1
|
Zhang M, Liu C, Tu J, Tang M, Ashrafizadeh M, Nabavi N, Sethi G, Zhao P, Liu S. Advances in cancer immunotherapy: historical perspectives, current developments, and future directions. Mol Cancer 2025; 24:136. [PMID: 40336045 PMCID: PMC12057291 DOI: 10.1186/s12943-025-02305-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/15/2025] [Indexed: 05/09/2025] Open
Abstract
Cancer immunotherapy, encompassing both experimental and standard-of-care therapies, has emerged as a promising approach to harnessing the immune system for tumor suppression. Experimental strategies, including novel immunotherapies and preclinical models, are actively being explored, while established treatments, such as immune checkpoint inhibitors (ICIs), are widely implemented in clinical settings. This comprehensive review examines the historical evolution, underlying mechanisms, and diverse strategies of cancer immunotherapy, highlighting both its clinical applications and ongoing preclinical advancements. The review delves into the essential components of anticancer immunity, including dendritic cell activation, T cell priming, and immune surveillance, while addressing the challenges posed by immune evasion mechanisms. Key immunotherapeutic strategies, such as cancer vaccines, oncolytic viruses, adoptive cell transfer, and ICIs, are discussed in detail. Additionally, the role of nanotechnology, cytokines, chemokines, and adjuvants in enhancing the precision and efficacy of immunotherapies were explored. Combination therapies, particularly those integrating immunotherapy with radiotherapy or chemotherapy, exhibit synergistic potential but necessitate careful management to reduce side effects. Emerging factors influencing immunotherapy outcomes, including tumor heterogeneity, gut microbiota composition, and genomic and epigenetic modifications, are also examined. Furthermore, the molecular mechanisms underlying immune evasion and therapeutic resistance are analyzed, with a focus on the contributions of noncoding RNAs and epigenetic alterations, along with innovative intervention strategies. This review emphasizes recent preclinical and clinical advancements, with particular attention to biomarker-driven approaches aimed at optimizing patient prognosis. Challenges such as immunotherapy-related toxicity, limited efficacy in solid tumors, and production constraints are highlighted as critical areas for future research. Advancements in personalized therapies and novel delivery systems are proposed as avenues to enhance treatment effectiveness and accessibility. By incorporating insights from multiple disciplines, this review aims to deepen the understanding and application of cancer immunotherapy, ultimately fostering more effective and widely accessible therapeutic solutions.
Collapse
Affiliation(s)
- Meiyin Zhang
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chaojun Liu
- Department of Breast Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Jing Tu
- Department of Pulmonary and Critical Care Medicine, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8 V 1P7, Canada
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR) Yong Loo Lin, School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Peiqing Zhao
- Translational Medicine Center, Zibo Central Hospital Affiliated to Binzhou Medical University, No. 54 Communist Youth League Road, Zibo, China.
| | - Shijian Liu
- Department of General Medicine, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, 150081, China.
| |
Collapse
|
2
|
Ma T, Yu Y, Gao Y, Jiang S, Ge W, Zeng Y, Wang X, Li S, Xie X, Guan G. Smart self-assembled peptide-based hydrogels: Mechanism, design and biomedical applications. Colloids Surf B Biointerfaces 2025; 253:114704. [PMID: 40300283 DOI: 10.1016/j.colsurfb.2025.114704] [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: 12/17/2024] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 05/01/2025]
Abstract
Peptide hydrogels have gained widespread attention in biomedical engineering due to their unique ability to mimic the cellular microenvironment in vivo. Stimulus-responsive self-assembled (SAP) hydrogels can undergo conformational changes in response to changes in the external environment, prompting a sol-gel transition. Their inherent biodegradability, excellent surface activity and biocompatibility make them ideal candidates for a wide range of biomedical applications, and these SAP hydrogels can be widely used in the fields of tissue engineering, cell and drug delivery, wound healing and medical diagnostic imaging. In this paper, the basic properties, design principles, preparation methods and self-assembly mechanisms of different types of stimuli-responsive SAP hydrogels are reviewed. By designing and constructing stimulus-responsive SAP hydrogels, we can create materials that mimic natural physiological environments, thereby better simulating cell behavior and tissue repair. In addition, it highlights specific applications of these hydrogels in biomedical engineering, supported by examples from recent literature. The report summarizes the current state of research, highlights key challenges, and provides insights into future prospects to encourage continued innovation and exploration in this rapidly evolving field.
