1
|
Tan H, Shen Z, Wang X, Shu S, Deng J, Lu L, Fan Z, Hu D, Cheng P, Cao X, Huang Q. Endoplasmic reticulum-targeted biomimetic nanoparticles induce apoptosis and ferroptosis by regulating endoplasmic reticulum function in colon cancer. J Control Release 2024; 375:422-437. [PMID: 39278355 DOI: 10.1016/j.jconrel.2024.09.018] [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: 06/25/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Colorectal cancer (CRC) is a major threat to human health, as it is one of the most common malignancies with a high incidence and mortality rate. The cancer cell membrane (CCM) has significant potential in targeted tumor drug delivery due to its membrane antigen-mediated homologous targeting ability. The endoplasmic reticulum (ER) in cancer cells plays a crucial role in apoptosis and ferroptosis. In this study, we developed an ER-targeted peptide-modified CCM-biomimetic nanoparticle-delivered lovastatin (LOV) nanomedicine delivery system (EMPP-LOV) for cancer treatment. Both in vitro and in vivo experiments demonstrated that EMPP could effectively target cancer cells and localize within the ER. EMPP-LOV modulated ER function to promote apoptosis and ferroptosis in tumor cells. Furthermore, synergistic antitumor efficacy was observed in both in vitro and in vivo models. EMPP-LOV induced apoptosis in CRC cells by over-activating endoplasmic reticulum stress and promoted ferroptosis by inhibiting the mevalonate pathway, leading to synergistic tumor growth inhibition with minimal toxicity to major organs. Overall, the EMPP-LOV delivery system, with its subcellular targeting capability within tumor cells, presents a promising therapeutic platform for CRC treatment.
Collapse
Affiliation(s)
- Hongxin Tan
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ziqi Shen
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Xiaohua Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Sicheng Shu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jie Deng
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Li Lu
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Ziyan Fan
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Danni Hu
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Pu Cheng
- Department of Gynaecology, The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xi Cao
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China; Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Qi Huang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
2
|
Xun J, Jiang X, Liu B, Hu Z, Liu J, Han Y, Gao R, Zhang H, Yang S, Yu X, Wang X, Yan C, Zhang Q. Neogambogic acid enhances anti-PD-1 immunotherapy efficacy by attenuating suppressive function of MDSCs in pancreatic cancer. Int Immunopharmacol 2024; 139:112696. [PMID: 39018692 DOI: 10.1016/j.intimp.2024.112696] [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: 03/04/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Anti-PD-1-based immunotherapy has limited benefits in patients with pancreatic cancer. Accumulating data indicate that natural products exert antitumor activity by remodeling the tumor immune microenvironment. It has been reported that neogambogic acid (NGA), an active natural monomer extracted from Garcinia, has anti-inflammatory and antitumor effects. Nevertheless, there are few systematic studies on the antitumor efficacy and immunomodulatory effects of NGA in pancreatic cancer. METHODS An orthotopic mouse model of pancreatic cancer was established and were treated with different doses of NGA. Tumor growth and ascites were observed. Flow cytometry and immunohistochemistry (IHC) were used to investigate the tumor immune microenvironment. CD11b+ MDSCs were infused back into mice with pancreatic cancer to observe tumor progression after NGA treatment. Bone marrow cells were induced to differentiate into MDSCs, and the effects of NGA on MDSCs were analyzed and the underlying mechanism was explored. The effects of NGA combined with an anti-PD-1 antibody on pancreatic cancer were further tested. RESULTS NGA significantly inhibited the tumor growth and improve ascites character in pancreatic cancer model mice. Flow cytometry and IHC analysis revealed that NGA decreased the MDSCs proportion and infiltration in the tumor microenvironment. Moreover, adoptive MDSCs largely attenuated the inhibitory effect of NGA on the progression of pancreatic cancer. In addition, we showed that NGA significantly promoted apoptosis and inhibited the differentiation, migration and immunosuppressive function of MDSCs and decreased level of STAT3 and p-STAT3. Furthermore, we demonstrated that NGA synergistically enhanced the efficacy of anti-PD-1 antibodies against pancreatic cancer. CONCLUSION NGA inhibited the progression of pancreatic cancer by inhibiting MDSCs in the tumor microenvironment, and enhanced the efficacy of anti-PD-1 therapy in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jing Xun
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin 300100, China; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin 300100, China
| | - Xiaolin Jiang
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin 300100, China; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin 300100, China
| | - Bin Liu
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin 300100, China; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin 300100, China
| | - Zhibo Hu
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin 300100, China; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin 300100, China
| | - Jinjin Liu
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin 300100, China; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin 300100, China; Integrated Chinese and Western Medicine Hospital, Tianjin University, Tianjin, China
| | - Yingdi Han
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin 300100, China; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin 300100, China
| | - Ruifang Gao
- Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, China
| | - Hui Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin 300100, China; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin 300100, China; Integrated Chinese and Western Medicine Hospital, Tianjin University, Tianjin, China
| | - Shimin Yang
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Xiangyang Yu
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Ximo Wang
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Chen Yan
- Tianjin Vocational College of Bioengineering, Tianjin 300301, China.
