1
|
Wu M, Zhao K, Tao X, Du L, Chen W, Guo H, Ren H, Zhang G. Biomimetic Copper Nanozyme Reprograms Cold Tumor via Cuproptosis-Pyroptosis Crosstalk for Potent Renal Carcinoma Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29291-29304. [PMID: 40338096 DOI: 10.1021/acsami.5c03559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Immune checkpoint blockade (ICB) therapy is an emerging strategy for renal cell carcinoma (RCC). However, its clinical efficacy remains constrained by its inherently poor immunogenicity and insufficient cytotoxic T lymphocyte (CTL) infiltration. Herein, we engineer a biomimetic copper nanozyme (Cu2O-OMV) by integrating Cu2O nanoparticles with bacterial outer-membrane vesicles (OMVs) to activate the antitumor immune response and synergize with ICB therapy. The Cu2O-OMV nanozyme exhibits peroxidase (POD)-like catalytic activity and releases Cu+ to exert Fenton-like activity, generating cytotoxic hydroxyl radicals (·OH) for tumor inhibition. Furthermore, Cu+ accumulation promotes the occurrence of cuproptosis, leading to the mitochondrial aggregation of lipoylated dihydrolipoamide S-acetyltransferase and depletion of ferredoxin 1. Notably, Cu2O-OMV concurrently activates pyroptosis via the noncanonical inflammasome pathway through its intrinsic lipopolysaccharide cargo, directly inhibiting tumor growth and inducing inflammatory cytokine release. The coordinated induction of cuproptosis and pyroptosis synergistically amplifies immunogenic cell death to enhance tumor immunogenicity, thereby promoting dendritic cell maturation and CTL infiltration. After combining with αPD-L1, it effectively destroys tumor cells to activate the antitumor immune response, thereby inhibiting tumor metastasis. Our study demonstrates a biomimetic nanozyme-driven strategy that harnesses dual cuproptosis-pyroptosis pathways to enhance the tumor immunogenicity and amplify the ICB efficacy, offering a transformative approach for RCC immunotherapy.
Collapse
Affiliation(s)
- Mengtong Wu
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, Jiangsu, China
| | - Kangkang Zhao
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Xinyue Tao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, Jiangsu, China
| | - Lin Du
- Department of Urology, The First People's Hospital of Yancheng, Yancheng 224006, Jiangsu ,China
| | - Weixu Chen
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, Jiangsu, China
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Hao Ren
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, Jiangsu, China
| | - Gutian Zhang
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, Jiangsu, China
| |
Collapse
|
2
|
Li Y, Lu Y, Zhu X, Wu T, Ali K, Zhang H, Ma Z, Liu H, Wang H, Zhu H, Bi Q, Wang J, Tuo Z, Tang L. An albumin-prodrug injectable formulation for synergistic cancer immunotherapy. J Colloid Interface Sci 2025; 686:1019-1032. [PMID: 39929010 DOI: 10.1016/j.jcis.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 02/12/2025]
Abstract
The emergence of immunotherapy has significantly transformed cancer therapy, and achieved promising outcomes. However, low patient response rates and the potential immune-related adverse events remain critical challenges, warranting extensive research. In this study, we developed an albumin-prodrug injection formulation (PIF) to enhance cancer immunotherapy. The stimulator of interferon genes (STING) agonist MSA-2 was esterified with the Indoleamine 2,3-dioxygenase (IDO) inhibitor NLG919 to synthesize the prodrug MSA-NLG. Subsequently, bovine serum albumin (BSA) was employed to prepare the anti-tumor injection formulation MSA-NLG@BSA, which effectively prolonged the circulation time of the prodrug, improved pharmacokinetic properties, and promoted tumor aggregation and penetration. This strategy ensures that the drug remains inactive in normal tissues, thereby reducing immune activation-induced damage. The prodrug was dissociated into MSA-2 and NLG919 within tumor cells overexpressing esterase. The MSA-2 molecule stimulates immune cells to secrete cytotoxic cytokines, triggering an anti-tumor immune response and reshaping immune cell population. Meanwhile, NLG919 regulates the recognition and killing of tumor cells by immune cells, effectively blocking the immunosuppression caused by IDO overexpression. This study highlights the potential of a tumor environment-responsive prodrug strategy to enhance the efficacy of immunotherapy, demonstrating the promising clinical translation of the novel MSA-NLG@BSA (PIF) formulation.
