T cells for prevention of glioma recurrence. Briefly, this gel is generated by a bio-orthogonal reaction between azide-modified chimeric exosomes and alkyne-modified alginate polymers. Particularly, these chimeric exosomes are generated from dendritic cell (DC)-tumor hybrid cells, allowing for direct and robust T-cell activation. The gel structure with chimeric exosomes as cross-linking points avoids the quick clearance by the immune system and thus prolongs the durability of antitumor T-cell immunity. Importantly, this exosome-containing immunotherapeutic gel provides chances for ameliorating functions of antigen-presenting cells (APCs) through accommodating different intracellular-acting adjuvants, such as stimulator of interferon genes (STING) agonists. This further enhances the antitumor T-cell response, resulting in the almost complete elimination of residual lesions after surgery. Our findings provide a promising strategy for postsurgical glioma immunotherapy that warrants further exploration in the clinical arena.
Collapse
Affiliation(s)
- Peng Bao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Hui-Yun Gu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Yuan-Cheng Jiang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Jia-Wei Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Meng Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Zhenlin Zhong
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| |
Collapse
|
31
|
Lin H, Liu C, Hu A, Zhang D, Yang H, Mao Y. Understanding the immunosuppressive microenvironment of glioma: mechanistic insights and clinical perspectives. J Hematol Oncol 2024; 17:31. [PMID: 38720342 PMCID: PMC11077829 DOI: 10.1186/s13045-024-01544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Glioblastoma (GBM), the predominant and primary malignant intracranial tumor, poses a formidable challenge due to its immunosuppressive microenvironment, thereby confounding conventional therapeutic interventions. Despite the established treatment regimen comprising surgical intervention, radiotherapy, temozolomide administration, and the exploration of emerging modalities such as immunotherapy and integration of medicine and engineering technology therapy, the efficacy of these approaches remains constrained, resulting in suboptimal prognostic outcomes. In recent years, intensive scrutiny of the inhibitory and immunosuppressive milieu within GBM has underscored the significance of cellular constituents of the GBM microenvironment and their interactions with malignant cells and neurons. Novel immune and targeted therapy strategies have emerged, offering promising avenues for advancing GBM treatment. One pivotal mechanism orchestrating immunosuppression in GBM involves the aggregation of myeloid-derived suppressor cells (MDSCs), glioma-associated macrophage/microglia (GAM), and regulatory T cells (Tregs). Among these, MDSCs, though constituting a minority (4-8%) of CD45+ cells in GBM, play a central component in fostering immune evasion and propelling tumor progression, angiogenesis, invasion, and metastasis. MDSCs deploy intricate immunosuppressive mechanisms that adapt to the dynamic tumor microenvironment (TME). Understanding the interplay between GBM and MDSCs provides a compelling basis for therapeutic interventions. This review seeks to elucidate the immune regulatory mechanisms inherent in the GBM microenvironment, explore existing therapeutic targets, and consolidate recent insights into MDSC induction and their contribution to GBM immunosuppression. Additionally, the review comprehensively surveys ongoing clinical trials and potential treatment strategies, envisioning a future where targeting MDSCs could reshape the immune landscape of GBM. Through the synergistic integration of immunotherapy with other therapeutic modalities, this approach can establish a multidisciplinary, multi-target paradigm, ultimately improving the prognosis and quality of life in patients with GBM.
Collapse
Affiliation(s)
- Hao Lin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Chaxian Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ankang Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Duanwu Zhang
- Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
32
|
Solomou G, Young AMH, Bulstrode HJCJ. Microglia and macrophages in glioblastoma: landscapes and treatment directions. Mol Oncol 2024. [PMID: 38712663 DOI: 10.1002/1878-0261.13657] [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: 11/28/2023] [Revised: 02/29/2024] [Accepted: 04/19/2024] [Indexed: 05/08/2024] Open
Abstract
Glioblastoma is the most common primary malignant tumour of the central nervous system and remains uniformly and rapidly fatal. The tumour-associated macrophage (TAM) compartment comprises brain-resident microglia and bone marrow-derived macrophages (BMDMs) recruited from the periphery. Immune-suppressive and tumour-supportive TAM cell states predominate in glioblastoma, and immunotherapies, which have achieved striking success in other solid tumours have consistently failed to improve survival in this 'immune-cold' niche context. Hypoxic and necrotic regions in the tumour core are found to enrich, especially in anti-inflammatory and immune-suppressive TAM cell states. Microglia predominate at the invasive tumour margin and express pro-inflammatory and interferon TAM cell signatures. Depletion of TAMs, or repolarisation towards a pro-inflammatory state, are appealing therapeutic strategies and will depend on effective understanding and classification of TAM cell ontogeny and state based on new single-cell and spatial multi-omic in situ profiling. Here, we explore the application of these datasets to expand and refine TAM characterisation, to inform improved modelling approaches, and ultimately underpin the effective manipulation of function.