| | - Qi Zhang
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin 300100, China; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin 300100, China.
| |
Collapse
|
3
|
Zhang Z, Li M, Zhang X, Zhou F. Novel Strategies for Tumor Treatment: Harnessing ROS-Inducing Active Ingredients from Traditional Chinese Medicine Through Multifunctional Nanoformulations. Int J Nanomedicine 2024; 19:9659-9688. [PMID: 39309188 PMCID: PMC11416109 DOI: 10.2147/ijn.s479212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Reactive oxygen species (ROS) encompass a diverse group of chemically reactive molecules or ions distinguished by their substantial oxidative potential. Empirical studies have shown that the targeted administration of high toxic concentrations of ROS can effectively induce tumor cell death in various types. Numerous bioactive ingredients derived from traditional Chinese medicine (TCM), recognized for their ROS-inducing properties, have demonstrated significant anti-tumor activity. Nonetheless, their clinical application has been hindered by challenges such as low solubility, limited bioavailability, and poor selectivity. Multifunctional nanoformulations possess the potential to overcome these challenges and enhance the anticancer efficacy of ROS-inducing active compounds. Through extensive searches of various academic databases and a thorough review and screening of relevant literature, this study aims to systematically summarize and generalize multiple active ingredients in TCM that induce ROS generation, along with their multifunctional nanoformulations, from various perspectives. The objective is to provide new insights and references for fundamental cancer research and clinical treatments. Furthermore, we acknowledge that although numerous active ingredients and their nanoformulations in TCM have demonstrated ROS-inducing and anti-tumor potentials, potentially offering novel strategies for tumor therapy, the underlying mechanisms require further comprehensive investigation.
Collapse
Affiliation(s)
- Zhengguang Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Xiaolong Zhang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Fuqiong Zhou
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| |
Collapse
|
4
|
Jin C, Zhang Y, Li B, Gao T, Wang B, Hua P. Robust anti-tumor immunity through the integration of targeted lipid nanoparticle-based mRNA nanovaccines with PD-1/PD-L1 blockade. Mater Today Bio 2024; 27:101136. [PMID: 39015802 PMCID: PMC11251012 DOI: 10.1016/j.mtbio.2024.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/18/2024] Open
Abstract
Tumor mRNA vaccines present a personalized approach in cancer immunotherapy, encoding distinct tumor antigens to evoke robust immune responses and offering the potential against emerging tumor variants. Despite this, the clinical advancement of tumor mRNA vaccines has been hampered by their limited delivery capacity and inefficient activation of antigen-presenting cells (APCs). Herein, we employed microfluidics technology to engineer mannose-modified lipid-based nanovaccines for specifically targeting APCs. The encapsulation process efficiently entrapped the cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) agonist along with mRNA encoding antigens. The targeted nanovaccines (TNVs) exhibited a narrow particle size distribution, ensuring consistent and efficient delivery. These TNVs significantly enhanced gene expression of mRNA, facilitating antigen presentation and immune activation. When compared to non-targeted nanovaccines, TNVs outperformed in terms of antigen presentation and immune activation. Furthermore, the combination of anti-PD-L1 antibodies with TNVs elicited a synergistic anti-tumor effect. This was attributed to the anti-PD-L1 antibodies' ability to overcome the immune suppression of tumor cells. Our findings suggest that the combination treatment elicited the most robust anti-tumor immune activation and immune memory effect. These results indicate that integrating tumor mRNA vaccines with immune checkpoint inhibitors or other immunostimulatory agents may be crucial for enhancing the immune response.