Collapse
Affiliation(s)
- Yang Li
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Yuting Lu
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Xiaoling Zhu
- Department of Colorectal Surgery, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Tao Wu
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kamran Ali
- Department of Colorectal Surgery, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Huamiao Zhang
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Zhangqiang Ma
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Huiping Liu
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Hanxi Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Heping Zhu
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Qiuchen Bi
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Jianwei Wang
- Department of Colorectal Surgery, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China.
| | - Zhan Tuo
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Longguang Tang
- Department of Pharmacy, Center for Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
3
|
Jiang Y, Wang Z, Li W, Ma T, Li M, Wu S, Lin E, Flader KE, Ma M, Chang M, Li H, Wang W, Lu J. Enhanced delivery of camptothecin to colorectal carcinoma using a tumor-penetrating peptide targeting p32. Acta Biomater 2025:S1742-7061(25)00361-7. [PMID: 40379119 DOI: 10.1016/j.actbio.2025.05.036] [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: 12/26/2024] [Revised: 04/04/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Camptothesome, a sphingomyelin (SM)-conjugated camptothecin (CPT) vesicular nanotherapeutic, addresses the poor solubility and lactone instability of CPT while enhancing drug loading, pharmacokinetics, and tumor distribution compared to CPT physically entrapped in conventional liposomes. Despite these improvements, the tumor uptake remains limited. To further enhance the tumor delivery efficiency and minimize the off-target distribution, we functionalize Camptothesome with the LinTT1 peptide, a CendR motif, which binds to overexpressed p32 proteins on tumor cell surface, initiating effective transcytosis for deep tumor penetration. Via systematic screening, the optimal peptide ratio on Camptothesome is identified. LinTT1/Camptothesome significantly increases cancer cell uptake without affecting normal cell internalization, resulting in enhanced anti-colorectal cancer cells activity. Additionally, decorating Camptothesome with the LinTT1 cell-penetrating peptide enables effective transcytosis via a Golgi-dependent intracellular trafficking mechanism, significantly improving the intratumoral delivery while reducing distribution to normal tissues. In a human HCT116 xenograft colorectal cancer (CRC) mouse model, LinTT1/Camptothesome demonstrates superior antitumor efficacy compared to both Camptothesome and Onivyde by upregulating cleaved caspase-3 and γH2AX. Our study substantiates the potential of leveraging a tumor-penetrating peptide to enhance the tumor delivery efficiency of Camptothesome, maximizing its therapeutic index for improved treatment of human CRC. STATEMENT OF SIGNIFICANCE: Despite the improved tumor delivery achieved by Camptothesome, its tumor distribution and penetration remain limited. This is because the enhanced permeability and retention effect only facilitates nanotherapeutic distribution to tumor periphery through leaky vasculature. The C-end Rule (CendR) motif-neuropilin receptor system enhances tumor-homing peptides by binding to cellular surface receptors, triggering transcytosis. Herein, LinTT1, the most potent CendR peptide that binds to the overexpressed p32 receptor on cancer cells, was effectively engineered onto Camptothesome using thiol-maleimide lipid chemistry. The LinTT1/Camptothesome significantly enhanced tumor uptake and penetration while minimizing accumulation in normal tissues, demonstrating remarkable anticancer efficacy in a human xenograft colorectal cancer model. Our findings highlight the critical role of tumor-homing peptides in unlocking the full therapeutic potential of Camptothesome.