Collapse
Affiliation(s)
- Georgios Solomou
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, UK
- Department of Neurosurgery, Addenbrooke's Hospital, Cambridge, UK
| | - Adam M H Young
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, UK
- Department of Neurosurgery, Addenbrooke's Hospital, Cambridge, UK
| | - Harry J C J Bulstrode
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, UK
- Department of Neurosurgery, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
33
|
Vazaios K, van Berkum RE, Calkoen FG, van der Lugt J, Hulleman E. OV Modulators of the Paediatric Brain TIME: Current Status, Combination Strategies, Limitations and Future Directions. Int J Mol Sci 2024; 25:5007. [PMID: 38732225 PMCID: PMC11084613 DOI: 10.3390/ijms25095007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Oncolytic viruses (OVs) are characterised by their preference for infecting and replicating in tumour cells either naturally or after genetic modification, resulting in oncolysis. Furthermore, OVs can elicit both local and systemic anticancer immune responses while specifically infecting and lysing tumour cells. These characteristics render them a promising therapeutic approach for paediatric brain tumours (PBTs). PBTs are frequently marked by a cold tumour immune microenvironment (TIME), which suppresses immunotherapies. Recent preclinical and clinical studies have demonstrated the capability of OVs to induce a proinflammatory immune response, thereby modifying the TIME. In-depth insights into the effect of OVs on different cell types in the TIME may therefore provide a compelling basis for using OVs in combination with other immunotherapy modalities. However, certain limitations persist in our understanding of oncolytic viruses' ability to regulate the TIME to enhance anti-tumour activity. These limitations primarily stem from the translational limitations of model systems, the difficulties associated with tracking reliable markers of efficacy throughout the course of treatment and the role of pre-existing viral immunity. In this review, we describe the different alterations observed in the TIME in PBTs due to OV treatment, combination therapies of OVs with different immunotherapies and the hurdles limiting the development of effective OV therapies while suggesting future directions based on existing evidence.
Collapse
Affiliation(s)
| | | | | | | | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (K.V.); (F.G.C.); (J.v.d.L.)
| |
Collapse
|
34
|
Chuang ST, Stein JB, Nevins S, Kilic Bektas C, Choi HK, Ko WK, Jang H, Ha J, Lee KB. Enhancing CAR Macrophage Efferocytosis Via Surface Engineered Lipid Nanoparticles Targeting LXR Signaling. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308377. [PMID: 38353580 PMCID: PMC11081841 DOI: 10.1002/adma.202308377] [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: 08/17/2023] [Revised: 02/05/2024] [Indexed: 02/24/2024]
Abstract
The removal of dying cells, or efferocytosis, is an indispensable part of resolving inflammation. However, the inflammatory microenvironment of the atherosclerotic plaque frequently affects the biology of both apoptotic cells and resident phagocytes, rendering efferocytosis dysfunctional. To overcome this problem, a chimeric antigen receptor (CAR) macrophage that can target and engulf phagocytosis-resistant apoptotic cells expressing CD47 is developed. In both normal and inflammatory circumstances, CAR macrophages exhibit activity equivalent to antibody blockage. The surface of CAR macrophages is modified with reactive oxygen species (ROS)-responsive therapeutic nanoparticles targeting the liver X receptor pathway to improve their cell effector activities. The combination of CAR and nanoparticle engineering activated lipid efflux pumps enhances cell debris clearance and reduces inflammation. It is further suggested that the undifferentiated CAR-Ms can transmigrate within a mico-fabricated vessel system. It is also shown that our CAR macrophage can act as a chimeric switch receptor (CSR) to withstand the immunosuppressive inflammatory environment. The developed platform has the potential to contribute to the advancement of next-generation cardiovascular disease therapies and further studies include in vivo experiments.