Collapse
Affiliation(s)
- Chengyan Jin
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, 130022, China
| | - Yan Zhang
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, 130022, China
| | - Baofeng Li
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, 130022, China
| | - Tianci Gao
- College of Clinical Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154007, China
| | - Bin Wang
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, 130022, China
| | - Peiyan Hua
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, 130022, China
| |
Collapse
|
5
|
Ma Y, Yi J, Ruan J, Ma J, Yang Q, Zhang K, Zhang M, Zeng G, Jin L, Huang X, Li J, Yang H, Wu W, Sun D. Engineered Cell Membrane-Coated Nanoparticles: New Strategies in Glioma Targeted Therapy and Immune Modulation. Adv Healthc Mater 2024; 13:e2400514. [PMID: 38652681 DOI: 10.1002/adhm.202400514] [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: 02/08/2024] [Revised: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Gliomas, the most prevalent primary brain tumors, pose considerable challenges due to their heterogeneity, intricate tumor microenvironment (TME), and blood-brain barrier (BBB), which restrict the effectiveness of traditional treatments like surgery and chemotherapy. This review provides an overview of engineered cell membrane technologies in glioma therapy, with a specific emphasis on targeted drug delivery and modulation of the immune microenvironment. This study investigates the progress in engineered cell membranes, encompassing physical, chemical, and genetic alterations, to improve drug delivery across the BBB and effectively target gliomas. The examination focuses on the interaction of engineered cell membrane-coated nanoparticles (ECM-NPs) with the TME in gliomas, emphasizing their potential to modulate glioma cell behavior and TME to enhance therapeutic efficacy. The review further explores the involvement of ECM-NPs in immunomodulation techniques, highlighting their impact on immune reactions. While facing obstacles related to membrane stability and manufacturing scalability, the review outlines forthcoming research directions focused on enhancing membrane performance. This review underscores the promise of ECM-NPs in surpassing conventional therapeutic constraints, proposing novel approaches for efficacious glioma treatment.
Collapse
Affiliation(s)
- Yilei Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Maolan Zhang
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Guoming Zeng
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Xiaobei Huang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Haifeng Yang
- JinFeng Laboratory, Chongqing, 401329, China
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, 400044, China
| | - Wei Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
- JinFeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
6
|
Chen Y, Fan W, Zhao Y, Liu M, Hu L, Zhang W. Progress in the Regulation of Immune Cells in the Tumor Microenvironment by Bioactive Compounds of Traditional Chinese Medicine. Molecules 2024; 29:2374. [PMID: 38792234 PMCID: PMC11124165 DOI: 10.3390/molecules29102374] [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: 04/10/2024] [Revised: 05/04/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
The tumor microenvironment (TME) can aid tumor cells in evading surveillance and clearance by immune cells, creating an internal environment conducive to tumor cell growth. Consequently, there is a growing focus on researching anti-tumor immunity through the regulation of immune cells within the TME. Various bioactive compounds in traditional Chinese medicine (TCM) are known to alter the immune balance by modulating the activity of immune cells in the TME. In turn, this enhances the body's immune response, thus promoting the effective elimination of tumor cells. This study aims to consolidate recent findings on the regulatory effects of bioactive compounds from TCM on immune cells within the TME. The bioactive compounds of TCM regulate the TME by modulating macrophages, dendritic cells, natural killer cells and T lymphocytes and their immune checkpoints. TCM has a long history of having been used in clinical practice in China. Chinese medicine contains various chemical constituents, including alkaloids, polysaccharides, saponins and flavonoids. These components activate various immune cells, thereby improving systemic functions and maintaining overall health. In this review, recent progress in relation to bioactive compounds derived from TCM will be covered, including TCM alkaloids, polysaccharides, saponins and flavonoids. This study provides a basis for further in-depth research and development in the field of anti-tumor immunomodulation using bioactive compounds from TCM.
Collapse
Affiliation(s)
- Yuqian Chen
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (Y.C.); (W.F.); (Y.Z.); (M.L.)
- Shandong Engineering Research Center for Smart Materials and Regenerative Medicine, Weifang 261053, China
| | - Wenshuang Fan
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (Y.C.); (W.F.); (Y.Z.); (M.L.)
| | - Yanyan Zhao
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (Y.C.); (W.F.); (Y.Z.); (M.L.)
- Shandong Engineering Research Center for Smart Materials and Regenerative Medicine, Weifang 261053, China
| | - Meijun Liu
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (Y.C.); (W.F.); (Y.Z.); (M.L.)
