1
|
Sun L, Liu Y, Sun Q, Wang G, Du B, Liu B, Gao T, Zhao P, Yang Y, Rong R. Polysaccharides from traditional Chinese medicine and their nano-formulated delivery systems for cancer immunotherapy. Carbohydr Polym 2025; 357:123416. [PMID: 40158963 DOI: 10.1016/j.carbpol.2025.123416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
Cancer immunotherapy has evolved into a new generation strategy in the field of anti-tumor treatment. Polysaccharides derived from Traditional Chinese Medicine (TCM) are gaining recognition as powerful immunomodulators in cancer therapy, noted for their multi-target and multi-pathway actions. Owing to their beneficial properties such as water solubility, biocompatibility, and chemical structure modifiability, TCM polysaccharides can also serve as carriers for hydrophobic drugs in the development of innovative drug delivery systems, enhancing synergistic antitumor effects. In this article, we summarize the diverse mechanisms of immunoregulation by TCM polysaccharides in tumor therapy. The applications of these polysaccharides as both active ingredients and drug carriers within nanodelivery systems for cancer immunotherapy are also introduced. Additionally, extensive research on TCM polysaccharides in clinical settings has been collected. Furthermore, discussions are presented on the development prospects and challenges faced by these polysaccharides in the field of tumor immunotherapy. Our goal is to improve researchers' comprehension of TCM polysaccharides in cancer immunotherapy, providing promising strategies to optimize cancer treatment and benefit diverse patient populations.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yuting Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Qihui Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Guimei Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Bodong Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Tian Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; Collaborative Innovation Center for Antiviral Traditional Chinese Medicine in Shandong Province, Jinan 250355, PR China; Shandong Antiviral Engineering Research Center of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
2
|
Chen Y, Zhang M, Li Z, Zheng J, Zhang Y, Guo Q, Liu S, Chen Y, Wei W, Jiang X, Tang J. Multifunctional Nanoplatform Based on Gelatin Nanoparticles with Immunomodulatory Capabilities for Combined Immunotherapy and Chemotherapy of Melanoma. Biomacromolecules 2025; 26:2996-3010. [PMID: 40207885 DOI: 10.1021/acs.biomac.5c00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Tumor immunotherapy has shown considerable therapeutic potential, especially when combined with chemotherapy. In this study, we developed a multifunctional nanoplatform GNPs-DOX/R848 that combined immunotherapy and chemotherapy for the treatment of melanoma, in which gelatin nanoparticles (GNPs) were loaded with the immunomodulatory agent resiquimod (R848) and the chemotherapy drug doxorubicin (DOX). GNPs possessed inherent immunomodulatory properties; when combined with R848, they induced a more pronounced polarization of M1-like macrophages by activating the NF-κB signaling pathway, thereby reversing the immunosuppressive tumor microenvironment. Meanwhile, GNPs effectively delivered R848 and DOX to tumor cells, promoting stronger therapeutic effects of the drugs, which strongly induced the immunogenic cell death triggered by DOX, leading to the infiltration of T cells into the tumor tissue. The treatment of melanoma demonstrated that GNPs-DOX/R848 significantly reduced tumor volume, enhanced the therapeutic effects of chemotherapy, providing a new approach for the combined treatment of cancer with immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Ying Chen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P.R. China
| | - Miaomiao Zhang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Zongjia Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P.R. China
| | - Jinyao Zheng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P.R. China
| | - Yuanhao Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P.R. China
| | - Qianyu Guo
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P.R. China
| | - Suzhen Liu
- Department of Cardiology, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, PR China
| | - Yu Chen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P.R. China
| | - Wei Wei
- Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xiue Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P.R. China
| | - Jilin Tang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, P.R. China
| |
Collapse
|
3
|
Xu QH, Yin XY, Chen ZQ, Huang EK, Yao X, Li X, Liu PN. Construction of In Situ Personalized Cancer Vaccines by Bioorthogonal Catalytic Microneedles for Augmented Melanoma Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500015. [PMID: 40130650 DOI: 10.1002/smll.202500015] [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: 01/01/2025] [Revised: 03/06/2025] [Indexed: 03/26/2025]
Abstract
In situ personalized tumor vaccines are produced directly at the primary tumor site by killing cancer cells and stimulating immune cells, they are effective against individuals and bypass the complexity and high cost of in vitro vaccine production. However, their clinical application is hindered by insufficient efficiency in inducing immunogenic cancer cell death (ICD) and systemic inflammation caused by immune adjuvants. Here, personalized cancer vaccines are constructed in situ for melanoma immunotherapy based on bioorthogonal catalytic microneedles, which enable the catalytic release of prodrugs at tumor sites and mediate strong ICD and an enhanced tumor immune response while avoiding systemic immune storms and toxic side effects. By incorporating TiO2 nanosheets supported Pd into swellable microneedles, the bioorthogonal microneedles are constructed to catalyze the depropargylation reaction of doxorubicin (DOX) prodrug and imiquimod (IMQ) prodrug in situ. The activated DOX at subcutaneous tumor sites induced strong ICD and released tumor-associated antigens. Concurrently, the activated IMQ acts as a Toll-like receptor (TLR7) agonist, enhancing the anti-tumor immune response. In vivo experiments demonstrate that this immunotherapy achieves ≈97% inhibition of primary tumors and effectively inhibits untreated distant tumors (≈94% inhibition) and lung metastasis (≈92% inhibition).
Collapse
Affiliation(s)
- Qian-He Xu
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China
| | - Xiu-Yuan Yin
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China
| | - Zhen-Qiang Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China
| | - En-Kui Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China
| | - Xiaojun Yao
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, 999078, China
| | - Xingguang Li
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China
| | - Pei-Nian Liu
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
4
|
He S, Huang Y, Liu J, Liu H, Chen Y, Zou T, Sun J, Wang W, Wei H, Yu CY. A Metformin-Based Multifunctional Nanoplatform as a DNA Damage Amplifier for Maximized Radio-Immunotherapy to Overcome Radiotherapy Resistance. ACS NANO 2025; 19:14848-14864. [PMID: 40207668 DOI: 10.1021/acsnano.4c18627] [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: 04/11/2025]
Abstract
Radiotherapy (RT) has been highlighted to be an effective strategy for antitumor immunity activation by causing direct DNA damages, but it generally suffers from low response rates due to the compromised cytosolic DNA (cDNA) recognition by cyclic GMP-AMP synthase (cGAS). Simultaneous DNA repair and clearance system regulation for enhanced cDNA accumulation is a useful approach to improve immune response rates, which remains seldom reported to our knowledge. Here, we report the construction of a metformin (MET)-based multifunctional nanocomplex, CS-MET/siTREX1 (CSMT), consisting of biguanide-decorated CS (CS-MET) as the vector and 3'-5' DNA exonuclease TREX1 siRNA (siTREX1) as the therapeutic gene for RT-induced antitumor immunity enhancement by amplifying the initial DNA damage signals. The uniqueness of this study is the development of CSMT as a specific DNA damage amplifier to promote cDNA accumulation for maximizing radio-immunotherapy and circumventing RT resistance. Specifically, the CSMT nanocomplexes show not only enhanced gene transfection efficiency by MET modification but also synergistic therapeutic effects including MET's inhibition on DNA repair and siTREX1's attenuation on cDNA clearance, which leads to the greatest inhibitory effect in a Hepa1-6 proximal/distal tumor model with a high tumor growth inhibition (TGI) value of 99.1% for the primary tumor and significantly compromised distal tumor growth by inducing immunogenic cell death (ICD), promoting tumor-associated neutrophil (TAN) polarization, and stimulating tumor-specific memory T-cell generation. Overall, the CSMT nanocomplexes developed herein hold great translatable promises for overcoming RT resistance in clinics.