Collapse
Affiliation(s)
- Skylar T Chuang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Joshua B Stein
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Sarah Nevins
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Cemile Kilic Bektas
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Hye Kyu Choi
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Wan-Kyu Ko
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Hyunjun Jang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Jihun Ha
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
35
|
Short L, Holt RA, Cullis PR, Evgin L. Direct in vivo CAR T cell engineering. Trends Pharmacol Sci 2024; 45:406-418. [PMID: 38614815 DOI: 10.1016/j.tips.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 04/15/2024]
Abstract
T cells modified to express intelligently designed chimeric antigen receptors (CARs) are exceptionally powerful therapeutic agents for relapsed and refractory blood cancers and have the potential to revolutionize therapy for many other diseases. To circumvent the complexity and cost associated with broad-scale implementation of ex vivo manufactured adoptive cell therapy products, alternative strategies to generate CAR T cells in vivo by direct infusion of nanoparticle-formulated nucleic acids or engineered viral vectors under development have received a great deal of attention in the past few years. Here, we outline the ex vivo manufacturing process as a motivating framework for direct in vivo strategies and discuss emerging data from preclinical models to highlight the potency of the in vivo approach, the applicability for new disease indications, and the remaining challenges associated with clinical readiness, including delivery specificity, long term efficacy, and safety.
Collapse
Affiliation(s)
- Lauralie Short
- Michael Smith Genome Sciences Department, BC Cancer Research Institute, Vancouver, BC, Canada; Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Robert A Holt
- Michael Smith Genome Sciences Department, BC Cancer Research Institute, Vancouver, BC, Canada; Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada; Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Laura Evgin
- Michael Smith Genome Sciences Department, BC Cancer Research Institute, Vancouver, BC, Canada; Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
36
|
Sun S, Yang Q, Jiang D, Zhang Y. Nanobiotechnology augmented cancer stem cell guided management of cancer: liquid-biopsy, imaging, and treatment. J Nanobiotechnology 2024; 22:176. [PMID: 38609981 PMCID: PMC11015566 DOI: 10.1186/s12951-024-02432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent both a key driving force and therapeutic target of tumoral carcinogenesis, tumor evolution, progression, and recurrence. CSC-guided tumor diagnosis, treatment, and surveillance are strategically significant in improving cancer patients' overall survival. Due to the heterogeneity and plasticity of CSCs, high sensitivity, specificity, and outstanding targeting are demanded for CSC detection and targeting. Nanobiotechnologies, including biosensors, nano-probes, contrast enhancers, and drug delivery systems, share identical features required. Implementing these techniques may facilitate the overall performance of CSC detection and targeting. In this review, we focus on some of the most recent advances in how nanobiotechnologies leverage the characteristics of CSC to optimize cancer diagnosis and treatment in liquid biopsy, clinical imaging, and CSC-guided nano-treatment. Specifically, how nanobiotechnologies leverage the attributes of CSC to maximize the detection of circulating tumor DNA, circulating tumor cells, and exosomes, to improve positron emission computed tomography and magnetic resonance imaging, and to enhance the therapeutic effects of cytotoxic therapy, photodynamic therapy, immunotherapy therapy, and radioimmunotherapy are reviewed.
Collapse
Affiliation(s)
- Si Sun
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiang Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
37
|
Huang J, Yang Q, Wang W, Huang J. CAR products from novel sources: a new avenue for the breakthrough in cancer immunotherapy. Front Immunol 2024; 15:1378739. [PMID: 38665921 PMCID: PMC11044028 DOI: 10.3389/fimmu.2024.1378739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has transformed cancer immunotherapy. However, significant challenges limit its application beyond B cell-driven malignancies, including limited clinical efficacy, high toxicity, and complex autologous cell product manufacturing. Despite efforts to improve CAR T cell therapy outcomes, there is a growing interest in utilizing alternative immune cells to develop CAR cells. These immune cells offer several advantages, such as major histocompatibility complex (MHC)-independent function, tumor microenvironment (TME) modulation, and increased tissue infiltration capabilities. Currently, CAR products from various T cell subtypes, innate immune cells, hematopoietic progenitor cells, and even exosomes are being explored. These CAR products often show enhanced antitumor efficacy, diminished toxicity, and superior tumor penetration. With these benefits in mind, numerous clinical trials are underway to access the potential of these innovative CAR cells. This review aims to thoroughly examine the advantages, challenges, and existing insights on these new CAR products in cancer treatment.
