1
|
Giram P, Md Mahabubur Rahman K, Aqel O, You Y. In Situ Cancer Vaccines: Redefining Immune Activation in the Tumor Microenvironment. ACS Biomater Sci Eng 2025. [PMID: 40223683 DOI: 10.1021/acsbiomaterials.5c00121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Cancer is one of the leading causes of mortality worldwide. Nanomedicines have significantly improved life expectancy and survival rates for cancer patients in current standard care. However, recurrence of cancer due to metastasis remains a significant challenge. Vaccines can provide long-term protection and are ideal for preventing bacterial and viral infections. Cancer vaccines, however, have shown limited therapeutic efficacy and raised safety concerns despite extensive research. Cancer vaccines target and stimulate responses against tumor-specific antigens and have demonstrated great potential for cancer treatment in preclinical studies. However, tumor-associated immunosuppression and immune tolerance driven by immunoediting pose significant challenges for vaccine design. In situ vaccination represents an alternative approach to traditional cancer vaccines. This strategy involves the intratumoral administration of immunostimulants to modulate the growth and differentiation of innate immune cells, such as dendritic cells, macrophages, and neutrophils, and restore T-cell activity. Currently approved in situ vaccines, such as T-VEC, have demonstrated clinical promise, while ongoing clinical trials continue to explore novel strategies for broader efficacy. Despite these advancements, failures in vaccine research highlight the need to address tumor-associated immune suppression and immune escape mechanisms. In situ vaccination strategies combine innate and adaptive immune stimulation, leveraging tumor-associated antigens to activate dendritic cells and cross-prime CD8+ T cells. Various vaccine modalities, such as nucleotide-based vaccines (e.g., RNA and DNA vaccines), peptide-based vaccines, and cell-based vaccines (including dendritic, T-cell, and B-cell approaches), show significant potential. Plant-based viral approaches, including cowpea mosaic virus and Newcastle disease virus, further expand the toolkit for in situ vaccination. Therapeutic modalities such as chemotherapy, radiation, photodynamic therapy, photothermal therapy, and Checkpoint blockade inhibitors contribute to enhanced antigen presentation and immune activation. Adjuvants like CpG-ODN and PRR agonists further enhance immune modulation and vaccine efficacy. The advantages of in situ vaccination include patient specificity, personalization, minimized antigen immune escape, and reduced logistical costs. However, significant barriers such as tumor heterogeneity, immune evasion, and logistical challenges remain. This review explores strategies for developing potent cancer vaccines, examines ongoing clinical trials, evaluates immune stimulation methods, and discusses prospects for advancing in situ cancer vaccination.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Kazi Md Mahabubur Rahman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Osama Aqel
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| |
Collapse
|
2
|
Chen L, Tang W, Liu J, Zhu M, Mu W, Tang X, Liu T, Zhu Z, Weng L, Cheng Y, Zhang Y, Chen X. On-demand reprogramming of immunosuppressive microenvironment in tumor tissue via multi-regulation of carcinogenic microRNAs and RNAs dependent photothermal-immunotherapy using engineered gold nanoparticles for malignant tumor treatment. Biomaterials 2025; 315:122956. [PMID: 39549441 DOI: 10.1016/j.biomaterials.2024.122956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
The frequent immune escape of tumor cells and fluctuating therapeutic efficiency vary with each individual are two critical issues for immunotherapy against malignant tumor. Herein, we fabricated an intelligent core-shell nanoparticle (SNAs@CCMR) to significantly inhibit the PD-1/PD-L1 mediated immune escape by on-demand regulation of various oncogenic microRNAs and perform RNAs dependent photothermal-immunotherapy to achieve precise and efficient treatment meeting the individual requirements of specific patients by in situ generation of customized tumor-associated antigens. The SNAs@CCMR consisted of antisense oligonucleotides grafted gold nanoparticles (SNAs) as core and TLR7 agonist imiquimod (R837) functionalized cancer cell membrane (CCM) as shell, in which the acid-labile Schiff base bond was used to connect the R837 and CCM. During therapy, the acid environment of tumor tissue cleaved the Schiff base to generate free R837 and SNAs@CCM. The SNAs@CCM further entered tumor cells via CCM mediated internalization, and then specifically hybridized with over-expressed miR-130a and miR-21, resulting in effective inhibition of the migration and PD-L1 expression of tumor cells to avoid their immune escape. Meanwhile, the RNAs capture also caused significant aggregation of SNAs, which immediately generated photothermal agents within tumor cells to perform highly selective photothermal therapy under NIR irradiation. These chain processes not only damaged the primary tumor, but also produced plenty of tumor-associated antigens, which matured the surrounding dendritic cells (DCs) and activated anti-tumor T cells along with the released R837, resulting in the enhanced immunotherapy with suppressive immune escape. Both in vivo and in vitro experiments demonstrated that our nanoparticles were able to inhibit primary tumor and its metastasis via multi-regulation of carcinogenic microRNAs and RNAs dependent photothermal-immune activations, which provided a promising strategy to reprogram the immunosuppressive microenvironment in tumor tissue for better malignant tumor therapy.
Collapse
Affiliation(s)
- Li Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Wenjun Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jie Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wenyun Mu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiaoyu Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tao Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lin Weng
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yumeng Cheng
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
3
|
Kim D, Choi J, Jin D, Xu E, Lee J, Byun J, Oh YK. Hybrid lipid nanoparticles with tumor antigen-primed dendritic cell membranes for post-surgical tumor immunotherapy. J Control Release 2025; 379:537-548. [PMID: 39800239 DOI: 10.1016/j.jconrel.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/22/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Post-surgical tumor recurrence poses a major challenge in cancer treatment due to residual tumor cells and surgery-induced immunosuppression. Here, we developed hybrid nanoparticles, termed T-DCNPs, designed to promote antigen-specific activation of cytotoxic CD8+ T cells while concurrently inhibiting immunosuppressive pathways within the tumor microenvironment. T-DCNPs were formulated by co-extruding lipid nanoparticles containing a transforming growth factor β inhibitor with dendritic cells that were pre-treated with autologous neoantigens derived from surgically excised tumors. By using whole tumor antigens rather than specific peptides, T-DCNPs effectively overcame tumor heterogeneity and elicited a robust, targeted immune response. In vitro studies showed that T-DCNPs enhanced CD8+ T cell proliferation and reduced programmed death-1 (PD-1) expression, leading to increased antitumor cytotoxicity. In vivo experiments, involving intratumoral injections of T-DCNPs in distant tumor and post-surgical melanoma models, demonstrated a significant reduction in distant tumor growth, decreased recurrence rates, and extended survival compared to control groups. Flow cytometry and immunohistochemistry analyses further confirmed the enhanced infiltration of activated CD8+ T cells and a marked reduction in immunosuppressive markers, including PD-1 and Foxp3, within the treated tumors. These results suggest that T-DCNPs, through the dual mechanisms of tumor antigen-specific T cell activation and immune modulation, offer a promising strategy to prevent tumor recurrence following surgery and could potentially improve the efficacy of postoperative cancer immunotherapy.
Collapse
Affiliation(s)
- Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaehyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Dongun Jin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea.
| | - Junho Byun
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
4
|
Song T, Cui X, Lin J, Liu Z, Huang L, Xue W. Enhanced Antigen Capture via Cholinephosphate-Mediated Cell Membrane Interactions to Improve In Situ Tumor Vaccines. Adv Healthc Mater 2025; 14:e2403460. [PMID: 39901376 DOI: 10.1002/adhm.202403460] [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: 09/11/2024] [Revised: 12/20/2024] [Indexed: 02/05/2025]
Abstract
Inadequate antigen capture and insufficient antigen-presenting cell (APC) activity at tumor sites limit the effectiveness of in situ vaccines. To address this, poly(glutamic acid-cholinephosphate) (pGluCP) is introduced as a polymer with cell membrane adhesion properties capable of capturing both water-soluble and insoluble membrane antigens from necrotic tumor cells while recruiting more APCs. The approach uses manganese-mineralized black phosphorus (MnBP) coated with pGluCP and αPD-1 antibodies to create the MnBP@pGluCP-αPD-1 complex for in situ vaccines. MnBP eradicates tumor cells via photothermal effects, releasing antigens, while Mn2⁺ ions activate the intracellular STING pathway, acting as an adjuvant. pGluCP captures these antigens, forming pathogen-mimicking micro-nanoparticles, leading to an in situ vaccine (MnBP@pGluCP/antigens) that co-localizes antigens and adjuvants. The αPD-1 antibody alleviates tumor-induced immune suppression, enhancing tumor cell-specific killing. This study demonstrates the potential of leveraging cholinephosphate-cell membrane interactions to improve antigen presentation efficiency, significantly bolstering the efficacy of in situ tumor vaccines and opening new avenues for advanced cancer immunotherapy.
Collapse
Affiliation(s)
- Ting Song
- Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Xin Cui
- Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Jiansheng Lin
- Department of Anatomy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zonghua Liu
- Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Linghong Huang
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510230, China
| | - Wei Xue
- Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
5
|
Xu R, Wang S, Guo Q, Zhong R, Chen X, Xia X. Anti-Tumor Strategies of Photothermal Therapy Combined with Other Therapies Using Nanoplatforms. Pharmaceutics 2025; 17:306. [PMID: 40142970 PMCID: PMC11944535 DOI: 10.3390/pharmaceutics17030306] [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: 01/17/2025] [Revised: 02/02/2025] [Accepted: 02/15/2025] [Indexed: 03/28/2025] Open
Abstract
Conventional cancer treatments often have complications and serious side effects, with limited improvements in 5-year survival and quality of life. Photothermal therapy (PTT) employs materials that convert light to heat when exposed to near-infrared light to raise the temperature of the tumor site to directly ablate tumor cells, induce immunogenic cell death, and improve the tumor microenvironment. This therapy has several benefits, including minimal invasiveness, high efficacy, reduced side effects, and robust targeting capabilities. Beyond just photothermal conversion materials, nanoplatforms significantly contribute to PTT by supplying effective photothermal conversion materials and bolstering tumor targeting to amplify anti-tumor effects. However, the anti-tumor effects of PTT alone are ultimately limited and often need to be combined with other therapies. This narrative review describes the recent progress of PTT combined with chemotherapy, radiotherapy, photodynamic therapy, immunotherapy, gene therapy, gas therapy, chemodynamic therapy, photoacoustic imaging, starvation therapy, and multimodal therapy. Studies have shown that combining PTT with other treatments can improve efficacy, reduce side effects, and overcome drug resistance. Despite the encouraging results, challenges such as optimizing treatment protocols, addressing tumor heterogeneity, and overcoming biological barriers remain. This paper highlights the potential for personalized, multimodal approaches to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Rubing Xu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China (Q.G.)
| | - Shengmei Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China (Q.G.)
| | - Ruqian Zhong
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China (Q.G.)
| | - Xi Chen
- Hunan Provincial Center for Drug Evaluation and Adverse Reaction Monitoring, Changsha 410013, China;
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China (Q.G.)
| |
Collapse
|
6
|
Chiang JY, Lin TH, Cheng JX, Pan WY. Polyethyleneimine/fucoidan polyplexes as vaccine carriers for enhanced antigen loading and dendritic cell activation. Int J Biol Macromol 2025; 306:141336. [PMID: 39986515 DOI: 10.1016/j.ijbiomac.2025.141336] [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: 09/26/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Vaccination is one of the most effective strategies for preventing infectious diseases. Recently, most research has centered on the development of protein subunit vaccines due to their safety. However, their low immunogenicity remains a challenge. Nanoparticle vaccines offer advantages by protecting proteins from degradation and acting as adjuvants to stimulate the immune system. Herein, a polyplexe (OVA@PEI/Fu) formed by the electrostatic interaction between positively charged polyethyleneimine (PEI) and negatively charged fucoidan was prepared for the encapsulation of a model antigen, ovalbumin (OVA). Experimental results revealed that the incorporation of fucoidan in the polyplexes not only enhanced OVA loading efficiency but also contributed adjuvant effects, significantly boosting dendritic cell activation and maturation in vitro compared to OVA@PEI polyplexes. In vivo experiments showed that the OVA@PEI/Fu can induce strong anti-OVA specific antibody responses, as well as OVA-specific CD4+ and CD8+ T cell responses. The carrier developed in the present study shows promise as a platform for protein-based subunit vaccines.