- Shandong Engineering Research Center for Smart Materials and Regenerative Medicine, Weifang 261053, China
| | - Linlin Hu
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (Y.C.); (W.F.); (Y.Z.); (M.L.)
- Shandong Engineering Research Center for Smart Materials and Regenerative Medicine, Weifang 261053, China
| | - Weifen Zhang
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (Y.C.); (W.F.); (Y.Z.); (M.L.)
- Shandong Engineering Research Center for Smart Materials and Regenerative Medicine, Weifang 261053, China
| |
Collapse
|
7
|
Zhong Z, Deng W, Wu J, Shang H, Tong Y, He Y, Huang Q, Ba X, Chen Z, Tang K. Cell membrane coated nanoparticles as a biomimetic drug delivery platform for enhancing cancer immunotherapy. NANOSCALE 2024; 16:8708-8738. [PMID: 38634521 DOI: 10.1039/d4nr00284a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Cancer immunotherapy, a burgeoning modality for cancer treatment, operates by activating the autoimmune system to impede the growth of malignant cells. Although numerous immunotherapy strategies have been employed in clinical cancer therapy, the resistance of cancer cells to immunotherapeutic medications and other apprehensions impede the attainment of sustained advantages for most patients. Recent advancements in nanotechnology for drug delivery hold promise in augmenting the efficacy of immunotherapy. However, the efficacy is currently constrained by the inadequate specificity of delivery, low rate of response, and the intricate immunosuppressive tumor microenvironment. In this context, the investigation of cell membrane coated nanoparticles (CMNPs) has revealed their ability to perform targeted delivery, immune evasion, controlled release, and immunomodulation. By combining the advantageous features of natural cell membranes and nanoparticles, CMNPs have demonstrated their unique potential in the realm of cancer immunotherapy. This review aims to emphasize recent research progress and elucidate the underlying mechanisms of CMNPs as an innovative drug delivery platform for enhancing cancer immunotherapy. Additionally, it provides a comprehensive overview of the current immunotherapeutic strategies involving different cell membrane types of CMNPs, with the intention of further exploration and optimization.
Collapse
Affiliation(s)
- Zichen Zhong
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Wen Deng
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
8
|
Shin S, Ahn YR, Kim M, Choi J, Kim H, Kim HO. Mammalian Cell Membrane Hybrid Polymersomes for mRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38615329 DOI: 10.1021/acsami.4c00843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Cell membranes are structures essential to the cell function and adaptation. Recent studies have targeted cell membranes to identify their protective and interactive properties. Leveraging these attributes of cellular membranes and their application to vaccine delivery is gaining increasing prominence. This study aimed to fuse synthetic polymeric nanoparticles with cell membranes to develop cell membrane hybrid polymersomes (HyPSomes) for enhanced vaccine delivery. We designed a platform to hybridize cell membranes with methoxy-poly(ethylene glycol)-block-polylactic acid nanoparticles by using the properties of both components. The formed HyPSomes were optimized by using dynamic light scattering, transmission electron microscopy, and Förster resonance energy transfer, and their stability was confirmed. The synthesized HyPSomes replicated the antigenic surface of the source cells and possessed the stability and efficacy of synthetic nanoparticles. These HyPSomes demonstrated enhanced cellular uptake and translation efficiency and facilitated endosome escape. HyPSomes showed outstanding capabilities for the delivery of foreign mRNAs to antigen-presenting cells. HyPSomes may serve as vaccine delivery systems by bridging the gap between synthetic and natural systems. These systems could be used in other contexts, e.g., diagnostics and drug delivery.