Collapse
Affiliation(s)
| | | | - Wen Wang
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Juan Huang
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
38
|
Anas Z, Hasan SFS, Moiz MA, Zuberi MAW, Shah HH, Ejaz A, Dave T, Panjwani MH, Rauf SA, Hussain MS, Waseem R. The role of hydrogels in the management of brain tumours: a narrative review. Ann Med Surg (Lond) 2024; 86:2004-2010. [PMID: 38576913 PMCID: PMC10990399 DOI: 10.1097/ms9.0000000000001809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/29/2024] [Indexed: 04/06/2024] Open
Abstract
Conventional therapeutic techniques for brain tumours have limitations and side effects, necessitating the need for alternative treatment options. MRI-monitored therapeutic hydrogel systems show potential as a non-surgical approach for brain tumour treatment. Hydrogels have unique physical and chemical properties that make them promising for brain tumour treatment, including the ability to encapsulate therapeutic agents, provide sustained and controlled drug release, and overcome the blood-brain barrier for better penetration. By combining hydrogel systems with MRI techniques, it is possible to develop therapeutic approaches that provide real-time monitoring and controlled release of therapeutic agents. Surgical resection remains important, but there is a growing need for alternative approaches that can complement or replace traditional methods. The objective of this comprehensive narrative review is to evaluate the potential of MRI-monitored therapeutic hydrogel systems in non-surgical brain tumour treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tirth Dave
- Bukovinian State Medical University, Chernivtsi, Ukraine
| | | | | | | | | |
Collapse
|
39
|
Yang S, Wang Y, Jia J, Fang Y, Yang Y, Yuan W, Hu J. Advances in Engineered Macrophages: A New Frontier in Cancer Immunotherapy. Cell Death Dis 2024; 15:238. [PMID: 38561367 PMCID: PMC10985090 DOI: 10.1038/s41419-024-06616-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
Macrophages, as pivotal cells within the tumour microenvironment, significantly influence the impact of and reactions to treatments for solid tumours. The rapid evolution of bioengineering technology has revealed the vast potential of engineered macrophages in immunotherapy, disease diagnosis, and tissue engineering. Given this landscape, the goal of harnessing and innovating macrophages as a novel strategy for solid tumour immunotherapy cannot be overstated. The diverse strategies for engineered macrophages in the realm of cancer immunotherapy, encompassing macrophage drug delivery systems, chimeric antigen receptor macrophage therapy, and synergistic treatment approaches involving bacterial outer membrane vesicles and macrophages, are meticulously examined in this review. These methodologies are designed to enhance the therapeutic efficacy of macrophages against solid tumours, particularly those that are drug-resistant and metastatic. Collectively, these immunotherapies are poised to supplement and refine current solid tumour treatment paradigms, thus heralding a new frontier in the fight against malignant tumours.
Collapse
Affiliation(s)
- Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Jiachi Jia
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yingshuai Fang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yabing Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| |
Collapse
|
40
|
Chen Y, Liu C, Fang Y, Chen W, Qiu J, Zhu M, Wei W, Tu J. Developing CAR-immune cell therapy against SARS-CoV-2: Current status, challenges and prospects. Biochem Pharmacol 2024; 222:116066. [PMID: 38373592 DOI: 10.1016/j.bcp.2024.116066] [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: 01/12/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Chimeric antigen receptor (CAR)-immune cell therapy has revolutionized the anti-tumor field, achieving efficient and precise tumor clearance by directly guiding immune cell activity to target tumors. In addition, the use of CAR-immune cells to influence the composition and function of the immune system and ultimately achieve virus clearance and immune system homeostasis has attracted the interest of researchers. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) triggered a global pandemic of coronavirus disease 2019 (COVID-19). To date, the rapidly mutating SARS-CoV-2 continues to challenge existing therapies and has raised public concerns regarding reinfection. In patients with COVID-19, the interaction of SARS-CoV-2 with the immune system influences the course of the disease, and the coexistence of over-activated immune system components, such as macrophages, and severely compromised immune system components, such as natural killer cells, reveals a dysregulated immune system. Dysregulated immune-induced inflammation may impair viral clearance and T-cell responses, causing cytokine storms and ultimately leading to patient death. Here, we summarize the research progress on the use of CAR-immune cells against SARS-CoV-2 infection. Furthermore, we discuss the feasibility, challenges and prospect of CAR-immune cells as a new immune candidate therapy against SARS-CoV-2.