Collapse
Affiliation(s)
- Shuangyan He
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yun Huang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jia Liu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hongdu Liu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yalan Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ting Zou
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jian Sun
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - WuZhou Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hua Wei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Cui-Yun Yu
- Affiliated Hospital of Hunan Academy of Chinese Medicine Hunan Academy of Chinese Medicine, Changsha 410013, China
| |
Collapse
|
5
|
Wei W, Du Y, Kang X, Liu Y, Liu Y, Guo Q, He Q, Wang J, Zhu S, Zhou JY, Bu W. Nanoscale Rhodium(I) Based Metal-Organic Framework Demonstrating Intense NIR-II Luminescence for Bioimaging. NANO LETTERS 2025; 25:4613-4620. [PMID: 40052801 DOI: 10.1021/acs.nanolett.5c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Although luminescent metal-organic frameworks (MOFs) have been widely reported, rare examples were found to emit in the second near-infrared (NIR-II, 1000-1700 nm) window. In this work, two nanoscale rhodium(I)-based MOFs (Rh-1@SDS and Rh-1@DSPE-PEG) have been controllably constructed in the aqueous dispersions of sodium dodecyl sulfate (SDS) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-methoxy-poly(ethylene glycol) (DSPE-PEG), wherein micelle- and vesicle-like aggregates form, respectively, with high colloidal stability. The vesicular dispersion of Rh-1@DSPE-PEG exhibits intense NIR-II luminescence at 1125 (1245, shoulder) nm. Consequently, this nanoMOF was used as an NIR-II luminescence probe, indicative of high-resolution systemic and local vascular imaging, where the postoperative recovery process of flap transplantation was clearly visualized. Meanwhile, it also demonstrates superior tumor targeting in the NIR-II window. To the best of our knowledge, this research represents the first example of nanoMOFs having intense NIR-II luminescence and excellent imaging capabilities.
Collapse
Affiliation(s)
- Wenxuan Wei
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- School of Physical Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Yijing Du
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Xiaomei Kang
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Yilin Liu
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Yujia Liu
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Qian Guo
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Qun He
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jun Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jin Yuan Zhou
- School of Physical Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Weifeng Bu
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| |
Collapse
|
6
|
Jiang M, Liao L, Zhang J, Wei X, Yu CY, Wei H. Peptide core spherical nucleic acids circumvent tumor immunosuppression via supplementing methionine for enhanced photodynamic/gene immune/therapy of hepatocellular carcinoma. J Colloid Interface Sci 2025; 682:653-670. [PMID: 39642551 DOI: 10.1016/j.jcis.2024.11.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
Spherical nucleic acids (SNAs) with functional peptide cores are an emerging nanoplatform for synergistic cancer therapy but have been rarely reported. We construct herein the first SNA nanoplatform based on a biodegradable binary peptide backbone of methionine (Met) and cysteine (Cys) for codelivery of a photosensitizer, Chlorin e6 (Ce6) and human liver-specific miR122 for synergistic photodynamic-gene therapy of hepatic cell carcinoma (HCC). Met supplementation by the peptide core improves the infiltration of T cells and enhances the effector function of T cells for turning a "cold" tumor into a "hot" one. The resulting SNA(+) shows the most significant inhibitory effect in a Hepa1-6 HCC primary/distal tumor model, with tumor growth inhibition (TGI) values of 98.5 ± 0.5 % and 99.1 ± 0.4 % for the primary and distant tumors, respectively. This SNA nanoplatform achieves superior high TGI values reported thus far to our knowledge with almost complete eradication for both tumors due to the simultaneous adaptive and innate immunity activation via photodynamic therapy (PDT) induced immunogenic cell death (ICD) and Met supplementation-promoted adaptive immunity, and miR122-enhanced innate immunity. Overall, this study not only develops a reliable synthetic strategy toward peptide-backboned multifunctional SNA nanoplatform, but also reports the modulation of amino acid metabolism for enhanced innate immunity for highly efficient HCC immunotherapy.
Collapse
Affiliation(s)
- Mingchao Jiang
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Luanfeng Liao
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Jinyan Zhang
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Xiaojie Wei
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China
| | - Cui-Yun Yu
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China; Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha 410013, China.
| | - Hua Wei
- Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, MOE Key Lab of Rare Pediatric Disease, University of South China, Hengyang 421001, China.
| |
Collapse
|
7
|
Qu G, Liu K, Xu W, Li D. Integrated analysis and experimental validation reveal the prognostic and immunological features associated with coagulation in hepatocellular carcinoma. Sci Rep 2025; 15:8626. [PMID: 40074769 PMCID: PMC11904193 DOI: 10.1038/s41598-025-85491-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/03/2025] [Indexed: 03/14/2025] Open
Abstract
Coagulation is intensively related to various tumors, which affects their progression and prognosis. However, research on the impact of coagulation-associated genes (CAGs) on hepatocellular carcinoma (HCC) occurrence, prognosis, and immune microenvironment is limited. Consequently, our research aims to uncover how CAGs affect the prognosis and immune microenvironments of HCC. We integrated gene expression data and clinical information from three datasets (GSE14520, GSE76427, and TCGA-LIHC). 281 CAGs were obtained from the coagulation-related pathway (hsa04610). We obtained three CAG patterns through a consensus clustering algorithm. Afterward, differential analyses of prognosis, biological processes, immune infiltration, and functional and pathway enrichment were conducted on the three CAG patterns. We intersected CAGs with differentially expressed genes in GSE76427 and then conducted Cox regression analysis to obtain the prognostic genes in HCC. Glycerol-3-phosphate dehydrogenase 2 (GPD2) was selected for further analyses. TCGA-LIHC samples with different GPD2 expression levels were analyzed for prognosis, DNA methylation, immune infiltration, and drug sensitivity. The expression level of GPD2 was verified through quantitative real-time PCR (qPCR) and immunohistochemistry. The wound-healing and Transwell assays were used to analyze the tumor cell migration and the Matrigel invasion and apoptosis assays were performed to determine cell invasion and apoptosis. Three CAG patterns were obtained through an unsupervised consensus clustering algorithm. CAGclusterA held the best prognosis compared to the other two clusters. The CAGclusterC was characterized by poor prognosis and abundant immune cell infiltration. The TCGA-LIHC dataset, as an internal validation, also yielded similar subtype classifications. Afterward, we identified the GPD2 gene, which significantly affected the prognosis of HCC and was positively correlated with the tumor progression. The upregulation of GPD2 expression was closely related to tumorigenic signatures and immune escape. The qPCR confirmed the upregulation of GPD2 expression in HCC tumor cell lines, compared to normal liver cell lines. Immunohistochemical staining confirmed the high expression of GPD2 in HCC tumor tissues compared to normal tissues. Regulating the expression level of GPD2 can inhibit the proliferation, migration, invasion, and induce apoptosis of HCC cells. Our study comprehensively elucidated the coagulation characteristics in HCC and identified a promising oncogenic gene GPD2. Exploring targeted strategies based on coagulation-related characteristics and biomarkers may shed light on HCC treatment.