Collapse
Affiliation(s)
- Tao Ma
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yi Yu
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yijun Gao
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Shanshan Jiang
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Wenhui Ge
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yiyu Zeng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinying Wang
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Shuangjiang Li
- Department of Stomatology, Changsha Stomatological Hospital, Hunan University of Chinese Medicine, Changsha 410011, China
| | - Xiaoyan Xie
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Gaopeng Guan
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
3
|
Zhang Y, Dong X, Zhang Y, Chen Z, Zhou G, Chen N, Shen W, Yang K, Pei P. Biomaterials to regulate tumor extracellular matrix in immunotherapy. J Control Release 2024; 376:149-166. [PMID: 39389365 DOI: 10.1016/j.jconrel.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
The tumor extracellular matrix (ECM) provides physical support and influences tumor development, metastasis, and the tumor microenvironment, creating barriers to immune drug delivery and cell infiltration. Therefore, modulating or degrading the ECM is of significant importance to enhance the efficacy of tumor immunotherapy. This manuscript initially summarizes the main strategies and mechanisms of biomaterials in modulating various components of the ECM, including collagen, fibronectin, hyaluronic acid, and in remodeling the ECM. Subsequently, it discusses the benefits of biomaterials for immunotherapy following ECM modulation, such as promoting the infiltration of drugs and immune cells, regulating immune cell function, and alleviating the immunosuppressive microenvironment. The manuscript also briefly introduces the application of biomaterials that utilize and mimic the ECM for tumor immunotherapy. Finally, it addresses the current challenges and future directions in this field, providing a comprehensive overview of the potential and innovation in leveraging biomaterials to enhance cancer treatment outcomes. Our work will offer a comprehensive overview of ECM modulation strategies and their application in biomaterials to enhance tumor immunotherapy.
Collapse
Affiliation(s)
- Yujie Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xuexue Dong
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Yanxiang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zetong Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Ni Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Jiangsu, China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- Department of Nuclear Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province 230022, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| |
Collapse
|
4
|
Li S, Wang Y, Li C, Zhou B, Zeng X, Zhu H. Supramolecular nanomedicine in the intelligent cancer therapy: recent advances and future. Front Pharmacol 2024; 15:1490139. [PMID: 39464634 PMCID: PMC11502448 DOI: 10.3389/fphar.2024.1490139] [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: 09/02/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
In recent years, the incidence of cancer has been increasing year by year, and the burden of the disease and the economic burden caused by it has been worsening. Although chemotherapy, immunotherapy, targeted therapy and other therapeutic means continue to progress, they still inevitably have problems such as high toxicity and side effects, susceptibility to drug resistance, and high price. Photothermal therapy and photodynamic therapy have demonstrated considerable advantages in cancer imaging and treatment due to their minimally invasive and selective nature. However, their development has been constrained by challenges related to drug delivery. In recent times, drug delivery systems constructed based on supramolecular chemistry have been the subject of considerable interest, particularly in view of their compatibility with the high permeability and long retention effect of tumors. Furthermore, the advantage of dissociating the active ingredient under pH, light and other stimuli makes them unique in cancer therapy. This paper reviews the current status of supramolecular nanomedicines in cancer therapy, elucidating the challenges faced and providing a theoretical basis for the efficient and precise treatment of malignant tumors.