Collapse
Affiliation(s)
- Yizhao Chen
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Chong Liu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Yilong Fang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Weile Chen
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Jiaqi Qiu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Mengjuan Zhu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
| | - Jiajie Tu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
41
|
Nie S, Qin Y, Ou L, Chen X, Li L. In Situ Reprogramming of Immune Cells Using Synthetic Nanomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310168. [PMID: 38229527 DOI: 10.1002/adma.202310168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/12/2024] [Indexed: 01/18/2024]
Abstract
In the past decade, adoptive cell therapy with chimeric antigen receptor-T (CAR-T) cells has revolutionized cancer treatment. However, the complexity and high costs involved in manufacturing current adoptive cell therapy greatly inhibit its widespread availability and access. To address this, in situ cell therapy, which directly reprograms immune cells inside the body, has recently been developed as a promising alternative. Here, an overview of the recent progress in the development of synthetic nanomaterials is provided to deliver plasmid DNA or mRNA for in situ reprogramming of T cells and macrophages, focusing especially on in situ CAR therapies. Also, the main challenges for in situ immune cell reprogramming are discussed and some approaches to overcome these barriers to fulfill the clinical applications are proposed.
Collapse
Affiliation(s)
- Shihong Nie
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuyang Qin
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Liyuan Ou
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, 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, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Ling Li
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
42
|
Wu B, Liang J, Yang X, Fang Y, Kong N, Chen D, Wang H. A Programmable Peptidic Hydrogel Adjuvant for Personalized Immunotherapy in Resected Stage Tumors. J Am Chem Soc 2024; 146:8585-8597. [PMID: 38478659 DOI: 10.1021/jacs.4c00569] [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: 03/28/2024]
Abstract
Adjuvant treatment after surgical resection usually plays an important role in delaying disease recurrence. Immunotherapy displays encouraging results in increasing patients' chances of staying cancer-free after surgery, as reported by recent clinical trials. However, the clinical outcomes of current immunotherapy need to be improved due to the limited responses, patient heterogeneity, nontargeted distribution, and immune-related adverse effects. This work describes a programmable hydrogel adjuvant for personalized immunotherapy after surgical resection. By filling the hydrogel in the cavity, this system aims to address the limited secretion of granzyme B (GrB) during immunotherapy and improve the low immunotherapy responses typically observed, while minimizing immune-related side effects. The TLR7/8 agonist imidazoquinoline (IMDQ) is linked to the self-assembling peptide backbone through a GrB-responsive linkage. Its release could enhance the activation and function of immune cells, which will lead to increased secretion of GrB and enhance the effectiveness of immunotherapy together. The hydrogel adjuvant recruits immune cells, initiates dendritic cell maturation, and induces M1 polarized macrophages to reverse the immunosuppressive tumor microenvironment in situ. In multiple murine tumor models, the hydrogel adjuvant suppresses tumor growth, increases animal survival and long-term immunological memory, and protects mice against tumor rechallenge, leading to effective prophylactic and therapeutic responses. This work provides a potential chemical strategy to overcome the limitations associated with immunotherapy.
Collapse
Affiliation(s)
- Bihan Wu
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang 310024, China
| | - Juan Liang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang 310024, China
| | - Xuejiao Yang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang 310024, China
| | - Yu Fang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang 310024, China
| | - Nan Kong
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang 310024, China
| | - Dinghao Chen
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang 310024, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
43
|
Zhao R, Xiao Q, Wu Y, Zhang W, Liu J, Zeng Y, Zhan J, Cai Y, Fang C. Dual-crosslinking immunostimulatory hydrogel synchronously suppresses pancreatic fistula and pancreatic cancer relapse post-resection. Biomaterials 2024; 305:122453. [PMID: 38159361 DOI: 10.1016/j.biomaterials.2023.122453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/11/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
In pancreatic cancer (PC), surgical resection remains the sole curative option, albeit patients undergoing resection are susceptible to postoperative pancreatic fistula (PF) formation and tumor recurrence. An unmet need exists for a unified strategy capable of concomitantly averting PF and tumor relapse to mitigate morbidity in PC patients after surgery. Herein, an original dual crosslinked biological sealant hydrogel (methacrylate-hyaluronic acid-dopamine (MA-HA-DA) and sulfhydryl-hyaluronic acid-dopamine (SH-HA-DA)) was engineered as a drug depot and loaded with polydopamine-cloaked cytokine interleukin-15 and platelets conjugated with anti-TIGIT. In vitro analyses validated favorable tissue adhesion, cytocompatibility, and stability of the hydrogels. In a PF rodent model, the hydrogel effectively adhered to the pancreatic stump, sealing the severed pancreatic end and impeding post-operative elevations in amylase and lipase. In PC murine models, hydrogels potently stimulated CD8+ T and NK cells to deter residual tumor re-growth and distant metastasis. This innovative hydrogel strategy establishes a new framework for concomitant prevention of PF and PC recurrence.