Collapse
Affiliation(s)
- Guangzhen Qu
- Department of Interventional Radiology, Beijing Chao-Yang Hospital Affiliated with Capital Medical University, Beijing, 100020, China
| | - Kun Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Weiyu Xu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Dongming Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
8
|
Song Y, Lei L, Cai X, Wei H, Yu CY. Immunomodulatory Peptides for Tumor Treatment. Adv Healthc Mater 2025; 14:e2400512. [PMID: 38657003 DOI: 10.1002/adhm.202400512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/07/2024] [Indexed: 04/26/2024]
Abstract
Peptides exhibit various biological activities, including biorecognition, cell targeting, and tumor penetration, and can stimulate immune cells to elicit immune responses for tumor immunotherapy. Peptide self-assemblies and peptide-functionalized nanocarriers can reduce the effect of various biological barriers and the degradation by peptidases, enhancing the efficiency of peptide delivery and improving antitumor immune responses. To date, the design and development of peptides with various functionalities have been extensively reviewed for enhanced chemotherapy; however, peptide-mediated tumor immunotherapy using peptides acting on different immune cells, to the knowledge, has not yet been summarized. Thus, this work provides a review of this emerging subject of research, focusing on immunomodulatory anticancer peptides. This review introduces the role of peptides in the immunomodulation of innate and adaptive immune cells, followed by a link between peptides in the innate and adaptive immune systems. The peptides are discussed in detail, following a classification according to their effects on different innate and adaptive immune cells, as well as immune checkpoints. Subsequently, two delivery strategies for peptides as drugs are presented: peptide self-assemblies and peptide-functionalized nanocarriers. The concluding remarks regarding the challenges and potential solutions of peptides for tumor immunotherapy are presented.
Collapse
Affiliation(s)
- Yang Song
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Longtianyang Lei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xingyu Cai
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, 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, 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, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410013, China
| |
Collapse
|
9
|
Zhang L, Chen X, Zhou B, Meng W, Zeng H, Chen Y, Huang G, Zhang Y, Wang H, Chen M, Chen J. Cocktail strategy-based nanomedicine: A synergistic cascade of starvation, NIR-II photothermal, and gas therapy for enhanced tumor immunotherapy. Acta Biomater 2025; 193:316-333. [PMID: 39701339 DOI: 10.1016/j.actbio.2024.11.011] [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: 07/21/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 12/21/2024]
Abstract
Immunotherapy has emerged as a highly promising strategy in the realm of cancer treatment, wherein immunogenic cell death (ICD) is considered a potential trigger for anti-tumor immunity by inducing adaptive immunity to dying cell antigens. This process is often accompanied by the exposure, active secretion, or passive release of a large number of damage-associated molecular patterns (DAMPs), which activate dendritic cells (DCs) and enhance their antigen-presenting capacity. Subsequently, it promotes the recruitment and activation of cytotoxic T lymphocytes, ultimately leading to tumor growth inhibition. In addition, polarizing the M2 phenotype of tumor-associated macrophages (TAMs) to the M1 phenotype is another way to activate anti-tumor immunity, which can further enhance the effect of anti-tumor immunotherapy. In this study, we engineered a composite nanoparticle of UiO-66-NH2@Gold nanoshells@GOx-P-Arg (denoted as UGsGP). The gold nano shells in UGsGP exhibit a broad Near-Infrared-II (NIR-II) absorption to give a high photothermal conversion efficiency and achieve photothermal therapy (PTT). The GOx in UGsGP involves the breakdown of glucose, which results in a decrease in ATP levels and an inhibition of HSP90 and HSP70 production, ultimately enhancing the heat sensitivity of the tumor for PTT. In addition, GOx-mediated starvation therapy by glucose exhaustion produces a substantial amount of hydrogen peroxide (H2O2), which can then react with P-Arg to produce intratumoral NO Thus, the synergistic effect of PTT resensitization, the photothermally-enhanced GOx-mediated starvation, and NO-based gas therapy promote the induction of ICD and the polarization of TAMs. The combination therapy exhibits significant antitumor effects both in vitro and in vivo. STATEMENT OF SIGNIFICANCE: (1) Gold nanoshells on the surface of UiO-66-NH2 display a broad absorption spectrum ranging from 900 to 1700 nm, combined with a high photothermal conversion efficiency of 74.0 %, demonstrating their remarkable ability to harness and convert light energy into heat for effective tumor ablation. (2) Under laser irradiation, GOx within the UGsGPs effectively consumes glucose, increasing intratumoral H2O2 levels, which then reacts with P-Arg to produce NO within the tumor. Concurrently, the reduction in ATP levels suppresses HSP90 and HSP70 production, thereby enhancing the tumor's sensitivity to photothermal therapy. (3) The synergistic combination of NO gas therapy, starvation therapy, and PTT promotes ICD induction and TAM polarization, thereby improving the therapeutic outcomes for primary and distant tumors.
Collapse
Affiliation(s)
- Lianying Zhang
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaotong Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Beixian Zhou
- The People's Hospital of Gaozhou, Maoming 525200, China
| | - Wei Meng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Haifeng Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yongjian Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Guoqin Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yingshan Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Huimin Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ming Chen
- The People's Hospital of Gaozhou, Maoming 525200, China.
| | - Jinxiang Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
10
|
Yang ZC, Yang ZH, Wang ZQ, Li Y, Mao GJ, Hu L, Ouyang J, Li CY. Zero-Crosstalk Tumor-Targeting Ratiometric Near-Infrared γ-Glutamyltranspeptidase Probe for Fluorescent-Guided Surgical Resection of Orthotopic Hepatic Tumor. Anal Chem 2025; 97:185-193. [PMID: 39752550 DOI: 10.1021/acs.analchem.4c03540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The challenge of "false positive" signals significantly complicates tumor localization and surgical resection, which are pivotal for successful tumor surgeries. Therefore, the development of a method for preoperative tumor localization and intraoperative margin determination holds considerable promise for improving surgical outcomes. In this study, a zero-crosstalk ratiometric tumor-targeting near-infrared (NIR) fluorescent probe was developed for precise cancer diagnosis and intraoperative navigation via NIR fluorescence imaging. This probe integrates a tumor-targeting moiety that selectively homes in on hepatocellular carcinoma cells and exhibits a highly sensitive ratiometric response to γ-glutamyltranspeptidase (GGT), characterized by a substantial emission shift from 830 to 650 nm. This unique NIR ratiometric emission property significantly enhances its effectiveness in early diagnosis and imaging-guided surgery resection. Furthermore, this zero-crosstalk probe successfully monitors GGT activity in blood, cells, and in vivo, endowing its potential for early cancer diagnosis in the clinic. Due to its efficient targeting and sensitive in situ response to GGT in both subcutaneous tumors and orthotopic hepatic tumors, the probe exhibits accurate detection capability for hepatic tumors. Additionally, by leveraging zero-crosstalk ratiometric NIR fluorescence imaging, this tumor-targeting probe can serve as a sprayable tool for delineating tumor margins with precision, thereby furnishing real-time intraoperative imaging guidance during tumor resection surgery.