Collapse
Affiliation(s)
- Jian-Yi Chiang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Tsai-Hsuan Lin
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Jia-Xiang Cheng
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Yu Pan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan.
| |
Collapse
|
7
|
Zhang S, Wang H. Targeting the lung tumour stroma: harnessing nanoparticles for effective therapeutic interventions. J Drug Target 2025; 33:60-86. [PMID: 39356091 DOI: 10.1080/1061186x.2024.2410462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Lung cancer remains an influential global health concern, necessitating the development of innovative therapeutic strategies. The tumour stroma, which is known as tumour microenvironment (TME) has a central impact on tumour expansion and treatment resistance. The stroma of lung tumours consists of numerous cells and molecules that shape an environment for tumour expansion. This environment not only protects tumoral cells against immune system attacks but also enables tumour stroma to attenuate the action of antitumor drugs. This stroma consists of stromal cells like cancer-associated fibroblasts (CAFs), suppressive immune cells, and cytotoxic immune cells. Additionally, the presence of stem cells, endothelial cells and pericytes can facilitate tumour volume expansion. Nanoparticles are hopeful tools for targeted drug delivery because of their extraordinary properties and their capacity to devastate biological obstacles. This review article provides a comprehensive overview of contemporary advancements in targeting the lung tumour stroma using nanoparticles. Various nanoparticle-based approaches, including passive and active targeting, and stimuli-responsive systems, highlighting their potential to improve drug delivery efficiency. Additionally, the role of nanotechnology in modulating the tumour stroma by targeting key components such as immune cells, extracellular matrix (ECM), hypoxia, and suppressive elements in the lung tumour stroma.
Collapse
Affiliation(s)
- Shushu Zhang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Hui Wang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
8
|
Chen Y, Mao K, Han D, Ma R, Sun T, Zhang H, Han B. Nanomedicine based on chemotherapy-induced immunogenic death combined with immunotherapy to enhance antitumor immunity. Front Pharmacol 2024; 15:1511423. [PMID: 39697556 PMCID: PMC11652165 DOI: 10.3389/fphar.2024.1511423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Introduction Chemo-immunotherapy based on inducing tumor immunogenic cell death (ICD)with chemotherapy drugs has filled the gaps between traditional chemotherapy and immunotherapy. It is verified that paclitaxel (PTX) can induce breast tumor ICD. From this basis, a kind of nanoparticle that can efficiently deliver different drug components simultaneously is constructed. The purpose of this study is for the sake of exploring the scheme of chemotherapy-induced ICD combined with other immunotherapy to enhance tumor immunogenicity and inhibit the growth, metastasis, and recurrence of breast tumors, so as to provide a research basis for solving the tough problem of breast cancer treatment. Methods Nanomedicine loaded with PTX, small interference RNA that suppresses CD47 expression (CD47siRNA, siCD47), and immunomodulator R848 were prepared by the double emulsification method. The hydrodynamic diameter and zeta potential of NP/PTX/siCD47/R848 were characterized. Established the tumor-bearing mice model of mouse breast cancer cell line (4T1) in situ and observed the effect of intravenous injection of NP/PTX/siCD47/R848 on the growth of 4T1 tumor in situ. Flow cytometry was used to detect the effect of drugs on tumor immune cells. Results NP/PTX/siCD47/R848 nano-drug with tumor therapeutic potential were successfully prepared by double emulsification method, with particle size of 121.5 ± 4.5 nm and surface potential of 36.1 ± 2.5 mV. The calreticulin on the surface of cell membrane and extracellular ATP or HMGB1 of 4T1 cells increased through treatment with NPs. NP/PTX-treated tumor cells could cause activation of BMDCs and BMDMs. After intravenous injection, NP/PTX could quickly reach the tumor site and accumulate for 24 h. The weight and volume of tumor in situ in the breast cancer model mice injected with nanomedicine through the tail vein were significantly lower than those in the PBS group. The ratio of CD8+/CD4+ T cells in the tumor microenvironment and the percentage of dendritic cells in peripheral blood increased significantly in breast cancer model mice injected with nano-drugs through the tail vein. Discussion Briefly, the chemotherapeutic drug paclitaxel can induce breast cancer to induce ICD. The nanomedicine which can deliver PTX, CD47siRNA, and R848 at the same time was prepared by double emulsification. NP/PTX/siCD47/R848 nano-drug can be enriched in the tumor site. The experiment of 4T1 cell tumor-bearing mice shows that the nano-drug can enhance tumor immunogenicity and inhibit breast tumor growth, which provides a new scheme for breast cancer treatment. (Graphical abstract).
Collapse
Affiliation(s)
- Yichang Chen
- Department of Breast Surgery, General Surgery Center of The First Hospital, Jilin University, Changchun, China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Institute of Immunology, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Dongxiao Han
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Institute of Immunology, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Ruolin Ma
- Department of Breast Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Institute of Immunology, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Haipeng Zhang
- Department of Gynecology, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Changchun, China
| | - Bing Han
- Department of Breast Surgery, General Surgery Center of The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
9
|
Zhu Y, Ke L, Wei Y, Zheng X. A High-Precision Real-Time Temperature Acquisition Method Based on Magnetic Nanoparticles. SENSORS (BASEL, SWITZERLAND) 2024; 24:7716. [PMID: 39686253 DOI: 10.3390/s24237716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024]
Abstract
The unique magnetothermal properties of magnetic nanoparticles enable the development of a high-precision, real-time, noninvasive temperature measurement method with significant potential in the biomedical field. Based on a low-frequency alternating magnetic field excitation model, we construct two additional magnetic field excitation models-alternating current-direct current superposition and dual-frequency superposition-to extract harmonic amplitude components from the magnetization response. To increase the accuracy of harmonic information acquisition, the effects of the truncation error, excitation magnetic field frequency, and amplitude are thoroughly analyzed, and optimal parameter values are selected to minimize the error. A single algorithm is designed for temperature inversion, and a joint algorithm is proposed to optimize the performance of the single algorithm. Under low-frequency alternating-current magnetic field excitation, the autonomous group particle swarm optimization method achieves superior real-time performance in terms of temperature inversion and running time. Compared with the opposition learning gray wolf optimizer and particle swarm optimization-gray wolf optimization, the proposed method achieves reductions of 52% and 68%, respectively. Additionally, under dual-frequency superimposed magnetic field excitation, a higher temperature inversion accuracy is achieved compared with that of the particle swarm optimization-gray wolf optimization algorithm, reducing the error from 0.237 K to 0.094 K.
Collapse
Affiliation(s)
- Yuchang Zhu
- School of Electrical Engineering, Shenyang University of Technology, Shenyang 110870, China
| | - Li Ke
- School of Electrical Engineering, Shenyang University of Technology, Shenyang 110870, China
| | - Yijing Wei
- School of Electrical Engineering, Shenyang University of Technology, Shenyang 110870, China
| | - Xiao Zheng
- CAS State Key Laboratory of Forest and Management, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China
| |
Collapse
|
10
|
Li B, Chen G, Zhong H, Li T, Lin M, Wei H, Zhang Q, Chen Q, Huang J, Shuai X. γ-Glutamyl transpeptidase-activable nanoprobe crosses the blood-brain barrier for immuno-sonodynamic therapy of glioma. Nat Commun 2024; 15:10418. [PMID: 39613729 DOI: 10.1038/s41467-024-54382-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/05/2024] [Indexed: 12/01/2024] Open
Abstract
Effective treatment against glioma remains challenging nowadays because the protective blood-brain barrier (BBB) impedes drug penetration into brain and the limited efficacy of conventional chemotherapy. While strong positively charged nanoparticles have good permeability through the BBB, they often come with the caveat of cationic toxicity to healthy tissues and organs during blood circulation. Here we show a neutrally charged nanoprobe with a surface decorated with γ-glutamyl moieties that can be cleaved by γ-glutamyl transpeptidase, an enzyme overexpressed on brain capillaries. Upon the cleavage, positively charged primary amines are generated, facilitating the effective crossing of the nanoprobe through BBB via the adsorption-mediated transcytosis pathway, while avoiding the caveat of cationic toxicity. In addition, when reaching the acidic tumor microenvironment, the nanoprobe co-encapsulating sonosensitizer and immune agonist swells, which results in an accelerated drug release under ultrasound irradiation to induce a combined immune response, ultimately leading to a robust anticancer effect. Overall, we report an effective drug delivery nanoplatform across the BBB for an enhanced therapy of glioma.
Collapse
Affiliation(s)
- Bo Li
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Gengjia Chen
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Huihai Zhong
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Tan Li
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Minzhao Lin
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Huiye Wei
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Qiaoyun Zhang
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Qi Chen
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
11
|
Cai W, Sun T, Qiu C, Sheng H, Chen R, Xie C, Kou L, Yao Q. Stable triangle: nanomedicine-based synergistic application of phototherapy and immunotherapy for tumor treatment. J Nanobiotechnology 2024; 22:635. [PMID: 39420366 PMCID: PMC11488210 DOI: 10.1186/s12951-024-02925-3] [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: 10/03/2023] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
In recent decades, cancer has posed a challenging obstacle that humans strive to overcome. While phototherapy and immunotherapy are two emerging therapies compared to traditional methods, they each have their advantages and limitations. These limitations include easy metastasis and recurrence, low response rates, and strong side effects. To address these issues, researchers have increasingly focused on combining these two therapies by utilizing a nano-drug delivery system due to its superior targeting effect and high drug loading rate, yielding remarkable results. The combination therapy demonstrates enhanced response efficiency and effectiveness, leading to a preparation that is highly targeted, responsive, and with low recurrence rates. This paper reviews several main mechanisms of anti-tumor effects observed in combination therapy based on the nano-drug delivery system over the last five years. Furthermore, the challenges and future prospects of this combination therapy are also discussed.
Collapse
Affiliation(s)
- Wenjing Cai
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Tuyue Sun
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chenyu Qiu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Huixiang Sheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Congying Xie
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Wenzhou, 325000, China.
- Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou, 325000, China.
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Wenzhou, 325000, China.
- Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou, 325000, China.