Collapse
Affiliation(s)
- SoJin Shin
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
- Department of Smart Health Science and Technology, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| | - Yu-Rim Ahn
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
- Department of Smart Health Science and Technology, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| | - Minse Kim
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
- Department of Smart Health Science and Technology, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| | - Jaewon Choi
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
- Department of Smart Health Science and Technology, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| | - HakSeon Kim
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
- Department of Smart Health Science and Technology, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| | - Hyun-Ouk Kim
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
- Department of Smart Health Science and Technology, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| |
Collapse
|
9
|
Jia W, Shen X, Guo Z, Cheng X, Zhao R. The future of cancer vaccines against colorectal cancer. Expert Opin Biol Ther 2024; 24:269-284. [PMID: 38644655 DOI: 10.1080/14712598.2024.2341744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/08/2024] [Indexed: 04/23/2024]
Abstract
INTRODUCTION Colorectal cancer (CRC) is the second most lethal malignancy worldwide. Immune checkpoint inhibitors (ICIs) benefit only 15% of patients with mismatch repair-deficient/microsatellite instability (dMMR/MSI) CRC. The majority of patients are not suitable due to insufficient immune infiltration. Cancer vaccines are a potential approach for inducing tumor-specific immunity within the solid tumor microenvironment. AREA COVERED In this review, we have provided an overview of the current progress in CRC vaccines over the past three years and briefly depict promising directions for further exploration. EXPERT OPINION Cancer vaccines are certainly a promising field for the antitumor treatment against CRC. Compared to monotherapy, cancer vaccines are more appropriate as adjuvants to standard treatment, especially in combination with ICI blockade, for microsatellite stable patients. Improved vaccine construction requires neoantigens with sufficient immunogenicity, satisfactory HLA-binding affinity, and an ideal delivery platform with perfect lymph node retention and minimal off-target effects. Prophylactic vaccines that potentially prevent CRC carcinogenesis are also worth investigating. The exploration of appropriate biomarkers for cancer vaccines may benefit prognostic prediction analysis and therapeutic response prediction in patients with CRC. Although many challenges remain, CRC vaccines represent an exciting area of research that may become an effective addition to current guidelines.
Collapse
Affiliation(s)
- Wenqing Jia
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaonan Shen
- Department of Gastroenterology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zichao Guo
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Cheng
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ren Zhao
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Huang X, Zhu X, Yang H, Li Q, Gai L, Sui X, Lu H, Feng J. Nanomaterial Delivery Vehicles for the Development of Neoantigen Tumor Vaccines for Personalized Treatment. Molecules 2024; 29:1462. [PMID: 38611742 PMCID: PMC11012694 DOI: 10.3390/molecules29071462] [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: 02/22/2024] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Tumor vaccines have been considered a promising therapeutic approach for treating cancer in recent years. With the development of sequencing technologies, tumor vaccines based on neoantigens or genomes specifically expressed in tumor cells, mainly in the form of peptides, nucleic acids, and dendritic cells, are beginning to receive widespread attention. Therefore, in this review, we have introduced different forms of neoantigen vaccines and discussed the development of these vaccines in treating cancer. Furthermore, neoantigen vaccines are influenced by factors such as antigen stability, weak immunogenicity, and biosafety in addition to sequencing technology. Hence, the biological nanomaterials, polymeric nanomaterials, inorganic nanomaterials, etc., used as vaccine carriers are principally summarized here, which may contribute to the design of neoantigen vaccines for improved stability and better efficacy.
Collapse
Affiliation(s)
- Xiaoyu Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Xiaolong Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Huan Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Qinyi Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Lizhi Gai
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Hua Lu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Jiao Feng
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| |
Collapse
|
11
|
Tai Y, Chen M, Wang F, Fan Y, Zhang J, Cai B, Yan L, Luo Y, Li Y. The role of dendritic cells in cancer immunity and therapeutic strategies. Int Immunopharmacol 2024; 128:111548. [PMID: 38244518 DOI: 10.1016/j.intimp.2024.111548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Dendritic cells (DCs) are asserted as the most potent antigen-presenting cells (APCs) that orchestrate both innate and adaptive immunity, being extremely effective in the induction of robust anti-cancer T cell responses. Hence, the modulation of DCs function represents an attractive target for improving cancer immunotherapy efficacy. A better understanding of the immunobiology of DCs, the interaction among DCs, immune effector cells and tumor cells in tumor microenvironment (TME) and the latest advances in biomedical engineering technology would be required for the design of optimal DC-based immunotherapy. In this review, we focus on elaborating the immunobiology of DCs in healthy and cancer environments, the recent advances in the development of enhancing endogenous DCs immunocompetence via immunomodulators as well as DC-based vaccines. The rapidly developing field of applying nanotechnology to improve DC-based immunotherapy is also highlighted.