Collapse
Affiliation(s)
- Zhi-Chao Yang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, Hunan Provincial University Key Laboratory for Environmental and Ecological Health, College of Chemistry, Xiangtan University, Xiangtan 411105, P.R. China
| | - Zhi-Hao Yang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, Hunan Provincial University Key Laboratory for Environmental and Ecological Health, College of Chemistry, Xiangtan University, Xiangtan 411105, P.R. China
| | - Zhi-Qing Wang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, Hunan Provincial University Key Laboratory for Environmental and Ecological Health, College of Chemistry, Xiangtan University, Xiangtan 411105, P.R. China
| | - Yao Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, Hunan Provincial University Key Laboratory for Environmental and Ecological Health, College of Chemistry, Xiangtan University, Xiangtan 411105, P.R. China
| | - Guo-Jiang Mao
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P.R. China
| | - Liufang Hu
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, Hunan Provincial University Key Laboratory for Environmental and Ecological Health, College of Chemistry, Xiangtan University, Xiangtan 411105, P.R. China
| | - Juan Ouyang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, Hunan Provincial University Key Laboratory for Environmental and Ecological Health, College of Chemistry, Xiangtan University, Xiangtan 411105, P.R. China
| | - Chun-Yan Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, Hunan Provincial University Key Laboratory for Environmental and Ecological Health, College of Chemistry, Xiangtan University, Xiangtan 411105, P.R. China
| |
Collapse
|
11
|
Wang C, Zhang X, Li Q, Hou Y, Sun M, Sun J, Lou Z, Han X, Li Y. A review of carbohydrate polymer-synthesized nanoparticles in cancer immunotherapy: Past, present and future perspectives. Int J Biol Macromol 2025; 286:138195. [PMID: 39645110 DOI: 10.1016/j.ijbiomac.2024.138195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Cancer continues to be a leading factor in mortality and tackling it has been made difficult by the development of immune escape. Furthermore, alternative treatments like surgery, chemotherapy, and radiation have been unsuccessful in eradicating cancer. Despite being effective, they have not succeeded in providing a full cancer treatment and exhibit several negative effects. The field of immunotherapy has been improved by utilizing cancer vaccines, immune checkpoint inhibitors (ICIs), and adoptive cell transfer to enhance immune responses to tumors. Nevertheless, cancer cells need to adapt and become immune to immune reactions, leading to the need for innovative treatment methods. Carbohydrate polymers and their nanoparticles have been beneficial in improving cancer immunotherapy by being customizable to specifically target the immune system. These nanoparticles can change the tumor microenvironment and accelerate immunotherapy by affecting immune cells such as T cells and dendritic cells. Incorporating both chemotherapy and phototherapy into nanoparticles can improve immunotherapy. Furthermore, besides controlling immune reactions, carbohydrate polymer nanoparticles can also be used for theranostic purposes, where they are used to image tumor cells and activate the immune system to eradicate cancer.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China
| | - Xueyao Zhang
- Department of Cardiology, First Hospital of China Medical University, Shenyang, China
| | - Qiaobei Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China
| | - Yuxin Hou
- Department of Ultrasonic Diagnosis, The Benxi Hospital of China Medical University, Benxi, China
| | - Minglu Sun
- Department of Ultrasonic Diagnosis, The Cancer Hospital of China Medical University, Shenyang, China
| | - Jun Sun
- Department of Intervention, the Fourth Hospital of China Medical University, Shenyang, China
| | - Zhe Lou
- Department of Cardiovascular Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
12
|
Xie Y, Yang Z, Shen H, Chen J, Weitz DA, Chen D, Sheng J, Liang T. Interfacial Engineering of Biocompatible Nanocapsules for Near-Infrared-Triggered Drug Release and Photothermal Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410844. [PMID: 39573938 PMCID: PMC11727245 DOI: 10.1002/advs.202410844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/08/2024] [Indexed: 01/14/2025]
Abstract
Chemotherapy is an effective option for cancer treatment. However, its clinical application is often limited by the severe side effects of chemical drugs. To overcome these limitations, a novel drug-loaded phase-change nanocapsule system is developed. These nanocapsules are assembled via one-step electrostatic self-assembly through guided interfacial engineering. The phase change material core nanocapsules demonstrate great photothermal-controlled drug release performance and exhibit excellent tumor-targeting drug delivery performance both in vitro and in vivo via the binding of hyaluronic acid shell on the nanocapsule surface with corresponding receptors on the tumor cell membrane. The phototherapy function of the nanocapsules enhances immune activation within the tumor microenvironment, as demonstrated by flow cytometry and multiplex immunohistochemistry. The developed nanocapsules are biocompatible, versatile, and scalable and offer a promising smart delivery platform for controllable near-infrared triggered drug release and photothermal therapy.
Collapse
Affiliation(s)
- Yuting Xie
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Ze Yang
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
- College of Energy Engineering and State Key Laboratory of Clean Energy UtilizationZhejiang UniversityHangzhou310003China
| | - Hang Shen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Jingyi Chen
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
| | - David A. Weitz
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
| | - Dong Chen
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003China
- College of Energy Engineering and State Key Laboratory of Clean Energy UtilizationZhejiang UniversityHangzhou310003China
| | - Jianpeng Sheng
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| |
Collapse
|
13
|
Wei FL, Zhai Y, Wang TF, Zhao JW, Wang CL, Tang Z, Shen K, Wu H, Zheng R, Du MR, Heng W, Li XX, Yan XD, Gao QY, Guo Z, Qian JX, Zhou CP. Stem cell-homing biomimetic hydrogel promotes the repair of osteoporotic bone defects through osteogenic and angiogenic coupling. SCIENCE ADVANCES 2024; 10:eadq6700. [PMID: 39485837 PMCID: PMC11529719 DOI: 10.1126/sciadv.adq6700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024]
Abstract
Osteoporotic bone defects refer to the disruption of bone structural integrity in patients with osteoporosis and pose a substantial challenge to orthopedic surgeons. In this study, we developed a biomimetic hydrogel to improve the osteogenic microenvironment and promote stem cell homing. This hydrogel served as a container for S-nitrosoglutathione and Ca2+, promoting the release of bioactive nitric oxide (NO) from bone marrow mesenchymal stem cells (BMSCs) and human vascular endothelial cells and activating the NO/cyclic guanosine monophosphate signaling pathway. These changes promote osteogenic and angiogenic couplings. The hydrogel simultaneously recruited BMSCs by conjugating the stem cell homing peptide SKPPGTSS. Using a rat distal femoral defect model, it was demonstrated that this hydrogel can effectively increase the formation of bone tissue and new blood vessels and has immune-regulating functions. We envision that this hydrogel may be a minimally invasive yet highly effective strategy for expediting the healing of osteoporotic bone defects.
Collapse
Affiliation(s)
- Fei-Long Wei
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
- Department of Orthopaedics, General Hospital of Central Theater Command (Wuhan General Hospital of Guangzhou Command, previously), Wuhan 430030, China
| | - Yuan Zhai
- Basic Medical College, Fourth Military Medical University, Xi'an 710032, China
| | - Tian-Fu Wang
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Jing-Wei Zhao
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Chao-Li Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Zhen Tang
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Kuo Shen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 710032 Xi'an, China
| | - Hao Wu
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Rui Zheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ming-Rui Du
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Wei Heng
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Xiao-Xiang Li
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Xiao-Dong Yan
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Quan-You Gao
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Zheng Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Ji-Xian Qian
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| | - Cheng-Pei Zhou
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, 710038 Xi’an, China
| |
Collapse
|
14
|
Qin Y, Ling X, Li Y, Wang J, Wang J, Rong Z, Cheng Y, Tao Z, Zhang H, Wei H, Yu CY. Histidine phosphatase-ferroptosis crosstalk modulation for efficient hepatocellular carcinoma treatment. J Nanobiotechnology 2024; 22:622. [PMID: 39402673 PMCID: PMC11476632 DOI: 10.1186/s12951-024-02918-2] [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: 05/29/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Altering the mechanisms of tumor cell death and overcoming the limitations of traditional chemotherapy is pivotal to contemporary tumor treatment. Inducing ferroptosis, while circumventing safety concerns associated with ferrous vectors, through nonferrous ferroptosis is a promising but underexplored frontier in cancer therapy. Histidine phosphatase (LHPP) has emerged as a novel therapeutic target in treating hepatocellular carcinoma (HCC), but the precise mechanism of LHPP against HCC remains unclear. Herein, we explore the effects of upregulating LHPP expression on ferroptosis and tumor immunogenicity induction by simply delivering a miRNA-363-5p inhibitor (miR-363-5pi) via a previously optimized gemcitabine-oleic acid (GOA) prodrug. Efficient miRNA encapsulation was achieved through hydrogen bonding at an optimized GOA/miRNA molar feed ratio of 250:1, affording spherical nanoparticles with a uniform hydrodynamic size of 147.1 nm and a negative potential of -21.5 mV. The mechanism of this LHPP-ferroptosis crosstalk is disclosed to be an inhibited phosphorylation of the PI3K/Akt pathway, leading to a remarkable tumor inhibition rate of 88.2% in nude mice bearing Bel-7402 tumor xenografts via a combination of LHPP-triggered nonferrous ferroptosis and GOA-induced chemotherapy. The biocompatibility of GOA/miR-363-5pi is strongly supported by their non-hematologic toxicity and insignificant organ damage. In addition, the tumor immunogenic activation potential of GOA/miR-363-5pi was finally explored. Overall, this study is the first work that elucidates the precise mechanism of LHPP for treating HCC via ferroptosis induction and achieves the transformation of chemotherapy and gene therapy into ferroptosis activation with tumor cell immunogenicity, which lays a new therapeutic foundation for the clinical treatment of HCC.