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
12
|
Dai X, Liu Z, Zhao X, Guo K, Ding X, Xu FJ, Zhao N. NIR-II-Responsive Hybrid System Achieves Cascade-Augmented Antitumor Immunity via Genetic Engineering of Both Bacteria and Tumor Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407927. [PMID: 39185788 DOI: 10.1002/adma.202407927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/09/2024] [Indexed: 08/27/2024]
Abstract
The combination of nanoparticles and tumor-targeting bacteria for cancer immunotherapy can overcome the shortcomings of poor nanoparticle accumulation, limited penetration, and restricted distribution. However, it remains a great challenge for the hybrid system to improve therapeutic efficacy through the simultaneous and controllable regulation of immune cells and tumor cells. Herein, a hybrid therapeutic platform is rationally designed to achieve immune cascade-augmented cancer immunotherapy. To construct the hybrids, photothermal nanoparticles responsive to light in the second near-infrared (NIR-II) region are conjugated onto the surface of engineered bacteria through pH-responsive Schiff base bonds. Taking advantage of the hypoxia targeting and deep penetration characteristics of the bacteria, the hybrids can accumulate at tumor sites. Then nanoparticles detach from the bacteria to realize genetic engineering of tumor cells, which induces tumor cell apoptosis and down-regulate the expression of programmed cell death ligand 1 to alleviate immunosuppressive tumor microenvironment. The mild photothermal heating can not only induce tumor-associated antigen release, but also trigger sustainable expression of cytokine interleukin-2. Notably, a synergistic antitumor effect is achieved between the process of p53 transfection and NIR-II light-activated genetic engineering of bacteria. This work proposes a facile strategy for the construction of hybrid system to achieve cascade-augmented cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaoguang Dai
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhiwen Liu
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoyi Zhao
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Kangli Guo
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaokang Ding
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Nana Zhao
- State Key Laboratory of Chemical Resource Engineering, Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
13
|
Wang Z, Sha T, Li J, Luo H, Liu A, Liang H, Qiang J, Li L, Whittaker AK, Yang B, Sun H, Shi C, Lin Q. Turning foes to friends: Advanced " in situ nanovaccine" with dual immunoregulation for enhanced immunotherapy of metastatic triple-negative breast cancer. Bioact Mater 2024; 39:612-629. [PMID: 38883315 PMCID: PMC11179173 DOI: 10.1016/j.bioactmat.2024.04.023] [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: 02/19/2024] [Revised: 04/21/2024] [Accepted: 04/21/2024] [Indexed: 06/18/2024] Open
Abstract
As a "cold tumor", triple-negative breast cancer (TNBC) exhibits limited responsiveness to current immunotherapy. How to enhance the immunogenicity and reverse the immunosuppressive microenvironment of TNBC remain a formidable challenge. Herein, an "in situ nanovaccine" Au/CuNDs-R848 was designed for imaging-guided photothermal therapy (PTT)/chemodynamic therapy (CDT) synergistic therapy to trigger dual immunoregulatory effects on TNBC. On the one hand, Au/CuNDs-R848 served as a promising photothermal agent and nanozyme, achieving PTT and photothermal-enhanced CDT against the primary tumor of TNBC. Meanwhile, the released antigens and damage-associated molecular patterns (DAMPs) promoted the maturation of dendritic cells (DCs) and facilitated the infiltration of T lymphocytes. Thus, Au/CuNDs-R848 played a role as an "in situ nanovaccine" to enhance the immunogenicity of TNBC by inducing immunogenic cell death (ICD). On the other hand, the nanovaccine suppressed the myeloid-derived suppressor cells (MDSCs), thereby reversing the immunosuppressive microenvironment. Through the dual immunoregulation, "cold tumor" was transformed into a "hot tumor", not only implementing a "turning foes to friends" therapeutic strategy but also enhancing immunotherapy against metastatic TNBC. Furthermore, Au/CuNDs-R848 acted as an excellent nanoprobe, enabling high-resolution near-infrared fluorescence and computed tomography imaging for precise visualization of TNBC. This feature offers potential applications in clinical cancer detection and surgical guidance. Collectively, this work provides an effective strategy for enhancing immune response and offers novel insights into the potential clinical applications for tumor immunotherapy.
Collapse
Affiliation(s)
- Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Tong Sha
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Jinwei Li
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Huanyu Luo
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Hao Liang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Jinbiao Qiang
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Lei Li
- Department of Endocrinology, Lequn Branch, The First Hospital of Jilin University, 130021, PR China
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Hongchen Sun
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Ce Shi
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| |
Collapse
|
14
|
Wu L, Yang L, Qian X, Hu W, Wang S, Yan J. Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine Increased the Antitumor Immune Response by Modulating the Tumor Microenvironment. J Funct Biomater 2024; 15:229. [PMID: 39194667 DOI: 10.3390/jfb15080229] [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: 05/23/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
With the rapid development of tumor immunotherapy, nanoparticle vaccines have attracted much attention as potential therapeutic strategies. A systematic review and analysis must be carried out to investigate the effect of mannose modification on the immune response to nanoparticles in regulating the tumor microenvironment, as well as to explore its potential clinical application in tumor therapy. Despite the potential advantages of nanoparticle vaccines in immunotherapy, achieving an effective immune response in the tumor microenvironment remains a challenge. Tumor immune escape and the overexpression of immunosuppressive factors limit its clinical application. Therefore, our review explored how to intervene in the immunosuppressive mechanism in the tumor microenvironment through the use of mannan-decorated lipid calcium phosphate nanoparticle vaccines to improve the efficacy of immunotherapy in patients with tumors and to provide new ideas and strategies for the field of tumor therapy.
Collapse
Affiliation(s)
- Liusheng Wu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 19077, Singapore
| | - Lei Yang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xinye Qian
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wang Hu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shuang Wang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jun Yan
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
15
|
Lu YJ, Vayalakkara RK, Dash BS, Hu SH, Premji TP, Wu CY, Shen YJ, Chen JP. Immunomodulatory R848-Loaded Anti-PD-L1-Conjugated Reduced Graphene Oxide Quantum Dots for Photothermal Immunotherapy of Glioblastoma. Pharmaceutics 2024; 16:1064. [PMID: 39204409 PMCID: PMC11358977 DOI: 10.3390/pharmaceutics16081064] [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: 06/27/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most severe form of brain cancer and presents unique challenges to developing novel treatments due to its immunosuppressive milieu where receptors like programmed death ligand 1 (PD-L1) are frequently elevated to prevent an effective anti-tumor immune response. To potentially shift the GBM environment from being immunosuppressive to immune-enhancing, we engineered a novel nanovehicle from reduced graphene oxide quantum dot (rGOQD), which are loaded with the immunomodulatory drug resiquimod (R848) and conjugated with an anti-PD-L1 antibody (aPD-L1). The immunomodulatory rGOQD/R8/aPDL1 nanoparticles can actively target the PD-L1 on the surface of ALTS1C1 murine glioblastoma cells and release R848 to enhance the T-cell-driven anti-tumor response. From in vitro experiments, the PD-L1-mediated intracellular uptake and the rGOQD-induced photothermal response after irradiation with near-infrared laser light led to the death of cancer cells and the release of damage-associated molecular patterns (DAMPs). The combinational effect of R848 and released DAMPs synergistically produces antigens to activate dendritic cells, which can prime T lymphocytes to infiltrate the tumor in vivo. As a result, T cells effectively target and attack the PD-L1-suppressed glioma cells and foster a robust photothermal therapy elicited anti-tumor immune response from a syngeneic mouse model of GBM with subcutaneously implanted ALTS1C1 cells.
Collapse
Affiliation(s)
- Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
- College of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Reesha Kakkadavath Vayalakkara
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
| | - Banendu Sunder Dash
- Department of Chemical and Materials and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (B.S.D.)
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Thejas Pandaraparambil Premji
- Department of Chemical and Materials and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (B.S.D.)
| | - Chun-Yuan Wu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
| | - Yang-Jin Shen
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
| | - Jyh-Ping Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
- Department of Chemical and Materials and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (B.S.D.)
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
16
|
Pan X, Ni S, Hu K. Nanomedicines for reversing immunosuppressive microenvironment of hepatocellular carcinoma. Biomaterials 2024; 306:122481. [PMID: 38286109 DOI: 10.1016/j.biomaterials.2024.122481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 01/31/2024]
Abstract
Although immunotherapeutic strategies such as immune checkpoint inhibitors (ICIs) have gained promising advances, their limited efficacy and significant toxicity remain great challenges for hepatocellular carcinoma (HCC) immunotherapy. The tumor immunosuppressive microenvironment (TIME) with insufficient T-cell infiltration and low immunogenicity accounts for most HCC patients' poor response to ICIs. Worse still, the current immunotherapeutics without precise delivery may elicit enormous autoimmune side effects and systemic toxicity in the clinic. With a better understanding of the TIME in HCC, nanomedicines have emerged as an efficient strategy to achieve remodeling of the TIME and superadditive antitumor effects via targeted delivery of immunotherapeutics or multimodal synergistic therapy. Based on the typical characteristics of the TIME in HCC, this review summarizes the recent advancements in nanomedicine-based strategies for TIME-reversing HCC treatment. Additionally, perspectives on the awaiting challenges and opportunities of nanomedicines in modulating the TIME of HCC are presented. Acquisition of knowledge of nanomedicine-mediated TIME reversal will provide researchers with a better opportunity for clinical translation of HCC immunotherapy.
Collapse
Affiliation(s)
- Xier Pan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shuting Ni
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
17
|
Mozafari N, Jahanbekam S, Ashrafi H, Shahbazi MA, Azadi A. Recent Biomaterial-Assisted Approaches for Immunotherapeutic Inhibition of Cancer Recurrence. ACS Biomater Sci Eng 2024; 10:1207-1234. [PMID: 38416058 DOI: 10.1021/acsbiomaterials.3c01347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Biomaterials possess distinctive properties, notably their ability to encapsulate active biological products while providing biocompatible support. The immune system plays a vital role in preventing cancer recurrence, and there is considerable demand for an effective strategy to prevent cancer recurrence, necessitating effective strategies to address this concern. This review elucidates crucial cellular signaling pathways in cancer recurrence. Furthermore, it underscores the potential of biomaterial-based tools in averting or inhibiting cancer recurrence by modulating the immune system. Diverse biomaterials, including hydrogels, particles, films, microneedles, etc., exhibit promising capabilities in mitigating cancer recurrence. These materials are compelling candidates for cancer immunotherapy, offering in situ immunostimulatory activity through transdermal, implantable, and injectable devices. They function by reshaping the tumor microenvironment and impeding tumor growth by reducing immunosuppression. Biomaterials facilitate alterations in biodistribution, release kinetics, and colocalization of immunostimulatory agents, enhancing the safety and efficacy of therapy. Additionally, how the method addresses the limitations of other therapeutic approaches is discussed.