Collapse
Affiliation(s)
- Ruizhi Zhao
- Department of Hepatobiliary Surgery, Institute of Digital Intelligence, Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Pazhou Lab, Guangzhou, 510320, China
| | - Qiuqun Xiao
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yuanyuan Wu
- Department of Hepatobiliary Surgery, Institute of Digital Intelligence, Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Pazhou Lab, Guangzhou, 510320, China
| | - Weiqi Zhang
- Guangdong Cardiovascular Institute, Department of General Surgery, Department of Breast Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Science), Southern Medical University, Guangzhou, 510100, China
| | - Jiale Liu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Yinghua Zeng
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jie Zhan
- Department of Laboratory Medicine, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yanbin Cai
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Chihua Fang
- Department of Hepatobiliary Surgery, Institute of Digital Intelligence, Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Pazhou Lab, Guangzhou, 510320, China.
| |
Collapse
|
44
|
Kudruk S, Forsyth CM, Dion MZ, Hedlund Orbeck JK, Luo J, Klein RS, Kim AH, Heimberger AB, Mirkin CA, Stegh AH, Artzi N. Multimodal neuro-nanotechnology: Challenging the existing paradigm in glioblastoma therapy. Proc Natl Acad Sci U S A 2024; 121:e2306973121. [PMID: 38346200 PMCID: PMC10895370 DOI: 10.1073/pnas.2306973121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024] Open
Abstract
Integrating multimodal neuro- and nanotechnology-enabled precision immunotherapies with extant systemic immunotherapies may finally provide a significant breakthrough for combatting glioblastoma (GBM). The potency of this approach lies in its ability to train the immune system to efficiently identify and eradicate cancer cells, thereby creating anti-tumor immune memory while minimizing multi-mechanistic immune suppression. A critical aspect of these therapies is the controlled, spatiotemporal delivery of structurally defined nanotherapeutics into the GBM tumor microenvironment (TME). Architectures such as spherical nucleic acids or poly(beta-amino ester)/dendrimer-based nanoparticles have shown promising results in preclinical models due to their multivalency and abilities to activate antigen-presenting cells and prime antigen-specific T cells. These nanostructures also permit systematic variation to optimize their distribution, TME accumulation, cellular uptake, and overall immunostimulatory effects. Delving deeper into the relationships between nanotherapeutic structures and their performance will accelerate nano-drug development and pave the way for the rapid clinical translation of advanced nanomedicines. In addition, the efficacy of nanotechnology-based immunotherapies may be enhanced when integrated with emerging precision surgical techniques, such as laser interstitial thermal therapy, and when combined with systemic immunotherapies, particularly inhibitors of immune-mediated checkpoints and immunosuppressive adenosine signaling. In this perspective, we highlight the potential of emerging treatment modalities, combining advances in biomedical engineering and neurotechnology development with existing immunotherapies to overcome treatment resistance and transform the management of GBM. We conclude with a call to action for researchers to leverage these technologies and accelerate their translation into the clinic.