Collapse
Affiliation(s)
- Yunze Tai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Man Chen
- Hebei Yanda Lu Daopei Hospital, Langfang 065201, China
| | - Fang Wang
- Department of Medical Laboratory, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, China
| | - Yu Fan
- Department of Urology, National Clinical Research Center for Geriatrics and Organ Transplantation Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu 610041, China
| | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bei Cai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Yan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yao Luo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
12
|
Fahmy SA, Elghanam R, Rashid G, Youness RA, Sedky NK. Emerging tendencies for the nano-delivery of gambogic acid: a promising approach in oncotherapy. RSC Adv 2024; 14:4666-4691. [PMID: 38318629 PMCID: PMC10840092 DOI: 10.1039/d3ra08042k] [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: 11/23/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
Despite the advancements in cancer therapies during the past few years, chemo/photo resistance, severe toxic effects, recurrence of metastatic tumors, and non-selective targeting remain incomprehensible. Thus, much effort has been spent exploring natural anticancer compounds endowed with biosafety and high effectiveness in cancer prevention and therapy. Gambogic acid (GA) is a promising natural compound in cancer therapy. It is the major xanthone component of the dry resin extracted from the Garcinia hanburyi Hook. f. tree. GA has significant antiproliferative effects on different types of cancer, and it exerts its anticancer activities through various pathways. Nonetheless, the clinical translation of GA has been hampered, partly due to its water insolubility, low bioavailability, poor pharmacokinetics, rapid plasma clearance, early degradation in blood circulation, and detrimental vascular irritation. Lately, procedures have been invented demonstrating the ability of nanoparticles to overcome the challenges associated with the clinical use of natural compounds both in vitro and in vivo. This review sheds light on the recent emerging trends for the nanodelivery of GA to cancer cells. To the best of our knowledge, no similar recent review described the different nanoformulations designed to improve the anticancer therapeutic activity and targeting ability of GA.
Collapse
Affiliation(s)
- Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt +20 1222613344
| | - Rawan Elghanam
- Nanotechnology Department, School of Sciences & Engineering, The American University in Cairo AUC Avenue, P.O. Box 74 New Cairo 11835 Egypt
| | - Gowhar Rashid
- Amity Medical School, Amity University Gurugram Haryana 122413 India
| | - Rana A Youness
- Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU) Cairo 11835 Egypt
| | - Nada K Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo Egypt
| |
Collapse
|
13
|
Zhang Z, Zhao Y, Wang Y, Zhao Y, Guo J. Autophagy/ferroptosis in colorectal cancer: Carcinogenic view and nanoparticle-mediated cell death regulation. ENVIRONMENTAL RESEARCH 2023; 238:117006. [PMID: 37669735 DOI: 10.1016/j.envres.2023.117006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 09/07/2023]
Abstract
The cell death mechanisms have a long history of being evaluated in diseases and pathological events. The ability of triggering cell death is considered to be a promising strategy in cancer therapy, but some mechanisms have dual functions in cancer, requiring more elucidation of underlying factors. Colorectal cancer (CRC) is a disease and malignant condition of colon and rectal that causes high mortality and morbidity. The autophagy targeting in CRC is therapeutic importance and this cell death mechanism can interact with apoptosis in inhibiting or increasing apoptosis. Autophagy has interaction with ferroptosis as another cell death pathway in CRC and can accelerate ferroptosis in suppressing growth and invasion. The dysregulation of autophagy affects the drug resistance in CRC and pro-survival autophagy can induce drug resistance. Therefore, inhibition of protective autophagy enhances chemosensitivity in CRC cells. Moreover, autophagy displays interaction with metastasis and EMT as a potent regulator of invasion in CRC cells. The same is true for ferroptosis, but the difference is that function of ferroptosis is determined and it can reduce viability. The lack of ferroptosis can cause development of chemoresistance in CRC cells and this cell death mechanism is regulated by various pathways and mechanisms that autophagy is among them. Therefore, current review paper provides a state-of-art analysis of autophagy, ferroptosis and their crosstalk in CRC. The nanoparticle-mediated regulation of cell death mechanisms in CRC causes changes in progression. The stimulation of ferroptosis and control of autophagy (induction or inhibition) by nanoparticles can impair CRC progression. The engineering part of nanoparticle synthesis to control autophagy and ferroptosis in CRC still requires more attention.
Collapse
Affiliation(s)
- Zhibin Zhang
- Chengde Medical College, College of Traditional Chinese Medicine, Chengde, Hebei, 067000, China.
| | - Yintao Zhao
- Chengde Medical College, Chengde, Hebei, 067000, China
| | - Yuman Wang
- Chengde Medical College, Chengde, Hebei, 067000, China
| | - Yutang Zhao
- Chengde Medical College, Chengde, Hebei, 067000, China
| | - Jianen Guo
- Chengde Medical College, Chengde, Hebei, 067000, China
| |
Collapse
|