Collapse
Affiliation(s)
- Negin Mozafari
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Sheida Jahanbekam
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Hajar Ashrafi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Amir Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| |
Collapse
|
18
|
Ngo TLH, Wang KL, Pan WY, Ruan T, Lin YJ. Immunomodulatory Prodrug Micelles Imitate Mild Heat Effects to Reshape Tumor Microenvironment for Enhanced Cancer Immunotherapy. ACS NANO 2024; 18:5632-5646. [PMID: 38344992 PMCID: PMC10883120 DOI: 10.1021/acsnano.3c11186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Physical stimulation with mild heat possesses the notable ability to induce immunomodulation within the tumor microenvironment (TME). It transforms the immunosuppressive TME into an immune-active state, making tumors more receptive to immune checkpoint inhibitor (ICI) therapy. Transient receptor potential vanilloid 1 (TRPV1), which can be activated by mild heat, holds the potential to induce these alterations in the TME. However, achieving precise temperature control within tumors while protecting neighboring tissues remains a significant challenge when using external heat sources. Taking inspiration from the heat sensation elicited by capsaicin-containing products activating TRPV1, this study employs capsaicin to chemically stimulate TRPV1, imitating immunomodulatory benefits akin to those induced by mild heat. This involves developing a glutathione (GSH)-responsive immunomodulatory prodrug micelle system to deliver capsaicin and an ICI (BMS202) concurrently. Following intravenous administration, the prodrug micelles accumulate at the tumor site through the enhanced permeability and retention effect. Within the GSH-rich TME, the micelles disintegrate and release capsaicin and BMS202. The released capsaicin activates TRPV1 expressed in the TME, enhancing programmed death ligand 1 expression on tumor cell surfaces and promoting T cell recruitment into the TME, rendering it more immunologically active. Meanwhile, the liberated BMS202 blocks immune checkpoints on tumor cells and T cells, activating the recruited T cells and ultimately eradicating the tumors. This innovative strategy represents a comprehensive approach to fine-tune the TME, significantly amplifying the effectiveness of cancer immunotherapy by exploiting the TRPV1 pathway and enabling in situ control of immunomodulation within the TME.
Collapse
Affiliation(s)
- Thi-Lan-Huong Ngo
- Research Center for Applied Sciences, Academia Sinica, Taipei, 115201, Taiwan
| | - Kuan-Lin Wang
- Research Center for Applied Sciences, Academia Sinica, Taipei, 115201, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, 242062, Taiwan
| | - Wen-Yu Pan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, 110301, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, 110301, Taiwan
| | - Ting Ruan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, 242062, Taiwan
| | - Yu-Jung Lin
- Research Center for Applied Sciences, Academia Sinica, Taipei, 115201, Taiwan
| |
Collapse
|
19
|
Lei L, Liu P, Jing W, Wu Z. Advancements in modifying the efficacy of immunotherapies through the thermal effects of nanomaterials. NANO TRANSMED 2023; 2:100022. [DOI: 10.1016/j.ntm.2023.100022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
|
20
|
Wang Q, Xia G, Li J, Yuan L, Yu S, Li D, Yang N, Fan Z, Li J. Multifunctional Nanoplatform for NIR-II Imaging-Guided Synergistic Oncotherapy. Int J Mol Sci 2023; 24:16949. [PMID: 38069279 PMCID: PMC10707236 DOI: 10.3390/ijms242316949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Tumors are a major public health issue of concern to humans, seriously threatening the safety of people's lives and property. With the increasing demand for early and accurate diagnosis and efficient treatment of tumors, noninvasive optical imaging (including fluorescence imaging and photoacoustic imaging) and tumor synergistic therapies (phototherapy synergistic with chemotherapy, phototherapy synergistic with immunotherapy, etc.) have received increasing attention. In particular, light in the near-infrared second region (NIR-II) has triggered great research interest due to its penetration depth, minimal tissue autofluorescence, and reduced tissue absorption and scattering. Nanomaterials with many advantages, such as high brightness, great photostability, tunable photophysical properties, and excellent biosafety offer unlimited possibilities and are being investigated for NIR-II tumor imaging-guided synergistic oncotherapy. In recent years, many researchers have tried various approaches to investigate nanomaterials, including gold nanomaterials, two-dimensional materials, metal sulfide oxides, polymers, carbon nanomaterials, NIR-II dyes, and other nanomaterials for tumor diagnostic and therapeutic integrated nanoplatform construction. In this paper, the application of multifunctional nanomaterials in tumor NIR-II imaging and collaborative therapy in the past three years is briefly reviewed, and the current research status is summarized and prospected, with a view to contributing to future tumor therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhongxiong Fan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (Q.W.); (G.X.); (J.L.); (L.Y.); (S.Y.); (D.L.); (N.Y.)
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology & Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; (Q.W.); (G.X.); (J.L.); (L.Y.); (S.Y.); (D.L.); (N.Y.)
| |
Collapse
|
21
|
Lu S, Wu Y, Liu Y, Sun X, Li J, Li J. Multifunctional Photothermal Hydrogel in the Second Near-Infrared Window for Localized Tumor Therapy. ACS APPLIED BIO MATERIALS 2023; 6:4694-4702. [PMID: 37824829 DOI: 10.1021/acsabm.3c00492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
A copper selenide-embedded gellan gum hydrogel (Cu2-xSe@GG) is designed as an "all-in-one" antitumor agent. The obtained nanocomposite hydrogel exhibits strong near-infrared light absorption and high photothermal conversion efficiency in both the NIR-I and NIR-II biowindows. The photothermal conversion efficiency achieves 58.8% under the irradiation of 0.75 W/cm2 with a 1064 nm laser. Furthermore, the nanocomposite hydrogel has catalase- and peroxidase-mimicking activities, which could alter the tumor microenvironment by reducing hypoxia and/or increasing the production of reactive oxygen species. Moreover, the multifunctional Cu2-xSe@GG nanocomposite hydrogel can also be used as an immune agonist resiquimod (R848) carrier to promote immune regulation and enhance the therapeutic effect. The single-syringe R848/Cu2-xSe@GG treatment achieves synergetic photothermal immunotherapy, showing 97.4% of tumor regression rate from an initial large tumor of 300 mm3.
Collapse
Affiliation(s)
- Sha Lu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yingjiao Wu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yandi Liu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Xiaoyi Sun
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Jianghua Li
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Juan Li
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha 410008, China
| |
Collapse
|
22
|
Zhang Y, Li Z, Huang Y, Zou B, Xu Y. Amplifying cancer treatment: advances in tumor immunotherapy and nanoparticle-based hyperthermia. Front Immunol 2023; 14:1258786. [PMID: 37869003 PMCID: PMC10587571 DOI: 10.3389/fimmu.2023.1258786] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
In the quest for cancer treatment modalities with greater effectiveness, the combination of tumor immunotherapy and nanoparticle-based hyperthermia has emerged as a promising frontier. The present article provides a comprehensive review of recent advances and cutting-edge research in this burgeoning field and examines how these two treatment strategies can be effectively integrated. Tumor immunotherapy, which harnesses the immune system to recognize and attack cancer cells, has shown considerable promise. Concurrently, nanoparticle-based hyperthermia, which utilizes nanotechnology to promote selective cell death by raising the temperature of tumor cells, has emerged as an innovative therapeutic approach. While both strategies have individually shown potential, combination of the two modalities may amplify anti-tumor responses, with improved outcomes and reduced side effects. Key studies illustrating the synergistic effects of these two approaches are highlighted, and current challenges and future prospects in the field are discussed. As we stand on the precipice of a new era in cancer treatment, this review underscores the importance of continued research and collaboration in bringing these innovative treatments from the bench to the bedside.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Li
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Huang
- College of Management, Sichuan Agricultural University, Chengdu, China
| | - Bingwen Zou
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xu
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Chen KH, Nguyen N, Huang TY, Lin YJ, Yu YT, Song HL, Wang JT, Nguyen VK, Chen HL, Chu LA, Chiang HHK, Sung HW. Macrophage-Hitchhiked Orally Administered β-Glucans-Functionalized Nanoparticles as "Precision-Guided Stealth Missiles" for Targeted Pancreatic Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304735. [PMID: 37363886 DOI: 10.1002/adma.202304735] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/23/2023] [Indexed: 06/28/2023]
Abstract
The prognosis in cases of pancreatic ductal adenocarcinoma (PDAC) with current treatment modalities is poor owing to the highly desmoplastic tumor microenvironment (TME). Herein, a β-glucans-functionalized zinc-doxorubicin nanoparticle system (βGlus-ZnD NPs) that can be orally administered, is developed for targeted PDAC therapy. Following oral administration in PDAC-bearing mice, βGlus-ZnD NPs actively target/transpass microfold cells, overcome the intestinal epithelial barrier, and then undergo subsequent phagocytosis by endogenous macrophages (βGlus-ZnD@Mϕ). As hitchhiking cellular vehicles, βGlus-ZnD@Mϕ transits through the intestinal lymphatic system and enters systemic circulation, ultimately accumulating in the tumor tissue as a result of the tumor-homing and "stealth" properties that are conferred by endogenous Mϕ. Meanwhile, the Mϕ that hitchhikes βGlus-ZnD NPs is activated to produce matrix metalloproteinases, destroying the desmoplastic stromal barrier, and differentiates toward the M1 -like phenotype, modulating the TME and recruiting effector T cells, ultimately inducing apoptosis of the tumor cells. The combination of βGlus-ZnD@Mϕ and immune checkpoint blockade effectively inhibits the growth of the primary tumor and suppresses the development of metastasis. It thus represents an appealing approach to targeted PDAC therapy.
Collapse
Affiliation(s)
- Kuan-Hung Chen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Nhien Nguyen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Tun-Yu Huang
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Yu-Jung Lin
- Research Center for Applied Sciences, 11529, Academia Sinica, Taipei, Taiwan
| | - Yu-Tzu Yu
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Hsiang-Lin Song
- Department of Pathology, National Taiwan University Hospital, 300, Hsinchu Branch, Hsinchu, Taiwan
| | - Jui-To Wang
- Neurological Institute, Department of Neurosurgery, Taipei Veterans General Hospital, 11217, Taipei, Taiwan
- Institute of Brain Science, National Yang-Ming Chiao Tung University, 11221, Taipei, Taiwan
| | - Van Khanh Nguyen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Hsin-Lung Chen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Li-An Chu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Hui-Hua Kenny Chiang
- Institute of Biomedical Engineering, National Yang-Ming Chiao Tung University, 11221, Taipei, Taiwan
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| |
Collapse
|
24
|
Yuan Z, He H, Zou J, Wang H, Chen Y, Chen Y, Lan M, Zhao Y, Gao F. Polydopamine-coated ferric oxide nanoparticles for R848 delivery for photothermal immunotherapy in breast cancer. Int J Pharm 2023; 644:123249. [PMID: 37467816 DOI: 10.1016/j.ijpharm.2023.123249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/02/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023]
Abstract
Breast cancer, which requires comprehensive multifunctional treatment strategies, is a major threat to the health of women. To develop multifunctional treatment strategies, we combined photothermal therapy (PTT) with immunotherapy in multifunctional nanoparticles for enhancing the anti-tumor efficacy. Fe3O4 nanoparticles coated with the polydopamine shell modified with polyethylene glycol and cyclic arginine-glycyl-aspartic peptide/anisamide (tNP) for loading the immune adjuvant resiquimod (R848) (R848@tNP) were developed in this research. R848@tNP had a round-like morphology with a mean diameter of 174.7 ± 3.8 nm, the zeta potential of -20.9 ± 0.9 mV, the drug loading rate of 9.2 ± 1.1 %, the encapsulation efficiency of 81.7 ± 3.2 %, high photothermal conversion efficiency and excellent magnetic properties in vitro. Furthermore, this research also explored the anticancer efficacy of nanoparticles against the breast cancer under the near-infrared (NIR) light (808 nm) in vitro and in vivo. R848@tNP-based NIR therapy effectively inhibited the proliferation of breast cancer cells. Moreover, R848@tNP mediated PTT significantly enhanced the maturation of dendritic cells in vitro. Additionally, R848@tNP enhances the anti-tumor effect and evoked an immune response under NIR in vivo. Furthermore, the biosafety of R848@tNP was fully investigated in this study. Collectively, these results clearly demonstrate that R848@tNP, with magnetic resonance imaging characteristics, is a potential therapeutic for breast cancer that combines PTT with the immunotherapy.