Collapse
Affiliation(s)
- Sergej Kudruk
- Department of Chemistry, Northwestern University, Evanston, IL60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL60208
| | - Connor M. Forsyth
- Department of Chemistry, Northwestern University, Evanston, IL60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL60208
| | - Michelle Z. Dion
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Jenny K. Hedlund Orbeck
- Department of Chemistry, Northwestern University, Evanston, IL60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL60208
| | - Jingqin Luo
- The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO63110
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO63110
| | - Robyn S. Klein
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO63110
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO63110
| | - Albert H. Kim
- The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO63110
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO63110
| | - Amy B. Heimberger
- Department of Neurological Surgery, Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Chad A. Mirkin
- Department of Chemistry, Northwestern University, Evanston, IL60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL60208
| | - Alexander H. Stegh
- The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO63110
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO63110
| | - Natalie Artzi
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Medicine, Engineering in Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA02115
| |
Collapse
|
45
|
You Q, Liang F, Wu G, Cao F, Liu J, He Z, Wang C, Zhu L, Chen X, Yang Y. The Landscape of Biomimetic Nanovesicles in Brain Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306583. [PMID: 37713652 DOI: 10.1002/adma.202306583] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Brain diseases, such as brain tumors, neurodegenerative diseases, cerebrovascular diseases, and brain injuries, are caused by various pathophysiological changes, which pose a serious health threat. Brain disorders are often difficult to treat due to the presence of the blood-brain barrier (BBB). Biomimetic nanovesicles (BNVs), including endogenous extracellular vesicles (EVs) derived from various cells and artificial nanovesicles, possess the ability to penetrate the BBB and thus can be utilized for drug delivery to the brain. BNVs, especially endogenous EVs, are widely distributed in body fluids and usually carry various disease-related signal molecules such as proteins, RNA, and DNA, and may also be analyzed to understand the etiology and pathogenesis of brain diseases. This review covers the exhaustive classification and characterization of BNVs and pathophysiological roles involved in various brain diseases, and emphatically focuses on nanotechnology-integrated BNVs for brain disease theranostics, including various diagnosis strategies and precise therapeutic regulations (e.g., immunity regulation, disordered protein clearance, anti-neuroinflammation, neuroregeneration, angiogenesis, and the gut-brain axis regulation). The remaining challenges and future perspectives regarding the nanotechnology-integrated BNVs for the diagnosis and treatment of brain diseases are also discussed and outlined.
Collapse
Affiliation(s)
- Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, 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
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Fuming Liang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing, 400016, China
| | - Gege Wu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, 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
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, 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
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Jingyi Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhaohui He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing, 400016, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, 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
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
46
|
Cao L, Meng X, Zhang Z, Liu Z, He Y. Macrophage heterogeneity and its interactions with stromal cells in tumour microenvironment. Cell Biosci 2024; 14:16. [PMID: 38303024 PMCID: PMC10832170 DOI: 10.1186/s13578-024-01201-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/22/2024] [Indexed: 02/03/2024] Open
Abstract
Macrophages and tumour stroma cells account for the main cellular components in the tumour microenvironment (TME). Current advancements in single-cell analysis have revolutionized our understanding of macrophage diversity and macrophage-stroma interactions. Accordingly, this review describes new insight into tumour-associated macrophage (TAM) heterogeneity in terms of tumour type, phenotype, metabolism, and spatial distribution and presents the association between these factors and TAM functional states. Meanwhile, we focus on the immunomodulatory feature of TAMs and highlight the tumour-promoting effect of macrophage-tumour stroma interactions in the immunosuppressive TME. Finally, we summarize recent studies investigating macrophage-targeted therapy and discuss their therapeutic potential in improving immunotherapy by alleviating immunosuppression.
Collapse
Affiliation(s)
- Liren Cao
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiaoyan Meng
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhiyuan Zhang
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yue He
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
47
|
Furnari FB, Anastasaki C, Bian S, Fine HA, Koga T, Le LQ, Rodriguez FJ, Gutmann DH. Stem cell modeling of nervous system tumors. Dis Model Mech 2024; 17:dmm050533. [PMID: 38353122 PMCID: PMC10886724 DOI: 10.1242/dmm.050533] [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: 10/01/2023] [Accepted: 12/18/2023] [Indexed: 02/16/2024] Open
Abstract
Nervous system tumors, particularly brain tumors, represent the most common tumors in children and one of the most lethal tumors in adults. Despite decades of research, there are few effective therapies for these cancers. Although human nervous system tumor cells and genetically engineered mouse models have served as excellent platforms for drug discovery and preclinical testing, they have limitations with respect to accurately recapitulating important aspects of the pathobiology of spontaneously arising human tumors. For this reason, attention has turned to the deployment of human stem cell engineering involving human embryonic or induced pluripotent stem cells, in which genetic alterations associated with nervous system cancers can be introduced. These stem cells can be used to create self-assembling three-dimensional cerebral organoids that preserve key features of the developing human brain. Moreover, stem cell-engineered lines are amenable to xenotransplantation into mice as a platform to investigate the tumor cell of origin, discover cancer evolutionary trajectories and identify therapeutic vulnerabilities. In this article, we review the current state of human stem cell models of nervous system tumors, discuss their advantages and disadvantages, and provide consensus recommendations for future research.