Collapse
Affiliation(s)
- Yang Qin
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xiaoli Ling
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yunxian Li
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jieqiong Wang
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiaqi Wang
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zhuoyi Rong
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yao Cheng
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zhenghao Tao
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haitao Zhang
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Hua Wei
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Cui-Yun Yu
- Pharmaceutical and Biomedical Polymers Research Laboratory, Institute of Pharmacy & Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410006, China.
| |
Collapse
|
15
|
Liu Y, Peng W, Wei T, Yuan Y, Cao X, Ma M, Sun Q, Li M, Xie F. Strong, anti-swelling, and biodegradable seaweed-based straws with surface mineralized CaCO 3 armor. Carbohydr Polym 2024; 341:122347. [PMID: 38876717 DOI: 10.1016/j.carbpol.2024.122347] [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: 02/15/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024]
Abstract
While the extensive utilization of disposable plastic straws has resulted in significant environmental issues such as microplastics and soil and ocean pollution, the quest for alternative straws for versatile use remains a formidable challenge. Here, drawing inspiration from naturally water-resistant materials such as bones and sea urchins, we have developed seaweed-based straws with significantly improved water resistance and mechanical strength via in-situ mineralization of CaCO3 on their surfaces. Specifically, the COO- groups on the G (α-L-guluronate) blocks of alginate were employed to establish a robust cross-linked network, while the COO- groups on the M (β-D-mannuronate) blocks attracted free Ca2+ through electrostatic forces, thereby promoting CaCO3 nucleation. This effectively prevents COOH groups from hydrating, reducing swelling, and results in the fabrication of nano- to micron-sized CaCO3 particles that reinforce the structure without compromising the cross-linked network. Compared with the control group, the S5% sample (prepared with 5 % Na2CO3 solution) exhibited a 102 % increase in water contact angle, a 35 % decrease in swelling degree, and a 35.5 % and 37.5 % increase in ultimate flexural and tensile stress, respectively. Furthermore, the potential use of these straws as a waste for heavy metal adsorption was investigated, addressing environmental concerns while demonstrating economic feasibility.
Collapse
Affiliation(s)
- Yuanpu Liu
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China; Qingdao Special Food Research Institute, Qingdao 266109, PR China
| | - Wen Peng
- College of Horticulture, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Ting Wei
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China; Qingdao Special Food Research Institute, Qingdao 266109, PR China
| | - Yajie Yuan
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China; Qingdao Special Food Research Institute, Qingdao 266109, PR China
| | - Xianyu Cao
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China; Qingdao Special Food Research Institute, Qingdao 266109, PR China
| | - Meng Ma
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China; Qingdao Special Food Research Institute, Qingdao 266109, PR China
| | - Qingjie Sun
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China; Qingdao Special Food Research Institute, Qingdao 266109, PR China
| | - Man Li
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China; Qingdao Special Food Research Institute, Qingdao 266109, PR China.
| | - Fengwei Xie
- Department of Chemical Engineering, University of Bath, Bath BA2 7AY, United Kingdom
| |
Collapse
|
16
|
Wang CC, Yang BQ, Feng R, Tao H, Xu BC, Zhang B. Nanoparticles prepared with biotin-esterified debranched starch as an oral carrier to improve the stability and antioxidant activity of resveratrol. Int J Biol Macromol 2024; 278:134543. [PMID: 39111511 DOI: 10.1016/j.ijbiomac.2024.134543] [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: 03/21/2024] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
In this study, biotin esterified debranched starch (Bio-DBS) nanoparticles with different molecular weights were prepared to improve the stability and antioxidant activity of resveratrol. The molecular weights of branched starch (DBS3, DBS9 and DBSp) determined by high-performance size-exclusion chromatography (HPSEC) were 3306, 3696, and 4688, respectively. Biotin was covalently coupled to DBS through the esterification reaction as a new material to prepare nanoparticles. The morphology, particle size, and loading capacity of Bio-DBS nanoparticles were all related to the molecular weights of DBS. The 1H NMR results indicated that there was a hydrogen bonding interaction between Bio-DBS and resveratrol, which contributed to the photochemical and antioxidant activity of resveratrol in the nanoparticles. The highest encapsulation efficiency (78.9 %) and loading capacity (15.78 %) of resveratrol were observed in Bio-DBS3 nanoparticles. Additionally, the cell viability was over 80 % when the concentration of Bio-DBS3 reached to 200 μg/mL. The Bio-DBS nanoparticles significantly improved the thermal stability, photostability, and antioxidant properties of resveratrol. Therefore, the Bio-DBS nanoparticles prepared in this study can be used as a promising carrier to improve the stability and antioxidant activity of resveratrol and may have potential applications in oral delivery.
Collapse
Affiliation(s)
- Chen-Chen Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food and Biological Engineering, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China
| | - Bao-Qiu Yang
- Xinjiang Production & Construction Group Key Laboratory of Agricultural Products Processing in Xinjiang South, College of Food Science and Engineering, Tarim University, Alar 843300, Xinjiang Province, PR China
| | - Ran Feng
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food and Biological Engineering, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China
| | - Han Tao
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food and Biological Engineering, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China.
| | - Bao-Cai Xu
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food and Biological Engineering, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China
| | - Bao Zhang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food and Biological Engineering, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China.
| |
Collapse
|
17
|
Li QQ, Xu D, Dong QW, Song XJ, Chen YB, Cui YL. Biomedical potentials of alginate via physical, chemical, and biological modifications. Int J Biol Macromol 2024; 277:134409. [PMID: 39097042 DOI: 10.1016/j.ijbiomac.2024.134409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Alginate is a linear polysaccharide with a modifiable structure and abundant functional groups, offers immense potential for tailoring diverse alginate-based materials to meet the demands of biomedical applications. Given the advancements in modification techniques, it is significant to analyze and summarize the modification of alginate by physical, chemical and biological methods. These approaches provide plentiful information on the preparation, characterization and application of alginate-based materials. Physical modification generally involves blending and physical crosslinking, while chemical modification relies on chemical reactions, mainly including acylation, sulfation, phosphorylation, carbodiimide coupling, nucleophilic substitution, graft copolymerization, terminal modification, and degradation. Chemical modified alginate contains chemically crosslinked alginate, grafted alginate and oligo-alginate. Biological modification associated with various enzymes to realize the hydrolysis or grafting. These diverse modifications hold great promise in fully harnessing the potential of alginate for its burgeoning biomedical applications in the future. In summary, this review provides a comprehensive discussion and summary of different modification methods applied to improve the properties of alginate while expanding its biomedical potentials.