Collapse
Affiliation(s)
- Zeting Yuan
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Hai He
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jiafeng Zou
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Hongtao Wang
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - You Chen
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yang Chen
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Yuzheng Zhao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, 100000 Beijing, China
| | - Feng Gao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
25
|
Wang W, Zhu Q, Jin Y, Gao J, Li J, Zheng X, Gao W, Saeed M, Sheng W, Yu H. Self-Immolated Nanoadjuvant for In Situ Vaccination Immunotherapy of Colorectal Cancer. Adv Healthc Mater 2023; 12:e2300524. [PMID: 37269141 DOI: 10.1002/adhm.202300524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/24/2023] [Indexed: 06/04/2023]
Abstract
Vaccination immunotherapy has revolutionized cancer treatment modalities. Although the immunomodulatory adjuvant generally employs for potentiating vaccine response, systemic administration may drive immune-related side effects, even immune tolerance. Therefore, tunable immunoadjuvants are highly desirable to simultaneously stimulate the immune response and mitigate systemic toxicity. Self-immolated nanoadjuvants are herein reported to potentiate vaccination immunotherapy of cancer. The nanoadjuvants are engineered by co-assembling an intracellular acidity-ionizable polymeric agonist of toll-like receptor 7/8 resiquimod (R848) and polymeric photosensitizer pyropheophorbide a (PPa). The resultant nanoadjuvants specifically accumulate at the tumor site via passive targeting and are dissociated in the acidic endosome versicles to activate PPa via protonation of the polymer backbone. Upon 671 nm laser irradiation, PPa performed photodynamic therapy to induce immunogenic cell death of tumor cells and subsequently releases R848 in a customized manner, which synergistically activates dendritic cells (DCs), promotes antigen cross-presentation, and eventually recruits cytotoxic T lymphocytes for tumor regression. Furthermore, the synergistic in situ vaccination immunotherapy with immune checkpoint blockade induce sustained immunological memory to suppress tumor recurrence in the rechallenged colorectal tumor model.
Collapse
Affiliation(s)
- Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qiwen Zhu
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yilan Jin
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jing Gao
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jianan Li
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Xiaohua Zheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Weidong Gao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Madiha Saeed
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| | - Weizhong Sheng
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Haijun Yu
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| |
Collapse
|
26
|
Qin Y, Zhang H, Li Y, Xie T, Yan S, Wang J, Qu J, Ouyang F, Lv S, Guo Z, Wei H, Yu CY. Promotion of ICD via Nanotechnology. Macromol Biosci 2023; 23:e2300093. [PMID: 37114599 DOI: 10.1002/mabi.202300093] [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: 03/07/2023] [Revised: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Immunotherapy represents the most promising treatment strategy for cancer, but suffers from compromised therapeutic efficiency due to low immune activity of tumor cells and an immunosuppressive microenvironment, which significantly hampers the clinical translations of this treatment strategy. To promote immunotherapy with desired therapeutic efficiency, immunogenic cell death (ICD), a particular type of death capable of reshaping body's antitumor immune activity, has drawn considerable attention due to the potential to stimulate a potent immune response. Still, the potential of ICD effect remains unsatisfactory because of the intricate tumor microenvironment and multiple drawbacks of the used inducing agents. ICD has been thoroughly reviewed so far with a general classification of ICD as a kind of immunotherapy strategy and repeated discussion of the related mechanism. However, there are no published reviews, to the authors' knowledge, providing a systematic summarization on the enhancement of ICD via nanotechnology. For this purpose, this review first discusses the four stages of ICD according to the development mechanisms, followed by a comprehensive description on the use of nanotechnology to enhance ICD in the corresponding four stages. The challenges of ICD inducers and possible solutions are finally summarized for future ICD-based enhanced immunotherapy.
Collapse
Affiliation(s)
- Yang Qin
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haitao Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yunxian Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ting Xie
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shuang Yan
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiaqi Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jun Qu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Feijun Ouyang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shaoyang Lv
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zifen Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
27
|
Bushra R, Ahmad M, Seidi F, Qurtulen, Song J, Jin Y, Xiao H. Polysaccharide-based nanoassemblies: From synthesis methodologies and industrial applications to future prospects. Adv Colloid Interface Sci 2023; 318:102953. [PMID: 37399637 DOI: 10.1016/j.cis.2023.102953] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/23/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023]
Abstract
Polysaccharides, due to their remarkable features, have gained significant prominence in the sustainable production of nanoparticles (NPs). High market demand and minimal production cost, compared to the chemically synthesised NPs, demonstrate a drive towards polysaccharide-based nanoparticles (PSNPs) benign to environment. Various approaches are used for the synthesis of PSNPs including cross-linking, polyelectrolyte complexation, and self-assembly. PSNPs have the potential to replace a wide diversity of chemical-based agents within the food, health, medical and pharmacy sectors. Nevertheless, the considerable challenges associated with optimising the characteristics of PSNPs to meet specific targeting applications are of utmost importance. This review provides a detailed compilation of recent accomplishments in the synthesis of PSNPs, the fundamental principles and critical factors that govern their rational fabrication, as well as various characterisation techniques. Noteworthy, the multiple use of PSNPs in different disciplines such as biomedical, cosmetics agrochemicals, energy storage, water detoxification, and food-related realms, is accounted in detail. Insights into the toxicological impacts of the PSNPs and their possible risks to human health are addressed, and efforts made in terms of PSNPs development and optimising strategies that allow for enhanced delivery are highlighted. Finally, limitations, potential drawbacks, market diffusion, economic viability and future possibilities for PSNPs to achieve widespread commercial use are also discussed.
Collapse
Affiliation(s)
- Rani Bushra
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China
| | - Mehraj Ahmad
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; College of Light Industry and Food, Department of Food Science and Engineering, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China.
| | - Farzad Seidi
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China
| | - Qurtulen
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Junlong Song
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China
| | - Yongcan Jin
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, Jiangsu, China; Joint International Research Lab of Lignocellulosic Functional Materials and Provincial Key Lab of Pulp and Paper Sci & Tech, Nanjing Forestry University, Nanjing 210037, Jiangsu, China
| | - Huining Xiao
- Department of Chemical Engineering, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| |
Collapse
|
28
|
Dai Y, Li X, Xue Y, Chen K, Jiao G, Zhu L, Li M, Fan Q, Dai Y, Zhao Q, Shen Q. Self-delivery of metal-coordinated NIR-II nanoadjuvants for multimodal imaging-guided photothermal-chemodynamic amplified immunotherapy. Acta Biomater 2023; 166:496-511. [PMID: 37230439 DOI: 10.1016/j.actbio.2023.05.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/01/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
The effectiveness of phototheranostics induced immunotherapy is still hampered by limited light penetration depth, the complex immunosuppressive tumor microenvironment (TME) and the low efficiency of immunomodulator drug delivery. Herein, self-delivery and TME responsive NIR-II phototheranostic nanoadjuvants (NAs) were fabricated to suppress the growth and metastasis of melanoma through the integration of photothermal-chemodynamic therapy (PTT-CDT) and immune remodeling. The NAs were constructed by the self-assembly of ultrasmall NIR-II semiconducting polymer dots and the toll-like receptor agonist resiquimod (R848) utilizing manganese ions (Mn2+) as coordination nodes. Under acidic TME, the NAs responsively disintegrated and released therapeutic components, which enable NIR-II fluorescence/photoacoustic/magnetic resonance imaging-guided tumor PTT-CDT. Moreover, the synergistic treatment of PTT-CDT could induce significant tumor immunogenic cell death and evoke highly efficacious cancer immunosurveillance. The released R848 stimulated the maturation of dendritic cells, which both amplified the antitumor immune response by modulating and remodeling the TME. The NAs present a promising integration strategy of polymer dot-metal ion coordination and immune adjuvants for precise diagnosis and amplified anti-tumor immunotherapy against deep-seated tumors. STATEMENT OF SIGNIFICANCE: The efficiency of phototheranostics induced immunotherapy is still limited by insufficient light penetration depth, low immune response and the complex immunosuppressive tumor microenvironment (TME). In order to improve the efficacy of immunotherapy, self-delivery NIR-II phototheranostic nanoadjuvants (PMR NAs) were successfully fabricated via the facile coordination self-assembly of ultra-small NIR-II semiconducting polymer dots and toll-like receptor agonist resiquimod (R848) utilizing manganese ions (Mn2+) as coordination nodes. PMR NAs not only enable TME responsive cargo release and NIR-II fluorescence/photoacoustic/magnetic resonance imaging mediated precise localization of tumors, but also achieve synergistic photothermal-chemodynamic therapy, evoking an effective anti-tumor immune response by ICD effect. The responsively released R848 could further amplify the efficiency of immunotherapy by reversing and remodeling the immunosuppressive tumor microenvironment, thereby effectively inhibiting tumor growth and lung metastasis.
Collapse
Affiliation(s)
- Yeneng Dai
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China; Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Xiangyu Li
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Yuwen Xue
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Kai Chen
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Guanda Jiao
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Lipeng Zhu
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China; School of Life Sciences, Central South University, Changsha 410013, China
| | - Meixing Li
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Quli Fan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Yunlu Dai
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Qi Zhao
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China.
| | - Qingming Shen
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| |
Collapse
|
29
|
Zare I, Taheri-Ledari R, Esmailzadeh F, Salehi MM, Mohammadi A, Maleki A, Mostafavi E. DNA hydrogels and nanogels for diagnostics, therapeutics, and theragnostics of various cancers. NANOSCALE 2023. [PMID: 37337663 DOI: 10.1039/d3nr00425b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
As an efficient class of hydrogel-based therapeutic drug delivery systems, deoxyribonucleic acid (DNA) hydrogels (particularly DNA nanogels) have attracted massive attention in the last five years. The main contributor to this is the programmability of these 3-dimensional (3D) scaffolds that creates fundamental effects, especially in treating cancer diseases. Like other active biological ingredients (ABIs), DNA hydrogels can be functionalized with other active agents that play a role in targeting drug delivery and modifying the half-life of the therapeutic cargoes in the body's internal environment. Considering the brilliant advantages of DNA hydrogels, in this survey, we intend to submit an informative collection of feasible methods for the design and preparation of DNA hydrogels and nanogels, and the responsivity of the immune system to these therapeutic cargoes. Moreover, the interactions of DNA hydrogels with cancer biomarkers are discussed in this account. Theragnostic DNA nanogels as an advanced species for both detection and therapeutic purposes are also briefly reviewed.