Collapse
Affiliation(s)
- Frank B Furnari
- Department of Medicine, University of California, San Diego, San Diego, CA 92037, USA
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shan Bian
- Institute for Regenerative Medicine, School of Life Sciences and Technology, Tongji University, 200070 Shanghai, China
| | - Howard A Fine
- Department of Neurology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Tomoyuki Koga
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lu Q Le
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Fausto J Rodriguez
- Division of Neuropathology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
48
|
Luo EY, Sugimura RR. Taming microglia: the promise of engineered microglia in treating neurological diseases. J Neuroinflammation 2024; 21:19. [PMID: 38212785 PMCID: PMC10785527 DOI: 10.1186/s12974-024-03015-9] [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: 06/09/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Microglia, the CNS-resident immune cells, are implicated in many neurological diseases. Nearly one in six of the world's population suffers from neurological disorders, encompassing neurodegenerative and neuroautoimmune diseases, most with dysregulated neuroinflammation involved. Activated microglia become phagocytotic and secret various immune molecules, which are mediators of the brain immune microenvironment. Given their ability to penetrate through the blood-brain barrier in the neuroinflammatory context and their close interaction with neurons and other glial cells, microglia are potential therapeutic delivery vehicles and modulators of neuronal activity. Re-engineering microglia to treat neurological diseases is, thus, increasingly gaining attention. By altering gene expression, re-programmed microglia can be utilized to deliver therapeutics to targeted sites and control neuroinflammation in various neuroinflammatory diseases. This review addresses the current development in microglial engineering, including genetic targeting and therapeutic modulation. Furthermore, we discuss limitations to the genetic engineering techniques and models used to test the functionality of re-engineered microglia, including cell culture and animal models. Finally, we will discuss future directions for the application of engineered microglia in treating neurological diseases.
Collapse
Affiliation(s)
- Echo Yongqi Luo
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Pokfulam, Hong Kong
| | - Rio Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
49
|
Tian M, Ma Z, Yang GZ. Micro/nanosystems for controllable drug delivery to the brain. Innovation (N Y) 2024; 5:100548. [PMID: 38161522 PMCID: PMC10757293 DOI: 10.1016/j.xinn.2023.100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/26/2023] [Indexed: 01/03/2024] Open
Abstract
Drug delivery to the brain is crucial in the treatment for central nervous system disorders. While significant progress has been made in recent years, there are still major challenges in achieving controllable drug delivery to the brain. Unmet clinical needs arise from various factors, including controlled drug transport, handling large drug doses, methods for crossing biological barriers, the use of imaging guidance, and effective models for analyzing drug delivery. Recent advances in micro/nanosystems have shown promise in addressing some of these challenges. These include the utilization of microfluidic platforms to test and validate the drug delivery process in a controlled and biomimetic setting, the development of novel micro/nanocarriers for large drug loads across the blood-brain barrier, and the implementation of micro-intervention systems for delivering drugs through intraparenchymal or peripheral routes. In this article, we present a review of the latest developments in micro/nanosystems for controllable drug delivery to the brain. We also delve into the relevant diseases, biological barriers, and conventional methods. In addition, we discuss future prospects and the development of emerging robotic micro/nanosystems equipped with directed transportation, real-time image guidance, and closed-loop control.
Collapse
Affiliation(s)
- Mingzhen Tian
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guang-Zhong Yang
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
50
|
Zhang Z, He C, Chen X. Designing Hydrogels for Immunomodulation in Cancer Therapy and Regenerative Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308894. [PMID: 37909463 DOI: 10.1002/adma.202308894] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/26/2023] [Indexed: 11/03/2023]
Abstract
The immune system not only acts as a defense against pathogen and cancer cells, but also plays an important role in homeostasis and tissue regeneration. Targeting immune systems is a promising strategy for efficient cancer treatment and regenerative medicine. Current systemic immunomodulation therapies are usually associated with low persistence time, poor targeting to action sites, and severe side effects. Due to their extracellular matrix-mimetic nature, tunable properties and diverse bioactivities, hydrogels are intriguing platforms to locally deliver immunomodulatory agents and cells, as well as provide an immunomodulatory microenvironment to recruit, activate, and expand host immune cells. In this review, the design considerations, including polymer backbones, crosslinking mechanisms, physicochemical nature, and immunomodulation-related components, of the hydrogel platforms, are focused on. The immunomodulatory effects and therapeutic outcomes in cancer therapy and tissue regeneration of different hydrogel systems are emphasized, including hydrogel depots for delivery of immunomodulatory agents, hydrogel scaffolds for cell delivery, and immunomodulatory hydrogels depending on the intrinsic properties of materials. Finally, the remained challenges in current systems and future development of immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Zhen Zhang
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|