Collapse
Affiliation(s)
- Shuo Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yujiao Wang
- Division of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Binghao Zhou
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoxi Zeng
- Division of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Wells K, Liu T, Zhu L, Yang L. Immunomodulatory nanoparticles activate cytotoxic T cells for enhancement of the effect of cancer immunotherapy. NANOSCALE 2024; 16:17699-17722. [PMID: 39257225 DOI: 10.1039/d4nr01780c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Cancer immunotherapy represents a promising targeted treatment by leveraging the patient's immune system or adoptive transfer of active immune cells to selectively eliminate cancer cells. Despite notable clinical successes, conventional immunotherapies face significant challenges stemming from the poor infiltration of endogenous or adoptively transferred cytotoxic T cells in tumors, immunosuppressive tumor microenvironment and the immune evasion capability of cancer cells, leading to limited efficacy in many types of solid tumors. Overcoming these hurdles is essential to broaden the applicability of immunotherapies. Recent advances in nanotherapeutics have emerged as an innovative tool to overcome these challenges and enhance the therapeutic potential of tumor immunotherapy. The unique biochemical and biophysical properties of nanomaterials offer advantages in activation of immune cells in vitro for cell therapy, targeted delivery, and controlled release of immunomodulatory agents in vivo. Nanoparticles are excellent carriers for tumor associated antigens or neoantigen peptides for tumor vaccine, empowering activation of tumor specific T cell responses. By precisely delivering immunomodulatory agents to the tumor site, immunoactivating nanoparticles can promote tumor infiltration of endogenous T cells or adoptively transferred T cells into tumors, to overcoming delivery and biological barriers in the tumor microenvironment, augmenting the immune system's ability to recognize and eliminate cancer cells. This review provides an overview of the current advances in immunotherapeutic approaches utilizing nanotechnology. With a focus on discussions concerning strategies to enhance activity and efficacy of cytotoxic T cells and explore the intersection of engineering nanoparticles and immunomodulation aimed at bolstering T cell-mediated immune responses, we introduce various nanoparticle formulations designed to deliver therapeutic payloads, tumor antigens and immunomodulatory agents for T cell activation. Diverse mechanisms through which nanoparticle-based approaches influence T cell responses by improving antigen presentation, promoting immune cell trafficking, and reprogramming immunosuppressive tumor microenvironments to potentiate anti-tumor immunity are examined. Additionally, the synergistic potential of combining nanotherapeutics with existing immunotherapies, such as immune checkpoint inhibitors and adoptive T cell therapies is explored. In conclusion, this review highlights emerging research advances on activation of cytotoxic T cells using nanoparticle agents to support the promises and potential applications of nanoparticle-based immunomodulatory agents for cancer immunotherapy.
Collapse
Affiliation(s)
- Kory Wells
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tongrui Liu
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| | - Lei Zhu
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
6
|
Hua Y, Shen Y. Applications of self-assembled peptide hydrogels in anti-tumor therapy. NANOSCALE ADVANCES 2024; 6:2993-3008. [PMID: 38868817 PMCID: PMC11166105 DOI: 10.1039/d4na00172a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024]
Abstract
Peptides are a class of active substances composed of a variety of amino acids with special physiological functions. The rational design of peptide sequences at the molecular level enables their folding into diverse secondary structures. This property has garnered significant attention in the biomedical sphere owing to their favorable biocompatibility, adaptable mechanical traits, and exceptional loading capabilities. Concurrently with advancements in modern medicine, the diagnosis and treatment of tumors have increasingly embraced targeted and personalized approaches. This review explores recent applications of self-assembled peptides derived from natural amino acids in chemical therapy, immunotherapy, and other adjunctive treatments. We highlighted the utilization of peptide hydrogels as delivery systems for chemotherapeutic drugs and other bioactive molecules and then discussed the challenges and prospects for their future application.
Collapse
Affiliation(s)
- Yue Hua
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University Nanjing Jiangsu 210009 China
| | - Yang Shen
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University Nanjing Jiangsu 210009 China
| |
Collapse
|
7
|
Peng Y, Liang S, Meng QF, Liu D, Ma K, Zhou M, Yun K, Rao L, Wang Z. Engineered Bio-Based Hydrogels for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313188. [PMID: 38362813 DOI: 10.1002/adma.202313188] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/01/2024] [Indexed: 02/17/2024]
Abstract
Immunotherapy represents a revolutionary paradigm in cancer management, showcasing its potential to impede tumor metastasis and recurrence. Nonetheless, challenges including limited therapeutic efficacy and severe immune-related side effects are frequently encountered, especially in solid tumors. Hydrogels, a class of versatile materials featuring well-hydrated structures widely used in biomedicine, offer a promising platform for encapsulating and releasing small molecule drugs, biomacromolecules, and cells in a controlled manner. Immunomodulatory hydrogels present a unique capability for augmenting immune activation and mitigating systemic toxicity through encapsulation of multiple components and localized administration. Notably, hydrogels based on biopolymers have gained significant interest owing to their biocompatibility, environmental friendliness, and ease of production. This review delves into the recent advances in bio-based hydrogels in cancer immunotherapy and synergistic combinatorial approaches, highlighting their diverse applications. It is anticipated that this review will guide the rational design of hydrogels in the field of cancer immunotherapy, fostering clinical translation and ultimately benefiting patients.