Collapse
Affiliation(s)
- Yanhao Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Teng Ma
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Mengwen Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Shuang Wu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Ethan Lin
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Karlie Elizabeth Flader
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Mengjiao Ma
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Mengyang Chang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Hongmin Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Wei Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, United States; Clinical and Translational Oncology Program, The University of Arizona Cancer Center, Tucson, AZ 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ 85721, United States; Southwest Environmental Health Sciences Center, The University of Arizona, Tucson 85721, United States.
| |
Collapse
|
4
|
Bai XF, Ma JC, Zhang C, Chen Z, He J, Cheng SX, Zhang XZ. Click Chemistry-Assisted Rejuvenation of Aging T Cells Sensitizes Aged Mice to Tumor Immunotherapy. J Am Chem Soc 2025; 147:16694-16704. [PMID: 40310278 DOI: 10.1021/jacs.5c05312] [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: 05/02/2025]
Abstract
Enormous resources have been devoted to address the suboptimal response of tumor patients to immunotherapy. However, a crucial yet often overlooked factor in these effects is the strong correlation between the occurrence and development of tumors and the immune dysfunction associated with aging. Our study aims to rejuvenate aging T cells within tumor-draining lymph nodes (TdLNs) by using targeted delivery of rapamycin, a macrolide capable of mitigating aging-related decline in immune function, thereby enhancing the antitumor efficacy of immunotherapy in aged mice. The targeted delivery system relies on a bioorthogonal reaction that harnesses the click chemistry between the azide (N3) groups artificially introduced onto TdLNs and the dibenzocyclooctyne (DBCO) groups attached to the rapamycin-loaded micelles administered intradermally. Experimental data demonstrate that this approach has effectively restored the functionality of impaired CD8+ T cells in aged mice, thereby enhancing the antitumor response to immune checkpoint blockade (ICB) therapy to levels comparable to those in young mice. This study presents a promising strategy to combat the resistance to immunotherapeutic approaches commonly encountered among elderly tumor patients.
Collapse
Affiliation(s)
- Xue-Feng Bai
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Jun-Chi Ma
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Zhu Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Jinlian He
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Department of Traditional Chinese Medicine of Zhongnan Hospital, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
5
|
Zhang W, Wang X, Wang J, Su C, Bao Y, Xiong M. The Relationship between In Vivo Toxicity and Responsive pH in Transistor-Like pH-Sensitive Nanodetergents. SMALL METHODS 2025; 9:e2401263. [PMID: 39676467 DOI: 10.1002/smtd.202401263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Subacidity-responsive materials (saRMs) have attracted considerable attention for disease-specific pH-responsive imaging and therapy. However, the guidance for their pH-responsive design, aimed at achieving effective responses at lesion sites while minimizing unwanted responses in normal tissues, is inadequate and challenged by the subtle pH difference between the desired responsive pH and the pH of normal tissues. Here, the correlation between the responsive pH of 'proton transistor' nanodetergents (pTNTs) is investigated and the in vivo toxicity caused by unwanted responses in normal tissues, taking advantage of their refined responsive pH and the easily characterized membranolytic activity and cytotoxicity following response. It is designed and selected five pTNTs that undergo a refined transition from an inactive "OFF" state with sealed membranolytic activity and cytotoxicity to an active "ON" state with potent membranolytic activity and cytotoxicity within a 0.1 pH perturbation at transition pH (pHt) values of 7.2, 7.1, 6.9, 6.8, and 6.7, respectively. A significant correlation between the in vivo toxicity of these pTNTs and their pHt for membranolytic activity is observed. And non-negligible changes in the organ toxicity of pTNTs are induced by every 0.1 or 0.2 pH shift of pHt. After intravenous administration, pTNTs with a pHt value of 7.2 or 7.1 induced significant hepatotoxicity and cardiotoxicity, while no significant toxicity is detected for pTNTs with pHt values ranging from 6.8 to 6.7. This hepatoxicity is found to be associated with the tissue's pH environment-dependent activation of membranolytic activity. This study can provide guidance for designing pH-responsive membranolytic materials and saRMs to minimize their toxicity and unwanted response in normal tissues.