Collapse
Affiliation(s)
- Qiao-Qiao Li
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Dong Xu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Qin-Wei Dong
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Xu-Jiao Song
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Yi-Bing Chen
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
18
|
Liu Y, Wu Y, Li Z, Wan D, Pan J. Targeted Drug Delivery Strategies for the Treatment of Hepatocellular Carcinoma. Molecules 2024; 29:4405. [PMID: 39339402 PMCID: PMC11434448 DOI: 10.3390/molecules29184405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent malignant tumors, exhibiting a high incidence rate that presents a substantial threat to human health. The use of sorafenib and lenvatinib, commonly employed as single-agent targeted inhibitors, complicates the treatment process due to the absence of definitive targeting. Nevertheless, the advent of nanotechnology has injected new optimism into the domain of liver cancer therapy. Nanocarriers equipped with active targeting or passive targeting mechanisms have demonstrated the capability to deliver drugs to tumor cells with high efficiency. This approach not only facilitates precise delivery to the affected site but also enables targeted drug release, thereby enhancing therapeutic efficacy. As medical technology progresses, there is an increasing call for innovative treatment modalities, including novel chemotherapeutic agents, gene therapy, phototherapy, immunotherapy, and combinatorial treatments for HCC. These emerging therapies are anticipated to yield improved clinical outcomes for patients, while minimizing systemic toxicity and adverse effects. Consequently, the application of nanotechnology is poised to significantly improve HCC treatment. This review focused on targeted strategies for HCC and the application of nanotechnology in this area.
Collapse
Affiliation(s)
- Yonghui Liu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Yanan Wu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Zijian Li
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Dong Wan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Jie Pan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| |
Collapse
|
19
|
Chen Y, Zhou Q, Jia Z, Cheng N, Zhang S, Chen W, Wang L. Enhancing cancer immunotherapy: Nanotechnology-mediated immunotherapy overcoming immunosuppression. Acta Pharm Sin B 2024; 14:3834-3854. [PMID: 39309502 PMCID: PMC11413684 DOI: 10.1016/j.apsb.2024.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/12/2024] [Accepted: 05/24/2024] [Indexed: 09/25/2024] Open
Abstract
Immunotherapy is an important cancer treatment method that offers hope for curing cancer patients. While immunotherapy has achieved initial success, a major obstacle to its widespread adoption is the inability to benefit the majority of patients. The success or failure of immunotherapy is closely linked to the tumor's immune microenvironment. Recently, there has been significant attention on strategies to regulate the tumor immune microenvironment in order to stimulate anti-tumor immune responses in cancer immunotherapy. The distinctive physical properties and design flexibility of nanomedicines have been extensively utilized to target immune cells (including tumor-associated macrophages (TAMs), T cells, myeloid-derived suppressor cells (MDSCs), and tumor-associated fibroblasts (TAFs)), offering promising advancements in cancer immunotherapy. In this article, we have reviewed treatment strategies aimed at targeting various immune cells to regulate the tumor immune microenvironment. The focus is on cancer immunotherapy models that are based on nanomedicines, with the goal of inducing or enhancing anti-tumor immune responses to improve immunotherapy. It is worth noting that combining cancer immunotherapy with other treatments, such as chemotherapy, radiotherapy, and photodynamic therapy, can maximize the therapeutic effects. Finally, we have identified the challenges that nanotechnology-mediated immunotherapy needs to overcome in order to design more effective nanosystems.
Collapse
Affiliation(s)
- Yunna Chen
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Qianqian Zhou
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Zongfang Jia
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Nuo Cheng
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Sheng Zhang
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Weidong Chen
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Lei Wang
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| |
Collapse
|
20
|
Wu X, Xin Y, Zhang H, Quan L, Ao Q. Biopolymer-Based Nanomedicine for Cancer Therapy: Opportunities and Challenges. Int J Nanomedicine 2024; 19:7415-7471. [PMID: 39071502 PMCID: PMC11278852 DOI: 10.2147/ijn.s460047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/18/2024] [Indexed: 07/30/2024] Open
Abstract
Cancer, as the foremost challenge among human diseases, has plagued medical professionals for many years. While there have been numerous treatment approaches in clinical practice, they often cause additional harm to patients. The emergence of nanotechnology has brought new directions for cancer treatment, which can deliver anticancer drugs specifically to tumor areas. This article first introduces the application scenarios of nanotherapies and treatment strategies of nanomedicine. Then, the noteworthy characteristics exhibited by biopolymer materials were described, which make biopolymers stand out in polymeric nanomedicine delivery. Next, we focus on summarizing the state-of-art studies of five categories of proteins (Albumin, Gelatin, Silk fibroin, Zein, Ferritin), nine varieties of polysaccharides (Chitosan, Starch, Hyaluronic acid, Dextran, cellulose, Fucoidan, Carrageenan, Lignin, Pectin) and liposomes in the field of anticancer drug delivery. Finally, we also provide a summary of the advantages and limitations of these biopolymers, discuss the prevailing impediments to their application, and discuss in detail the prospective research directions. This review not only helps readers understand the current development status of nano anticancer drug delivery systems based on biopolymers, but also is helpful for readers to understand the properties of various biopolymers and find suitable solutions in this field through comparative reading.
Collapse
Affiliation(s)
- Xixi Wu
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Yuan Xin
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Hengtong Zhang
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Liang Quan
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Qiang Ao
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| |
Collapse
|
21
|
Jia W, Yang M, Zhang W, Xu W, Zhang Y. Carrier-Free Self-Assembled Nanomedicines for Promoting Apoptosis and Inhibiting Proliferation in Hepatocellular Carcinoma. ACS Biomater Sci Eng 2024; 10:4347-4358. [PMID: 38841860 DOI: 10.1021/acsbiomaterials.4c00390] [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: 06/07/2024]
Abstract
In order to improve the effectiveness of tumor treatment and reduce the toxic side effects of drugs, we formed carrier-free multifunctional nanoparticles (BI NPs) by noncovalent interaction of berberine hydrochloride and IR780. BI NPs possessed the synergistic effects of promoting apoptosis, inhibiting proliferation and metastasis of tumors, and phototherapeutic treatment. Dispersive and passive targeting ability retention (EPR) effects of BI NPs on tumor sites in vivo could be monitored by fluorescence imaging. In addition, BI NPs exhibited effective reactive oxygen species (ROS) generation and photothermal conversion capabilities, photodynamic therapy (PDT), and photothermal therapy (PTT). Importantly, BI NPs inhibit tumor suppression through the AMPK/PI3K/AKT signaling pathway to inhibit tumor proliferation and metastasis. BI NPs not only have efficient in vivo multimodal therapeutic effects but also have good biosafety and potential clinical applications.