Collapse
Affiliation(s)
- Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz 7178795844, Iran
| | - Reza Taheri-Ledari
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Farhad Esmailzadeh
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Mohammad Mehdi Salehi
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Adibeh Mohammadi
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Ali Maleki
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
30
|
Liu T, Zhu M, Chang X, Tang X, Yuan P, Tian R, Zhu Z, Zhang Y, Chen X. Tumor-Specific Photothermal-Therapy-Assisted Immunomodulation via Multiresponsive Adjuvant Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300086. [PMID: 36782382 DOI: 10.1002/adma.202300086] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/05/2023] [Indexed: 05/05/2023]
Abstract
Multiresponsive adjuvant nanoparticles (RMmAGL) are fabricated to perform tumor-specific photothermal therapy while regulating the behavior of tumor-associated immune cells for primary tumor eradication and metastasis inhibition. Core-satellite-like RMmAGL have a core of mannose-functionalized mesoporous silica nanoparticles loaded with the TLR7 agonist imiquimod (R837@MSN-mannose) connected via hydrazone bonds to satellites of glutamine (Glu)- and lysine (Lys)-comodified gold nanoparticles (AuNPs-Glu/Lys). During therapy, the acidic environment in tumor tissue cleaves the hydrazone bonds to release AuNPs-Glu/Lys, which further accumulate in tumor cells. After internalization, photothermal agents (aggregated AuNPs-Glu/Lys) are generated in situ through the intratumoral enzyme-catalyzed reaction between Glu and Lys, resulting in tumor-specific photothermal therapy. The detachment of AuNPs-Glu/Lys also triggers the release of R837, which matured dendritic cells (DCs) via a vaccine-like mechanism along with the tumor-associated antigens generated by photothermal therapy. These matured DCs further activates surrounding T cells for immunotherapy. Moreover, the resulting free MSN-mannose serves as an artificial glycocalyx to continuously induce the polarization of tumor-associated macrophages from an immunosuppressive phenotype to an inflammatory phenotype, thus further enhancing immunotherapy. Both in vivo and in vitro experiments demonstrate significant inhibition of malignant tumors after therapy.
Collapse
Affiliation(s)
- Tao Liu
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xiaowei Chang
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xiaoyu Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Pingyun Yuan
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Ran Tian
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
31
|
Sun Y, Lyu B, Yang C, He B, Zhang H, Wang X, Zhang Q, Dai W. An enzyme-responsive and transformable PD-L1 blocking peptide-photosensitizer conjugate enables efficient photothermal immunotherapy for breast cancer. Bioact Mater 2023; 22:47-59. [PMID: 36203955 PMCID: PMC9519467 DOI: 10.1016/j.bioactmat.2022.08.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/02/2022] [Accepted: 08/15/2022] [Indexed: 12/07/2022] Open
Abstract
Mild photothermal therapy combined with immune checkpoint blockade has received increasing attention for the treatment of advanced or metastatic cancers due to its good therapeutic efficacy. However, it remains a challenge to facilely integrate the two therapies and make it potential for clinical translation. This work designed a peptide-photosensitizer conjugate (PPC), which consisted of a PD-L1 antagonist peptide (CVRARTR), an MMP-2 specific cleavable sequence, a self-assembling motif, and the photosensitizer Purpurin 18. The single-component PPC can self-assemble into nanospheres which is suitable for intravenous injection. The PPC nanosphere is cleaved by MMP-2 when it accumulates in tumor sites, thereby initiating the cancer-specific release of the antagonist peptide. Simultaneously, the nanospheres gradually transform into co-assembled nanofibers, which promotes the retention of the remaining parts within the tumor. In vivo studies demonstrated that PPC nanospheres under laser irradiation promote the infiltration of cytotoxic T lymphocytes and maturation of DCs, which sensitize 4T1 tumor cells to immune checkpoint blockade therapy. Therefore, PPC nanospheres inhibit tumor growth efficiently both in situ and distally and blocked the formation of lung metastases. The present study provides a simple and efficient integrated strategy for breast cancer photoimmunotherapy.
A peptide-photosensitizer conjugate (PPC) with self-assembled ability. Self-assembled PPC realized enzyme-responsive PD-L1 blocking peptide release. Shape transformation from nanospheres to co-assembled nanofibers. Efficient integrated strategy for breast cancer photoimmunotherapy.
Collapse
Affiliation(s)
- Yanan Sun
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, 050017, China
| | - Bochen Lyu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Chang Yang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Corresponding author.
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Corresponding author.
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Corresponding author.
| |
Collapse
|
32
|
Li C, Guan N, Liu F. T7 peptide-decorated exosome-based nanocarrier system for delivery of Galectin-9 siRNA to stimulate macrophage repolarization in glioblastoma. J Neurooncol 2023; 162:93-108. [PMID: 36854924 DOI: 10.1007/s11060-023-04257-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/30/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE Exosomes are nano-vesicular carriers capable of delivering cargoes for intercellular communication, which holds potential as biocompatible and high efficiency systems for drug delivery. In this study, we evaluated the potential effect of T7 peptide-decorated exosome-loaded Galectin-9 siRNA (T7-Exo/siGalectin-9) in the M1 polarization of macrophages and immunosuppression of glioblastoma (GBM). METHODS Differentially expressed genes in GBM were in silico predicted and then experimentally verified. Galectin-9 was knocked down by siRNA to assess its role in tumor-bearing mice. T7 peptide-decorated exosomes (derived from human embryonic kidney [HEK]-293T cells) targeting GBM were prepared, and loaded with Galectin-9 siRNA by electroporation to prepare nanoformulations (T7-Exo/siGalectin-9). The role of T7-Exo/siGalectin-9 in CD8+ T cell cytotoxicity to target GBM cells and polarization of macrophages was evaluated after artificial modulation of Galectin-9 expression. Anti-tumor effects of T7-Exo/siGalectin-9 were elucidated in vitro and in vivo. RESULTS Galectin-9 was highly expressed in GBM tissues and cell lines. The siRNA-mediated knockdown of Galectin-9 repressed the growth of xenografts of GBM cells in C57BL/6 mice and activated immune response in the tumor microenvironment. T7-Exo/siGalectin-9 effectively delivered siGalectin-9 to GBM cells. T7-Exo/siGalectin-9 contributed to activation of the TLR7-IRF5 pathway, which polarized macrophages to M1 phenotype. By this mechanism, phagocytosis of GBM cells by macrophages was increased, the anti-tumor effect of CD8+ T cells was enhanced and the inflammatory responses were suppressed. CONCLUSION Overall, T7-Exo/siGalectin-9 promotes macrophage repolarization and restricts the immunosuppression of GBM, thus providing novel insights into and drug delivery system of immunotherapy for GBM.
Collapse
Affiliation(s)
- Chenguang Li
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Ning Guan
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121000, Liaoning, People's Republic of China.
| |
Collapse
|
33
|
Horvath D, Basler M. PLGA Particles in Immunotherapy. Pharmaceutics 2023; 15:pharmaceutics15020615. [PMID: 36839937 PMCID: PMC9965784 DOI: 10.3390/pharmaceutics15020615] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Poly(lactic-co-glycolic acid) (PLGA) particles are a widely used and extensively studied drug delivery system. The favorable properties of PLGA such as good bioavailability, controlled release, and an excellent safety profile due to the biodegradable polymer backbone qualified PLGA particles for approval by the authorities for the application as a drug delivery platform in humas. In recent years, immunotherapy has been established as a potent treatment option for a variety of diseases. However, immunomodulating drugs rely on targeted delivery to specific immune cell subsets and are often rapidly eliminated from the system. Loading of PLGA particles with drugs for immunotherapy can protect the therapeutic compounds from premature degradation, direct the drug delivery to specific tissues or cells, and ensure sustained and controlled drug release. These properties present PLGA particles as an ideal platform for immunotherapy. Here, we review recent advances of particulate PLGA delivery systems in the application for immunotherapy in the fields of allergy, autoimmunity, infectious diseases, and cancer.
Collapse
Affiliation(s)
- Dennis Horvath
- Division of Immunology, Department of Biology, University of Konstanz, D-78457 Konstanz, Germany
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, D-78457 Konstanz, Germany
| | - Michael Basler
- Division of Immunology, Department of Biology, University of Konstanz, D-78457 Konstanz, Germany
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland
- Correspondence:
| |
Collapse
|
34
|
Zhang Y, Chen J, Shi L, Ma F. Polymeric nanoparticle-based nanovaccines for cancer immunotherapy. MATERIALS HORIZONS 2023; 10:361-392. [PMID: 36541078 DOI: 10.1039/d2mh01358d] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Therapeutic cancer vaccines, which are designed to amplify tumor-specific T cell responses, have been envisioned as one of the most powerful tools for effective cancer immunotherapy. However, increasing the potency, quality and durability of the vaccine response remains a big challenge. In recent years, materials-based delivery systems focusing on the co-delivery of antigens and adjuvants to enhance cancer vaccination therapy have attracted increasing interest. Among various materials, polymeric nanoparticles (NPs) with different physicochemical properties which can incorporate multiple immunological cues are of great interest. In this review, the recent progress in the design and construction of both ex vivo subunit and in situ cancer vaccines using polymeric NPs is summarized. Especially, we will focus on how these NPs improve the adjuvanticity of vaccines. The design principles of polymeric NPs for ex vivo subunit cancer vaccines and in situ cancer vaccination are also discussed. Finally, we want to briefly discuss molecular chaperones in cancer immunity and the applications of our unique self-assembly mixed shell polymeric micelle-based nanochaperones for cancer vaccines.
Collapse
Affiliation(s)
- Yongxin Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
| | - Jiajing Chen
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300192, P. R. China
| | - Feihe Ma
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China.
| |
Collapse
|
35
|
Abstract
Immunotherapy has revolutionized the treatment of patients with cancer. However, promoting antitumour immunity in patients with tumours that are resistant to these therapies remains a challenge. Thermal therapies provide a promising immune-adjuvant strategy for use with immunotherapy, mostly owing to the capacity to reprogramme the tumour microenvironment through induction of immunogenic cell death, which also promotes the recruitment of endogenous immune cells. Thus, thermal immunotherapeutic strategies for various cancers are an area of considerable research interest. In this Review, we describe the role of the various thermal therapies and provide an update on attempts to combine these with immunotherapies in clinical trials. We also provide an overview of the preclinical development of various thermal immuno-nanomedicines, which are capable of combining thermal therapies with various immunotherapy strategies in a single therapeutic platform. Finally, we discuss the challenges associated with the clinical translation of thermal immuno-nanomedicines and emphasize the importance of multidisciplinary and inter-professional collaboration to facilitate the optimal translation of this technology from bench to bedside.
Collapse
|
36
|
Interventional Oncolytic Immunotherapy with LTX-315 for Residual Tumor after Incomplete Radiofrequency Ablation of Liver Cancer. Cancers (Basel) 2022; 14:cancers14246093. [PMID: 36551579 PMCID: PMC9777024 DOI: 10.3390/cancers14246093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Objective: To investigate the feasibility of interventional oncolytic immunotherapy with LTX-315 for residual tumors after incomplete radiofrequency ablation (iRFA) of VX2 liver tumors in a rabbit model. Methods: For in vitro experiments, VX2 tumor cells were treated with: (1) phosphate buffered saline, (2) radiofrequency hyperthermia (RFH), (3) LTX-315, and (4) RFH plus LTX-315. The residual tumors after iRFA of VX2 liver tumors were treated with: (1) phosphate buffered saline served as control, (2) 2 mg LTX-315, and (3) 4 mg LTX-315. MTS assay, fluorescence microscopy, and flow cytometry were used to compare cell viabilities and apoptosis among different groups. Ultrasound imaging was used to follow up the tumor growth, which were correlated with the optical imaging and subsequent histology. Results: For in vitro experiments, compared with the other three groups, MTS assay demonstrated the lowest cell viability, fluorescence microscopy showed the least survival cells, and apoptosis analysis revealed the highest percentage of apoptosis cells in the combination treatment groups (p < 0.001). For in vivo experiments, ultrasound imaging showed the smallest tumor volume in the group with 4 mg LTX-315 therapy compared with the other two groups (p < 0.001). The optical imaging and histopathological analysis showed complete necrosis of the tumors in the group with 4 mg LTX-315 therapy. A significant increase of CD8+ T cells and HSP70 and a significant decrease of Tregs were observed in residual tumors in the group with 2 mg LTX-315 therapy compared with the control group (p < 0.001). Conclusion: Interventional oncolytic immunotherapy with LTX-315 for residual tumors after iRFA of liver cancer is feasible, which may open up new avenues to prevent residual tumors after RFA of intermediate-to-large liver cancers.