Collapse
Affiliation(s)
- Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Qian-Fang Meng
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kaiqing Yun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
8
|
Sun W, Xie S, Liu SF, Hu X, Xing D. Evolving Tumor Characteristics and Smart Nanodrugs for Tumor Immunotherapy. Int J Nanomedicine 2024; 19:3919-3942. [PMID: 38708176 PMCID: PMC11070166 DOI: 10.2147/ijn.s453265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Typical physiological characteristics of tumors, such as weak acidity, low oxygen content, and upregulation of certain enzymes in the tumor microenvironment (TME), provide survival advantages when exposed to targeted attacks by drugs and responsive nanomedicines. Consequently, cancer treatment has significantly progressed in recent years. However, the evolution and adaptation of tumor characteristics still pose many challenges for current treatment methods. Therefore, efficient and precise cancer treatments require an understanding of the heterogeneity degree of various factors in cancer cells during tumor evolution to exploit the typical TME characteristics and manage the mutation process. The highly heterogeneous tumor and infiltrating stromal cells, immune cells, and extracellular components collectively form a unique TME, which plays a crucial role in tumor malignancy, including proliferation, invasion, metastasis, and immune escape. Therefore, the development of new treatment methods that can adapt to the evolutionary characteristics of tumors has become an intense focus in current cancer treatment research. This paper explores the latest understanding of cancer evolution, focusing on how tumors use new antigens to shape their "new faces"; how immune system cells, such as cytotoxic T cells, regulatory T cells, macrophages, and natural killer cells, help tumors become "invisible", that is, immune escape; whether the diverse cancer-associated fibroblasts provide support and coordination for tumors; and whether it is possible to attack tumors in reverse. This paper discusses the limitations of targeted therapy driven by tumor evolution factors and explores future strategies and the potential of intelligent nanomedicines, including the systematic coordination of tumor evolution factors and adaptive methods, to meet this therapeutic challenge.
Collapse
Affiliation(s)
- Wenshe Sun
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Shaowei Xie
- Department of Ultrasound, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Shi Feng Liu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Xiaokun Hu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
| |
Collapse
|
9
|
Song H, Sun H, He N, Xu C, Du L, Ji K, Wang J, Zhang M, Gu Y, Wang Y, Liu Q. Glutathione Depletion-Induced Versatile Nanomedicine for Potentiating the Ferroptosis to Overcome Solid Tumor Radioresistance and Enhance Immunotherapy. Adv Healthc Mater 2024; 13:e2303412. [PMID: 38245863 DOI: 10.1002/adhm.202303412] [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: 10/07/2023] [Revised: 12/04/2023] [Indexed: 01/22/2024]
Abstract
A high level of reduced glutathione is a major factor contributing to the radioresistance observed in solid tumors. To address this radioresistance associated with glutathione, a cinnamaldehyde (CA) polymer prodrug, referred to as PDPCA, is fabricated. This prodrug is created by synthesizing a pendent CA prodrug with acetal linkages in a hydrophobic block, forming a self-assembled into a core-shell nanoparticle in aqueous media. Additionally, it encapsulates all-trans retinoic acid (ATRA) for synchronous delivery, resulting in PDPCA@ATRA. The PDPCA@ATRA nanoparticles accumulate reactive oxygen species through both endogenous and exogenous pathways, enhancing ferroptosis by depleting glutathione. This approach demonstrates efficacy in overcoming tumor radioresistance in vivo and in vitro, promoting the ferroptosis, and enhancing the cytotoxic T lymphocyte (CTL) response for lung tumors to anti-PD-1 (αPD-1) immunotherapy. Furthermore, this study reveals that PDPCA@ATRA nanoparticles promote ferroptosis through the NRF2-GPX4 signaling pathway, suggesting the potential for further investigation into the combination of radiotherapy and αPD-1 immune checkpoint inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Huijuan Song
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| | - Hao Sun
- School of Preventive Medicine Sciences (Institute of Radiation Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, 250062, China
| | - Ningning He
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| | - Chang Xu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| | - Liqing Du
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| | - Kaihua Ji