Collapse
Affiliation(s)
- WeiLu Jia
- Medical School, Southeast University, Nanjing 210009, China
| | - Meng Yang
- Department of Ultrasound, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - WenNing Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China
| | - WenJing Xu
- Medical School, Southeast University, Nanjing 210009, China
| | - YeWei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China
| |
Collapse
|
22
|
Xie T, Huang C, Wang Y, Zhang H, Guo P, Phann TT, Cheng Y, Lei L, Tao Z, Gao Q, Wei H, Yu CY. An "All-In-One" Immunomodulator-Engineered Clinical Translatable Immunotherapy of Advanced Hepatocellular Carcinoma. Adv Healthc Mater 2024; 13:e2304476. [PMID: 38519415 DOI: 10.1002/adhm.202304476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Clinical treatment of advanced hepatocellular carcinoma (HCC) remains a significant challenge. Utilizing 1-bromoacetyl-3,3-dinitroazetidine (RRx-001) to downregulate the expression of innate immune checkpoint molecule, cluster of differentiation 47 (CD47), provides a powerful means for treating advanced HCC containing abundant immunosuppressive macrophages. Herein engineering of a previously optimized Doxorubicin (DOX)-delivery nanoplatform based on sodium alginate is reported to further co-deliver RRx-001 (biotinylated aldehyde alginate-doxorubicin micelle prodrug nanoplatform, BEA-D@R) for efficient immunotherapy of advanced HCC. This groundbreaking technique reveals the "all-in-one" immunotherapeutic functionalities of RRx-001. Besides the previously demonstrated functions of downregulating CD47 expression and increasing reactive nitrogen species (RNS) generation, another key function of RRx-001 for downregulating the expression of the adaptive immune checkpoint molecule programmed cell death 1 ligand 1 (PDL1) is first uncovered here. Combined with the reactive oxygen species (ROS) generation and an upregulated "eat me" signal level of DOX, BEA-D@R collectively increases RNS generation, enhances T-cell infiltration, and maximizes macrophage phagocytosis, leading to an average of 40% tumor elimination in a mice model bearing an initial tumor volume of ≈300 mm3 that mimics advanced HCC. Overall, the "all-in-one" immunotherapeutic functionalities of a clinical translatable nanoplatform are uncovered for enhanced immunotherapy of advanced HCC.
Collapse
Affiliation(s)
- Ting Xie
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cong Huang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yuqing Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, 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, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Pei Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Thuy Thu Phann
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yao Cheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Longtianyang Lei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zhenghao Tao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Qing Gao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, 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, 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, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410013, China
| |
Collapse
|
23
|
Xie X, Cui M, Wang T, Yang J, Li W, Wang K, Lin M. Constructing Stiff β-Sheet for Self-Reinforced Alginate Fibers. MATERIALS (BASEL, SWITZERLAND) 2024; 17:3047. [PMID: 38998130 PMCID: PMC11242387 DOI: 10.3390/ma17133047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024]
Abstract
The application of alginate fibers is limited by relatively low mechanical properties. Herein, a self-reinforcing strategy inspired by nature is proposed to fabricate alginate fibers with minimal changes in the wet-spinning process. By adapting a coagulation bath composing of CaCl2 and ethanol, the secondary structure of sodium alginate (SA) was regulated during the fibrous formation. Ethanol mainly increased the content of β-sheet in SA. Rheological analysis revealed a reinforcing mechanism of stiff β-sheet for enhanced modulus and strength. In combination with Ca2+ crosslinking, the self-reinforced alginate fibers exhibited an increment of 39.0% in tensile strength and 71.9% in toughness. This work provides fundamental understanding for β-sheet structures in polysaccharides and a subsequent self-reinforcing mechanism. It is significant for synthesizing strong and tough materials. The self-reinforcing strategy involved no extra additives and preserved the degradability of the alginate. The reinforced alginate fibers exhibited promising potentials for biological applications.
Collapse
Affiliation(s)
- Xuelai Xie
- State Key Laboratory of Bio-Fibers and Eco-Textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, China
| | - Min Cui
- State Key Laboratory of Bio-Fibers and Eco-Textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, China
| | - Tianyuan Wang
- State Key Laboratory of Bio-Fibers and Eco-Textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, China
| | - Jinhong Yang
- State Key Laboratory of Bio-Fibers and Eco-Textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, China
| | - Wenli Li
- State Key Laboratory of Bio-Fibers and Eco-Textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, China
| | - Kai Wang
- State Key Laboratory of Bio-Fibers and Eco-Textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, China
- Institute of Flexible Electronics (IFE), Northwestern Polytechnical University (NPU), Xi’an 710072, China
| | - Min Lin
- State Key Laboratory of Bio-Fibers and Eco-Textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, China
| |
Collapse
|
24
|
Cui M, Liu S, Xie X, Yang J, Wang T, Jiao Y, Lin M, Sui K. Self-Assembly Reinforced Alginate Fibers for Enhanced Strength, Toughness, and Bone Regeneration. Biomacromolecules 2024; 25:3475-3485. [PMID: 38741285 DOI: 10.1021/acs.biomac.4c00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Material reinforcement commonly exists in a contradiction between strength and toughness enhancement. Herein, a reinforced strategy through self-assembly is proposed for alginate fibers. Sodium alginate (SA) microstructures with regulated secondary structures are assembled in acidic and ethanol as reinforcing units for alginate fibers. Acidity increases the flexibility of the helix and contributes to enhanced extendibility. Ethanol is responsible for formation of a stiff β-sheet, which enhances the modulus and strength. The structurally engineered SA assembly exhibits robust mechanical compatibility, and thus reinforced alginate fibers possess an improved tensile strength of 2.1 times, a prolonged elongation of 1.5 times, and an enhanced toughness of 3.0 times compared with SA fibers without reinforcement. The reinforcement through self-assembly provides an understanding of strengthening and toughening mechanism based on secondary structures. Due to a similar modulus with bones, reinforced alginate fibers exhibit good efficacy in accelerating bone regeneration in vivo.
Collapse
Affiliation(s)
- Min Cui
- State Key Laboratory of Bio-Fibers and Eco-textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, P. R. China
| | - Shuwei Liu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130012, P. R. China
| | - Xuelai Xie
- State Key Laboratory of Bio-Fibers and Eco-textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, P. R. China
| | - Jinhong Yang
- State Key Laboratory of Bio-Fibers and Eco-textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, P. R. China
| | - Tianyuan Wang
- State Key Laboratory of Bio-Fibers and Eco-textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, P. R. China
| | - Yuyang Jiao
- State Key Laboratory of Bio-Fibers and Eco-textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, P. R. China
| | - Min Lin
- State Key Laboratory of Bio-Fibers and Eco-textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, P. R. China
| | - Kunyan Sui
- State Key Laboratory of Bio-Fibers and Eco-textiles, College of Materials Science and Engineering, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, Qingdao University, Qingdao 266071, P. R. China
| |
Collapse
|
25
|
Jiang MC, Fang ZL, Zhang JY, Ma W, Liao LF, Yu CY, Wei H. A fully biodegradable spherical nucleic acid nanoplatform for self-codelivery of doxorubicin and miR122 for innate and adaptive immunity activation. Acta Biomater 2024; 180:407-422. [PMID: 38614414 DOI: 10.1016/j.actbio.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/12/2024] [Accepted: 04/07/2024] [Indexed: 04/15/2024]
Abstract
Facile construction of a fully biodegradable spherical nucleic acid (SNA) nanoplatform is highly desirable for clinical translations but remains rarely explored. We developed herein the first polycarbonate-based biodegradable SNA nanoplatform for self-codelivery of a chemotherapeutic drug, doxorubicin (DOX), and a human liver-specific miR122 for synergistic chemo-gene therapy of hepatocellular carcinoma (HCC). Ring-opening polymerization (ROP) of a carbonate monomer leads to a well-defined polycarbonate backbone for subsequent DOX conjugation to the pendant side chains via acidic pH-cleavage Schiff base links and miR122 incorporation to the chain termini via click coupling, affording an amphiphilic polycarbonate-DOX-miR122 conjugate, PBis-Mpa30-DOX-miR122 that can self-assemble into stabilized SNA. Besides the desired biodegradability, another notable merit of this nanoplatform is the use of miR122 not only for gene therapy but also for enhanced innate immune response. Together with the ICD-triggering effect of DOX, PBis-Mpa30-DOX-miR122 SNA-mediated DOX and miR122 codelivery leads to synergistic immunogenicity enhancement, resulting in tumor growth inhibition value (TGI) of 98.1 % significantly higher than those of the groups treated with only drug or gene in a Hepa1-6-tumor-bearing mice model. Overall, this study develops a useful strategy toward biodegradable SNA construction, and presents a drug and gene-based self-codelivery SNA with synergistic immunogenicity enhancement for efficient HCC therapy. STATEMENT OF SIGNIFICANCE: Facile construction of a fully biodegradable SNA nanoplatform is useful for in vivo applications but remains relatively unexplored likely due to the synthetic challenge. We report herein construction of a polycarbonate-based SNA nanoplatform for co-delivering a chemotherapeutic drug, DOX, and a human liver-specific miR-122 for synergistic HCC treatment. In addition to the desired biodegradability properties, this SNA nanoplatform integrates DOX-triggered ICD and miR-122-enhanced innate immunity for simultaneously activating adaptive and innate immunities, which leads to potent antitumor efficiency with a TGI value of 98.1 % in a Hepa1-6-tumor-bearing mice model.