Collapse
|
37
|
Cytokine Therapy Combined with Nanomaterials Participates in Cancer Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14122606. [PMID: 36559100 PMCID: PMC9788370 DOI: 10.3390/pharmaceutics14122606] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/14/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Immunotherapy has gradually become an emerging treatment modality for tumors after surgery, radiotherapy, and chemotherapy. Cytokine therapy is a promising treatment for cancer immunotherapy. Currently, there are many preclinical theoretical bases to support this treatment strategy and a variety of cytokines in clinical trials. When cytokines were applied to tumor immunotherapy, it was found that the efficacy was not satisfactory. As research on tumor immunity has deepened, the role of cytokines in the tumor microenvironment has been further explored. Meanwhile, the study of nanomaterials in drug delivery has been fully developed in the past 20 years. Researchers have begun to think about the possibility of combining cytokine therapy with nanomaterials. Herein, we briefly review various nano-delivery systems that can directly deliver cytokines or regulate the expression of cytokines in tumor cells for cancer immunotherapy. We further discussed the feasibility of the combination of various therapies. We looked forward to the main challenges, opportunities, and prospects of tumor immunotherapy with multiple cytokines and a nano-delivery system.
Collapse
|
38
|
Kan X, Zhou G, Zhang F, Ji H, Shin DS, Monsky W, Zheng C, Yang X. Enhanced efficacy of direct immunochemotherapy for hepatic cancer with image-guided intratumoral radiofrequency hyperthermia. J Immunother Cancer 2022; 10:e005619. [PMID: 36450380 PMCID: PMC9717415 DOI: 10.1136/jitc-2022-005619] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND It is still a challenge to prevent tumor recurrence post radiofrequency ablation (RFA) of medium-to-large hepatocellular carcinomas (HCC). Immunochemotherapy, a combination of immunotherapy with chemotherapy, has demonstrated a great potential in augmenting the treatment efficacy for some malignancies. In this study, we validated the feasibility of using radiofrequency hyperthermia (RFH)-enhanced intratumoral immunochemotherapy of LTX-315 with liposomal doxorubicin for rat orthotopic HCC. METHODS Different groups of luciferase-labeled rat HCC cells and rat orthotopic HCC models were treated by: (1) phosphate buffered saline; (2) RFH; (3) LTX-315; (4) RFH+LTX-315; (5) liposomal doxorubicin; (6) RFH+liposomal doxorubicin; (7) LTX-315+liposomal doxorubicin; and (8) RFH+LTX-315+liposomal doxorubicin. Cell viabilities and apoptosis of different treatment groups were compared. Changes in tumor sizes were quantified by optical and ultrasound imaging, which were confirmed by subsequent histopathology. The potential underlying biological mechanisms of the triple combination treatment (RFH+LTX-315+liposomal doxorubicin) were explored. RESULTS Flow cytometry and MTS assay showed the highest percentage of apoptotic cells and lowest cell viability in the triple combination treatment group compared with other seven groups (p<0.001). Tumors in this group also presented the most profound decrease in bioluminescence signal intensities and the smallest tumor volumes compared with other seven groups (p<0.001). A significant increase of CD8+ T cells, CD8+/interferon (IFN)-γ+ T cells, CD8+/tumor necrosis factor (TNF)-α+ T cells, and natural killer cells, and a significant decrease of regulatory T cells were observed in the tumors (p<0.001). Meanwhile, a significantly higher level of Th1-type cytokines in both plasma (interleukin (IL)-2, IL-12, IL-18, IFN-γ) and tumors (IL-2, IL-18, IFN-γ, TNF-α), as well as a significantly lower Th2-type cytokines of IL-4 and IL-10 in plasma and tumor were detected. CONCLUSIONS Intratumoral RFA-associated RFH could enhance the efficacy of immunochemotherapy of LTX-315 with liposomal doxorubicin for HCC, which may provide a new strategy to increase the curative efficacy of thermal ablation for medium-to-large HCC.
Collapse
Affiliation(s)
- Xuefeng Kan
- Image-Guided Bio-Molecular Intervention Research and Section of Vascular and Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanhui Zhou
- Image-Guided Bio-Molecular Intervention Research and Section of Vascular and Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA
- Hepatobiliary and Pancreatic Interventional Treatment Center, Division of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Feng Zhang
- Image-Guided Bio-Molecular Intervention Research and Section of Vascular and Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Hongxiu Ji
- Image-Guided Bio-Molecular Intervention Research and Section of Vascular and Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Pathology, Overlake Medical Center and Incyte Diagnostics, Bellevue, WA, USA
| | - David S Shin
- Image-Guided Bio-Molecular Intervention Research and Section of Vascular and Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Wayne Monsky
- Image-Guided Bio-Molecular Intervention Research and Section of Vascular and Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoming Yang
- Image-Guided Bio-Molecular Intervention Research and Section of Vascular and Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
39
|
Meng Z, Fang X, Fu B, Qian C, Yang Z, Bai Y, Tao X, Huang H, Ma C, Miao W, Ren H, Wang A, Li X. Tumor immunotherapy boosted by R837 nanocrystals through combining chemotherapy and mild hyperthermia. J Control Release 2022; 350:841-856. [PMID: 36096366 DOI: 10.1016/j.jconrel.2022.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 12/07/2022]
Abstract
Melanoma is a malignant skin cancer that is prone to metastasis in the early stage and has a poor prognosis. Immunomodulatory therapy for melanoma has been a hot research topic in recent years. However, low immune cell infiltration and loss of tumor immunogenicity may occur in tumors, resulting in low response rates to immunotherapy. Thus, immunomodulatory therapy is usually used in combination with chemotherapy and radiotherapy. Development of combined therapeutic strategies with low systemic toxicity, high immune responsiveness and long-term inhibition of metastasis and recurrence of melanoma is the goal of current research. In this study, the insoluble immune adjuvant imiquimod (R837) was prepared as nanocrystals and coated with polydopamine (PDA) to form R837@PDA, which was then loaded into chitosan hydrogel (CGP) to form the drug-loaded gel system, R837@PDA@CGP (RPC), to combine immunomodulation effects, induction of immunogenic cell death (ICD) effects and immune-enhancement effects. After treatment with RPC, ICD in melanoma was induced, and the infiltration rate of cytotoxic T cells (CTLs) in melanoma was also significantly enhanced, which turned the tumor itself into an in situ vaccine and boosted the cancer-immunity cycle at the tumor site. Therefore, melanoma growth, metastasis and recurrence were notably inhibited.
Collapse
Affiliation(s)
- Zhengjie Meng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Xue Fang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Bowen Fu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Cheng Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zheng Yang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Yunhao Bai
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Xinyue Tao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Haixiao Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Chenyu Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Wenjun Miao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Hao Ren
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xueming Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
40
|
Xu N, Zhang X, Qi T, Wu Y, Xie X, Chen F, Shao D, Liao J. Biomedical applications and prospects of temperature‐orchestrated photothermal therapy. MEDCOMM – BIOMATERIALS AND APPLICATIONS 2022; 1. [DOI: 10.1002/mba2.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/10/2022] [Indexed: 01/06/2025]
Abstract
AbstractPhotothermal therapy (PTT) has been regarded as a promising strategy considering its advantages of high inherent specificity and a lower invasive burden. Since the photothermal killing of cells/bacteria showed different patterns of death depending on the varying temperature in PTT, the temperature change of PTT is vital to cell/tissue response in scientific research and clinical application. On one hand, mild PTT has received substantial attention in the treatment of cancer and soft/hard tissue repair. On the other hand, the high temperature induced by PTT is capable of antibacterial capacity, which is better than conventional antibiotic therapy with drug resistance. Herein, we summarize the recent developments in the application of temperature‐dependent photothermal biomaterials, mainly covering the temperature ranges of 40–42°C, 43–50°C, and over 50°C. We highlight the biological mechanism of PTT and the latest progress in the treatment of different diseases. Finally, we conclude by discussing the challenges and perspectives of biomaterials in addressing temperature‐orchestrated PTT. Given a deep understanding of the interaction between temperature and biology, rationally designed biomaterials with sophisticated photothermal responsiveness will benefit the outcomes of personalized PTT toward various diseases.
Collapse
Affiliation(s)
- Nuo Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Xu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Tingting Qi
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine University of Electronic Science and Technology of China Chengdu China
| | - Yongzhi Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Xi Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Fangman Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences University of Macau Macau China
| | - Dan Shao
- School of Medicine South China University of Technology Guangzhou Guangdong China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China
| |
Collapse
|
41
|
Siboro PY, Nguyen VKT, Miao YB, Sharma AK, Mi FL, Chen HL, Chen KH, Yu YT, Chang Y, Sung HW. Ultrasound-Activated, Tumor-Specific In Situ Synthesis of a Chemotherapeutic Agent Using ZIF-8 Nanoreactors for Precision Cancer Therapy. ACS NANO 2022; 16:12403-12414. [PMID: 35920682 DOI: 10.1021/acsnano.2c03587] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The in situ transformation of low-toxicity precursors into a chemotherapeutic agent at a tumor site to enhance the efficacy of its treatment has long been an elusive goal. In this work, a zinc-based zeolitic imidazolate framework that incorporates pharmaceutically acceptable precursors is prepared as a nanoreactor (NR) system for the localized synthesis of an antitumor drug. The as-prepared NRs are administered intratumorally in a tumor-bearing mouse model and then irradiated with ultrasound (US) to activate the chemical synthesis. The US promotes the penetration of the administered NRs into the tumor tissue to cover the lesion entirely, although some NRs leak into the surrounding normal tissue. Nevertheless, only the tumor tissue, where the H2O2 concentration is high, is adequately exposed to the as-synthesized antitumor drug, which markedly impedes development of the tumor. No significant chemical synthesis is detected in the surrounding normal tissue, where the local H2O2 concentration is negligible and the US irradiation is not directly applied. The as-proposed tumor-specific in situ synthesis of therapeutic molecules induces hardly any significant in vivo toxicity and, thus, is potentially a potent biocompatible approach to precision chemotherapy.