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| | - Jinhan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| | - Manman Zhang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| | - Yeqing Gu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| | - Yan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin, 300192, China
| |
Collapse
|
10
|
Mohaghegh N, Ahari A, Zehtabi F, Buttles C, Davani S, Hoang H, Tseng K, Zamanian B, Khosravi S, Daniali A, Kouchehbaghi NH, Thomas I, Serati Nouri H, Khorsandi D, Abbasgholizadeh R, Akbari M, Patil R, Kang H, Jucaud V, Khademhosseini A, Hassani Najafabadi A. Injectable hydrogels for personalized cancer immunotherapies. Acta Biomater 2023; 172:67-91. [PMID: 37806376 DOI: 10.1016/j.actbio.2023.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/19/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
The field of cancer immunotherapy has shown significant growth, and researchers are now focusing on effective strategies to enhance and prolong local immunomodulation. Injectable hydrogels (IHs) have emerged as versatile platforms for encapsulating and controlling the release of small molecules and cells, drawing significant attention for their potential to enhance antitumor immune responses while inhibiting metastasis and recurrence. IHs delivering natural killer (NK) cells, T cells, and antigen-presenting cells (APCs) offer a viable method for treating cancer. Indeed, it can bypass the extracellular matrix and gradually release small molecules or cells into the tumor microenvironment, thereby boosting immune responses against cancer cells. This review provides an overview of the recent advancements in cancer immunotherapy using IHs for delivering NK cells, T cells, APCs, chemoimmunotherapy, radio-immunotherapy, and photothermal-immunotherapy. First, we introduce IHs as a delivery matrix, then summarize their applications for the local delivery of small molecules and immune cells to elicit robust anticancer immune responses. Additionally, we discuss recent progress in IHs systems used for local combination therapy, including chemoimmunotherapy, radio-immunotherapy, photothermal-immunotherapy, photodynamic-immunotherapy, and gene-immunotherapy. By comprehensively examining the utilization of IHs in cancer immunotherapy, this review aims to highlight the potential of IHs as effective carriers for immunotherapy delivery, facilitating the development of innovative strategies for cancer treatment. In addition, we demonstrate that using hydrogel-based platforms for the targeted delivery of immune cells, such as NK cells, T cells, and dendritic cells (DCs), has remarkable potential in cancer therapy. These innovative approaches have yielded substantial reductions in tumor growth, showcasing the ability of hydrogels to enhance the efficacy of immune-based treatments. STATEMENT OF SIGNIFICANCE: As cancer immunotherapy continues to expand, the mode of therapeutic agent delivery becomes increasingly critical. This review spotlights the forward-looking progress of IHs, emphasizing their potential to revolutionize localized immunotherapy delivery. By efficiently encapsulating and controlling the release of essential immune components such as T cells, NK cells, APCs, and various therapeutic agents, IHs offer a pioneering pathway to amplify immune reactions, moderate metastasis, and reduce recurrence. Their adaptability further shines when considering their role in emerging combination therapies, including chemoimmunotherapy, radio-immunotherapy, and photothermal-immunotherapy. Understanding IHs' significance in cancer therapy is essential, suggesting a shift in cancer treatment dynamics and heralding a novel period of focused, enduring, and powerful therapeutic strategies.
Collapse
Affiliation(s)
- Neda Mohaghegh
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Amir Ahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Claire Buttles
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Indiana University Bloomington, Department of Biology, Bloomington, IN 47405, USA
| | - Saya Davani
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Hanna Hoang
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90024, USA
| | - Kaylee Tseng
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90007, USA
| | - Benjamin Zamanian
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, V6T1Z4, Canada
| | - Ariella Daniali
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Negar Hosseinzadeh Kouchehbaghi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Textile Engineering, Amirkabir University of Technology (Tehran Polytechnic), Hafez Avenue, Tehran, Iran
| | - Isabel Thomas
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Hamed Serati Nouri
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohsen Akbari
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Rameshwar Patil
- Department of Basic Science and Neurosurgery, Division of Cancer Science, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Heemin Kang
- Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA.