Collapse
Affiliation(s)
- Ming-Chao Jiang
- Hengyang Medical School, School of Resources Environment and Safety Engineering, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Zhou-Long Fang
- Hengyang Medical School, School of Resources Environment and Safety Engineering, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Jin-Yan Zhang
- Hengyang Medical School, School of Resources Environment and Safety Engineering, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Wei Ma
- Hengyang Medical School, School of Resources Environment and Safety Engineering, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Luan-Feng Liao
- Hengyang Medical School, School of Resources Environment and Safety Engineering, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Cui-Yun Yu
- Affiliated Hospital of Hunan Academy of Chinese Medicine Hunan, Academy of Chinese Medicine, Changsha 410013, China; Hengyang Medical School, School of Resources Environment and Safety Engineering, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China.
| | - Hua Wei
- Hengyang Medical School, School of Resources Environment and Safety Engineering, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China.
| |
Collapse
|
26
|
Shi Q, Zhang W, Zhou Y, Huang S, Yu J, Yang M, Zhang Z, Ma J, Luo J, Rao S, Lu D, Peng S, Cao Y, Liu L, Yan Z. Hypoxia-activated cascade nanovaccine for synergistic chemoembolization-immune therapy of hepatocellular carcinoma. Biomaterials 2024; 306:122480. [PMID: 38271787 DOI: 10.1016/j.biomaterials.2024.122480] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/25/2023] [Accepted: 01/20/2024] [Indexed: 01/27/2024]
Abstract
In this work, a promising treatment strategy for triggering robust antitumor immune responses in transarterial chemoembolization of hepatocellular carcinoma (HCC) is presented. The zeolitic imidazolate framework nanoparticles loaded with hypoxia-activated prodrug tirapazamine and immune adjuvant resiquimod facilitated in situ generation of nanovaccine via a facile approach. The nanovaccine can strengthen the ability of killing the liver cancer cells under hypoxic environment, while was capable of improving immunogenic tumor microenvironment and triggering strong antitumor immune responses by increasing the primary and distant intratumoral infiltration of immune cells such as cytotoxic T cells. Moreover, a porous microcarrier, approved by FDA as pharmaceutical excipient, was designed to achieve safe and effective delivery of the nanovaccine via transarterial therapy in rabbit orthotopic VX2 liver cancer model. The microcarrier exhibited the characteristics of excellent drug loading and occlusion of peripheral artery. The collaborative delivery of the microcarrier and nanovaccine demonstrated an exciting inhibitory effect on solid tumors and tumor metastases, which provided a great potential as novel combination therapy for HCC interventional therapy.
Collapse
Affiliation(s)
- Qin Shi
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wen Zhang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yongjie Zhou
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Songjiang Huang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiaze Yu
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Minjie Yang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zihan Zhang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jingqin Ma
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jianjun Luo
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shengxiang Rao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering and MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Shaojun Peng
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| | - Yongbin Cao
- State Key Laboratory of Genetic Engineering and MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China; Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Lingxiao Liu
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Zhiping Yan
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
27
|
Li Z, Wang J, Liu J, Yu J, Wang J, Wang H, Wei Q, Liu M, Xu M, Feng Z, Zhong T, Zhang X. Multifunctional ZnO@DOX/ICG-LMHP Nanoparticles for Synergistic Multimodal Antitumor Activity. J Funct Biomater 2024; 15:35. [PMID: 38391888 PMCID: PMC10889406 DOI: 10.3390/jfb15020035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Multifunctional nanoparticles are of significant importance for synergistic multimodal antitumor activity. Herein, zinc oxide (ZnO) was used as pH-sensitive nanoparticles for loading the chemotherapy agent doxorubicin (DOX) and the photosensitizer agent indocyanine green (ICG), and biocompatible low-molecular-weight heparin (LMHP) was used as the gatekeepers for synergistic photothermal therapy/photodynamic therapy/chemotherapy/immunotherapy. ZnO was decomposed into cytotoxic Zn2+ ions, leading to a tumor-specific release of ICG and DOX. ZnO simultaneously produced oxygen (O2) and reactive oxygen species (ROS) for photodynamic therapy (PDT). The released ICG under laser irradiation produced ROS for PDT and raised the tumor temperature for photothermal therapy (PTT). The released DOX directly caused tumor cell death for chemotherapy. Both DOX and ICG also induced immunogenic cell death (ICD) for immunotherapy. The in vivo and in vitro results presented a superior inhibition of tumor progression, metastasis and recurrence. Therefore, this study could provide an efficient approach for designing multifunctional nanoparticles for synergistic multimodal antitumor therapy.
Collapse
Affiliation(s)
- Zhuoyue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jingru Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Junwei Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jianming Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jingwen Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hui Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qingchao Wei
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Man Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Meiqi Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhenhan Feng
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ting Zhong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xuan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
28
|
Chen Z, Wang X, Zhao N, Chen H, Guo G. Advancements in pH-responsive nanocarriers: enhancing drug delivery for tumor therapy. Expert Opin Drug Deliv 2023; 20:1623-1642. [PMID: 38059646 DOI: 10.1080/17425247.2023.2292678] [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] [Received: 10/16/2023] [Accepted: 12/05/2023] [Indexed: 12/08/2023]
Abstract
INTRODUCTION Tumors pose a significant global economic and health burden, with conventional cancer treatments lacking tumor specificity, leading to limited efficiency and undesirable side effects. Targeted tumor therapy is imminent. Tumor cells produce lactate and hydrogen ions (H+) by Warburg effect, forming an acidic tumor microenvironment (TME), which can be employed to design targeted tumor therapy. Recently, progress in nanotechnology has led to the development of pH-responsive nanocarriers, which have gathered significant attention. Under acidic tumor conditions, they exhibit targeted accumulation within tumor sites and controlled release profiles of therapeutic reagents, enabling precise tumor therapy. AREAS COVERED This review comprehensively summarize the principles underlying pH-responsive features, discussing various types of pH-responsive nanocarriers, their advantages, and limitations. Innovative therapeutic drugs are also examined, followed by an exploration of recent advancements in applying various pH-responsive nanocarriers as delivery systems for enhanced tumor therapy. EXPERT OPINIONS pH-responsive nanocarriers have garnered significant attention for their capability to achieve targeted accumulation of therapeutic agents at tumor sites and controlled drug delivery profiles, ultimately increasing the efficiency of tumor eradication. It is anticipated that the employment of pH-responsive nanocarriers will elevate the effectiveness and safety of tumor therapy, contributing to improved overall outcomes.
Collapse
Affiliation(s)
- Zhouyun Chen
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxiao Wang
- West China School of Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Zhao
- School of Pharmacy, Shihezi University, Shihezi, China
| | - Haifeng Chen
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Guo
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|