Collapse
Affiliation(s)
- Putry Yosefa Siboro
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan (ROC)
| | - Van Khanh Thi Nguyen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan (ROC)
| | - Yang-Bao Miao
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan (ROC)
| | - Amit Kumar Sharma
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan (ROC)
| | - Fwu-Long Mi
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan (ROC)
| | - Hsin-Lung Chen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan (ROC)
| | - Kuan-Hung Chen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan (ROC)
| | - Yu-Tzu Yu
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan (ROC)
| | - Yen Chang
- Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and School of Medicine, Tzu Chi University, Hualien 97004, Taiwan (ROC)
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan (ROC)
| |
Collapse
|
42
|
Functionalized chitosan as a promising platform for cancer immunotherapy: A review. Carbohydr Polym 2022; 290:119452. [DOI: 10.1016/j.carbpol.2022.119452] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/20/2022]
|
43
|
Rostamizadeh L, Molavi O, Rashid M, Ramazani F, Baradaran B, Lavasanaifar A, Lai R. Recent advances in cancer immunotherapy: Modulation of tumor microenvironment by Toll-like receptor ligands. BIOIMPACTS : BI 2022; 12:261-290. [PMID: 35677663 PMCID: PMC9124882 DOI: 10.34172/bi.2022.23896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/18/2022]
Abstract
![]()
Immunotherapy is considered a promising approach for cancer treatment. An important strategy for cancer immunotherapy is the use of cancer vaccines, which have been widely used for cancer treatment. Despite the great potential of cancer vaccines for cancer treatment, their therapeutic effects in clinical settings have been limited. The main reason behind the lack of significant therapeutic outcomes for cancer vaccines is believed to be the immunosuppressive tumor microenvironment (TME). The TME counteracts the therapeutic effects of immunotherapy and provides a favorable environment for tumor growth and progression. Therefore, overcoming the immunosuppressive TME can potentially augment the therapeutic effects of cancer immunotherapy in general and therapeutic cancer vaccines in particular. Among the strategies developed for overcoming immunosuppression in TME, the use of toll-like receptor (TLR) agonists has been suggested as a promising approach to reverse immunosuppression. In this paper, we will review the application of the four most widely studied TLR agonists including agonists of TLR3, 4, 7, and 9 in cancer immunotherapy.
Collapse
Affiliation(s)
- Leila Rostamizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ramazani
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Lavasanaifar
- Faculty of Pharmacy and Pharmaceutical Science, University of Alberta, Edmonton, Canada
| | - Raymond Lai
- Department of Laboratory Medicine & Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
44
|
Huang L, Ge X, Liu Y, Li H, Zhang Z. The Role of Toll-like Receptor Agonists and Their Nanomedicines for Tumor Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14061228. [PMID: 35745800 PMCID: PMC9230510 DOI: 10.3390/pharmaceutics14061228] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/20/2022] [Accepted: 06/07/2022] [Indexed: 01/11/2023] Open
Abstract
Toll-like receptors (TLRs) are a class of pattern recognition receptors that play a critical role in innate and adaptive immunity. Toll-like receptor agonists (TLRa) as vaccine adjuvant candidates have become one of the recent research hotspots in the cancer immunomodulatory field. Nevertheless, numerous current systemic deliveries of TLRa are inappropriate for clinical adoption due to their low efficiency and systemic adverse reactions. TLRa-loaded nanoparticles are capable of ameliorating the risk of immune-related toxicity and of strengthening tumor suppression and eradication. Herein, we first briefly depict the patterns of TLRa, followed by the mechanism of agonists at those targets. Second, we summarize the emerging applications of TLRa-loaded nanomedicines as state-of-the-art strategies to advance cancer immunotherapy. Additionally, we outline perspectives related to the development of nanomedicine-based TLRa combined with other therapeutic modalities for malignancies immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Hui Li
- Correspondence: (H.L.); (Z.Z.)
| | | |
Collapse
|
45
|
Tan L, Shen X, He Z, Lu Y. The Role of Photodynamic Therapy in Triggering Cell Death and Facilitating Antitumor Immunology. Front Oncol 2022; 12:863107. [PMID: 35692783 PMCID: PMC9184441 DOI: 10.3389/fonc.2022.863107] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/22/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer is a major threat to human health because of its high mortality, easy recurrence, strong invasion, and metastasis. Photodynamic therapy (PDT) is a promising minimally invasive treatment for tumor. Compared with traditional treatment methods, PDT is less invasive and does not easily damage normal tissues. Most of the effects of this treatment are due to the direct effects of singlet oxygen together with reactive oxygen species. PDT can provide the source of active oxygen for the Fenton reaction, which enhances ferroptosis and also improves the efficacy of PDT in antitumor therapy. Additionally, in contrast to chemotherapy and radiotherapy, PDT has the effect of stimulating the immune response, which can effectively induce immunogenic cell death (ICD) and stimulate immunity. PDT is an ideal minimally invasive treatment method for tumors. In this paper, according to the characteristics of anti-tumor immunity of PDT, some tumor treatment strategies of PDT combined with anti-tumor immunotherapy are reviewed.
Collapse
|
46
|
Shim MK, Song SK, Jeon SI, Hwang KY, Kim K. Nano-sized drug delivery systems to potentiate the immune checkpoint blockade therapy. Expert Opin Drug Deliv 2022; 19:641-652. [DOI: 10.1080/17425247.2022.2081683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Man Kyu Shim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Su Kyung Song
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Biosystems & Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Seong Ik Jeon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Kwang Yeon Hwang
- Department of Biosystems & Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
47
|
Maravajjala KS, Swetha KL, Roy A. pH-responsive nanoparticles for multidimensional combined chemo-immunotherapy of cancer. J Pharm Sci 2022; 111:2353-2368. [DOI: 10.1016/j.xphs.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 10/18/2022]
|
48
|
Zhou S, Wang H, Li R, Wang Y, Wang Z, Feng L. Multifunctional Self-Assembly with NIR Light-Activated Cascade Effect for Improving Local Treatment on Solid Tumors. ACS APPLIED MATERIALS & INTERFACES 2022; 14:14087-14096. [PMID: 35297244 DOI: 10.1021/acsami.2c00448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Incomplete local treatment of solid tumors is the main cause of tumor difficult to cure, and easily leads to tumor metastasis and recurrence. The dense external matrix and hypoxic microenvironment of solid tumors severely restrict the therapy efficacy of local tumors. Enhancing the infiltration ability of agents to tumor tissues and adapting the therapy mode favored to hypoxic microenvironments are beneficial to improve the cure rate of tumors. In this work, we designed and developed a self-assembled biomaterial with a cascade effect triggered by near-infrared light. The self-assembly was combined of biotin, phase change material (PNIPAM), photochemical agent (ATT-2), and alkyl radical generator (AIPH). In the assembly, biotin acted as a targeted group. ATT-2 was used to provide heat to synergistically induce the phase change and decompose alkyl radicals. The superficial and deep tumors were ablated by heat and alkyl radicals with white light irradiation of the assembly, respectively. The assay in vivo showed that the self-assembly could effectively eliminate local lesions of solid tumors. This work provides new insights for improving the cure rate of tumors, which not only develops biomaterials adapted to the tumor microenvironment, but also proposes new therapies for complete elimination of solid tumors.
Collapse
Affiliation(s)
- Sirong Zhou
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, P. R. China
| | - Haoping Wang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, P. R. China
| | - Ruipeng Li
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, P. R. China
| | - Yunxia Wang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, P. R. China
| | - Zhijun Wang
- Department of Chemistry, Changzhi University, Changzhi 046011, P. R. China
| | - Liheng Feng
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, P. R. China
| |
Collapse
|
49
|
Barchi JJ. Glycoconjugate Nanoparticle-Based Systems in Cancer Immunotherapy: Novel Designs and Recent Updates. Front Immunol 2022; 13:852147. [PMID: 35432351 PMCID: PMC9006936 DOI: 10.3389/fimmu.2022.852147] [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: 01/10/2022] [Accepted: 03/04/2022] [Indexed: 11/15/2022] Open
Abstract
For many years, cell-surface glycans (in particular, Tumor-Associated Carbohydrate Antigens, TACAs) have been the target of both passive and active anticancer immunotherapeutic design. Recent advances in immunotherapy as a treatment for a variety of malignancies has revolutionized anti-tumor treatment regimens. Checkpoint inhibitors, Chimeric Antigen Receptor T-cells, Oncolytic virus therapy, monoclonal antibodies and vaccines have been developed and many approvals have led to remarkable outcomes in a subset of patients. However, many of these therapies are very selective for specific patient populations and hence the search for improved therapeutics and refinement of techniques for delivery are ongoing and fervent research areas. Most of these agents are directed at protein/peptide epitopes, but glycans-based targets are gaining in popularity, and a handful of approved immunotherapies owe their activity to oligosaccharide targets. In addition, nanotechnology and nanoparticle-derived systems can help improve the delivery of these agents to specific organs and cell types based on tumor-selective approaches. This review will first outline some of the historical beginnings of this research area and subsequently concentrate on the last 5 years of work. Based on the progress in therapeutic design, predictions can be made as to what the future holds for increasing the percentage of positive patient outcomes for optimized systems.
Collapse
Affiliation(s)
- Joseph J. Barchi
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| |
Collapse
|
50
|
Lee JH, Song J, Kim IG, You G, Kim H, Ahn JH, Mok H. Exosome-mediated delivery of transforming growth factor-β receptor 1 kinase inhibitors and toll-like receptor 7/8 agonists for combination therapy of tumors. Acta Biomater 2022; 141:354-363. [PMID: 35007784 DOI: 10.1016/j.actbio.2022.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/27/2021] [Accepted: 01/04/2022] [Indexed: 02/07/2023]
Abstract
In this study, combination therapy with the transforming growth factor-β receptor I (TGFβRI) kinase inhibitor SD-208 and a toll-like receptor (TLR)-7/8 agonist resiquimod (R848) was examined along with serum-derived exosomes (EXOs) as versatile carriers. SD-208-encapsulated EXOs (SD-208/EXOs) and R848-encapsulated EXOs (R848/EXOs) were successfully prepared with a size of 87 ± 8 nm and 51 ± 4 nm, respectively, which were stable in aqueous solution at pH 7.4. SD-208/EXOs and R848/EXOs reduced the migration of cancer cells (B16F10 and PC-3) and triggered the release of proinflammatory cytokines from stimulated macrophages and dendritic cells, respectively. The fluorescent dye-labeled EXOs showed significantly improved penetration through the PC-3/fibroblast co-culture spheroids and enhanced accumulation in the B16F10 mouse tumor model compared with the free fluorescent dye. In addition, the combination therapy of R848/EXOs (R848 dose of 0.36 mg/kg) and SD-208/EXOs (SD-208 dose of 0.75 mg/kg) reduced tumor growth and improved survival rate at low doses in the B16F10 tumor xenograft model. Taken together, the combination therapy using the TGFβRI kinase inhibitor and TLR 7/8 agonist with EXOs may serve as a promising strategy to treat melanoma and prostate cancer. STATEMENT OF SIGNIFICANCE: Owing to the prevalence of several non-responding cancers that resist treatment, it is necessary to identify a novel combined treatment strategy with biomaterials to maximize therapeutic efficacy and minimize the undesirable side effects. In this study, we aimed to examine the use of the TGFβRI kinase inhibitor SD-208 and the TLR7/8 agonist resiquimod (R848) encapsulated within serum-derived EXOs for their synergistic antitumor effects. We first demonstrated that combined treatment with SD-208 and R848 can be a convincing strategy to circumvent tumor growth in vivo using serum-derived exosomes as promising carriers. Therefore, we believe this manuscript would be of great interest to the biomaterial communities especially who are studying immunotherapy.
Collapse
|