| | | |
Collapse
|
11
|
Zhang M, Chen W, Ju Y, Zhao H, Wang C. Polymer-Protein Nanovaccine Synthesized via Reactive Self-Assembly with Potential Application in Cancer Immunotherapy: Physicochemical and Biological Characterization In Vitro and In Vivo. Macromol Rapid Commun 2023; 44:e2300438. [PMID: 37708966 DOI: 10.1002/marc.202300438] [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: 07/20/2023] [Revised: 09/10/2023] [Indexed: 09/16/2023]
Abstract
Nanovaccines composed of polymeric nanocarriers and protein-based antigens have attracted much attention in recent years because of their enormous potential in the prevention and treatment of diseases such as viral infections and cancer. While surface-conjugated protein antigens are known to be more immunoactive than encapsulated antigens, current surface conjugation methods often result in low and insufficient protein loading. Herein, reactive self-assembly is used to prepare nanovaccine from poly(ε-caprolactone) (PCL) and ovalbumin (OVA)-a model antigen. A rapid thiol-disulfide exchange reaction between PCL with pendant pyridyl disulfide groups and thiolated OVA results in the formation of nanoparticles with narrow size distribution. High OVA loading (≈70-80 wt%) is achieved, and the native secondary structure of OVA is preserved. Compared to free OVA, the nanovaccine is much superior in enhancing antigen uptake by bone marrow-derived dendritic cells (BMDCs), promoting BMDC maturation and antigen presentation via the MHC I pathway, persisting at the injection site and draining lymph nodes, activating both Th1 and Th2 T cell immunity, and ultimately, resisting tumor challenge in mice. This is the first demonstration of reactive self-assembly for the construction of a polymer-protein nanovaccine with clear potential in advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Mingming Zhang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Wenjuan Chen
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Yuanyuan Ju
- Key Laboratory of Functional Polymer Materials, Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Hanying Zhao
- Key Laboratory of Functional Polymer Materials, Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chun Wang
- Department of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo Hall, 312 Church Street S. E., Minneapolis, MN, 55455, USA
| |
Collapse
|
12
|
Zhang J, Zhao D, Lu K. Mechanisms and influencing factors of peptide hydrogel formation and biomedicine applications of hydrogels. SOFT MATTER 2023; 19:7479-7493. [PMID: 37756117 DOI: 10.1039/d3sm01057k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Self-assembled peptide-based hydrogels have shown great potential in bio-related applications due to their porous structure, strong mechanical stability, high biocompatibility, and easy functionalization. Herein, the structure and characteristics of hydrogels and the mechanism of action of several regular secondary structures during gelation are investigated. The factors influencing the formation of peptide hydrogels, especially the pH responsiveness and salt ion induction are analyzed and summarized. Finally, the biomedical applications of peptide hydrogels, such as bone tissue engineering, cell culture, antigen presentation, antibacterial materials, and drug delivery are reviewed.
Collapse
Affiliation(s)
- Jiahui Zhang
- School of Chemistry and Chemical Engineering, Henan University of Technology, Locus Street, High-Tech Industry Development Zone, Zhengzhou 450001, China.
| | - Dongxin Zhao
- School of Chemistry and Chemical Engineering, Henan University of Technology, Locus Street, High-Tech Industry Development Zone, Zhengzhou 450001, China.
| | - Kui Lu
- School of Chemistry and Chemical Engineering, Henan University of Technology, Locus Street, High-Tech Industry Development Zone, Zhengzhou 450001, China.
- School of Chemical Engineering and Food Science, Zhengzhou University of Technology, Yingcai Road 18, Zhengzhou, 450044, Henan Province, China.
| |
Collapse
|
13
|
Zhu H, Zheng J, Oh XY, Chan CY, Low BQL, Tor JQ, Jiang W, Ye E, Loh XJ, Li Z. Nanoarchitecture-Integrated Hydrogel Systems toward Therapeutic Applications. ACS NANO 2023; 17:7953-7978. [PMID: 37071059 DOI: 10.1021/acsnano.2c12448] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Hydrogels, as one of the most feasible soft biomaterials, have gained considerable attention in therapeutic applications by virtue of their tunable properties including superior patient compliance, good biocompatibility and biodegradation, and high cargo-loading efficiency. However, hydrogel application is still limited by some challenges like inefficient encapsulation, easy leakage of loaded cargoes, and the lack of controllability. Recently, nanoarchitecture-integrated hydrogel systems were found to be therapeutics with optimized properties, extending their bioapplication. In this review, we briefly presented the category of hydrogels according to their synthetic materials and further discussed the advantages in bioapplication. Additionally, various applications of nanoarchitecture hybrid hydrogels in biomedical engineering are systematically summarized, including cancer therapy, wound healing, cardiac repair, bone regeneration, diabetes therapy, and obesity therapy. Last, the current challenges, limitations, and future perspectives in the future development of nanoarchitecture-integrated flexible hydrogels are addressed.
Collapse
Affiliation(s)
- Houjuan Zhu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Jie Zheng
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Xin Yi Oh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Chui Yu Chan
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Beverly Qian Ling Low
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Jia Qian Tor
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Wenbin Jiang
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Enyi Ye
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore, Singapore 117574, Republic of Singapore
| |
Collapse
|