Collapse
Affiliation(s)
- Weinan Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- School of Biomedical Engineering, and Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, P. R. China
| | - Xiaochuan Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Jihong Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Chanjuan Su
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, and Medical Research Center, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Yan Bao
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, and Medical Research Center, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Menghua Xiong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
6
|
Quadros M, Goyal M, Chauhan G, Gadhave D, Gupta V. An Inhaled Nanoemulsion Encapsulating a Herbal Drug for Non-Small Cell Lung Cancer (NSCLC) Treatment. Pharmaceutics 2025; 17:540. [PMID: 40430833 DOI: 10.3390/pharmaceutics17050540] [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: 03/20/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Celastrol (Cela), a phytochemical extracted from Tripterygium wilfordii, has been extensively investigated for its potential anti-inflammatory, anti-psoriatic, antioxidant, neuroprotective, and antineoplastic properties. However, its clinical translation is limited due to poor bioavailability, low solubility, and nonspecific toxicity. This study aimed to develop and evaluate an inhalable Cela-loaded nanoemulsion (NE) formulation to enhance targeted drug delivery and therapeutic efficacy in non-small cell lung cancer (NSCLC). Methods: The NE formulation was optimized using Capmul MCM (25%), Tween 80 (20%), Transcutol HP (5%), and water (50%) as the oil, surfactant, co-surfactant, and aqueous phase, respectively. Physicochemical characterization included globule size, zeta potential, and drug release in simulated lung fluid. In vitro aerosolization performance, cytotoxicity in NSCLC cell lines (A549), scratch and clonogenic assays, and 3D tumor spheroid models were employed to assess therapeutic potential. Results: The NE showed a globule size of 201.4 ± 3.7 nm and a zeta potential of -15.7 ± 0.2 mV. Drug release was sustained, with 20.4 ± 5.5%, 29.1 ± 10%, 64.6 ± 4.1%, and 88.1 ± 5.2% released at 24, 48, 72, and 120 h, respectively. In vitro aerosolization studies indicated a median aerodynamic particle size of 4.8 ± 0.2 μm, confirming its respirability in the lung. Cell culture studies indicated higher toxicity of NE-Cela in NSCLC cells. NE-Cela significantly reduced A549 cell viability, showing a ~6-fold decrease in IC50 (0.2 ± 0.1 μM) compared to Cela alone (1.2 ± 0.2 μM). Migration and clonogenic assays demonstrated reduced cell proliferation, and 3D spheroid models supported its therapeutic activity in tumor-like environments. Conclusions: The inhalable NE-Cela formulation improved Cela's physicochemical limitations and demonstrated enhanced anti-cancer efficacy in NSCLC models. These findings support its potential as a targeted, well-tolerated therapeutic option for lung cancer treatment.
Collapse
Affiliation(s)
- Mural Quadros
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Mimansa Goyal
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Gautam Chauhan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Dnyandev Gadhave
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Vivek Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| |
Collapse
|
7
|
Zhou Y, Wu Y, Sun S, Wang W, Zhou S, Liu H, Guo Y, Hong S, Ding F, Cai H. Self-Assembled Glycopeptide as a Biocompatible mRNA Vaccine Platform Elicits Robust Antitumor Immunity. ACS NANO 2025; 19:14727-14741. [PMID: 40203215 DOI: 10.1021/acsnano.4c15187] [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
Since the emergence of the COVID-19 pandemic, mRNA vaccines have garnered significant attention. Delivery systems affect the effectiveness of mRNA vaccines, yet there remains a scarcity of vectors that can achieve safe and efficient delivery of mRNA. We took advantage of self-assembled glycopeptides (SAPs) to develop a vector named Man-MPm, which was coupled with mannose and manganese ions to achieve lymph node targeting and STING pathway activation. The Man-MPm-based mRNA vaccine exhibited high biosafety across various administration routes, eliciting robust antigen-specific immune responses within lymph nodes. Due to the elevated antitumor immunity, Man-MPm significantly suppressed tumor growth and extended the survival period of mice in melanoma prevention and treatment models as well as in a colon cancer model. Our findings show that Man-MPm addresses the challenges to safety and effectiveness associated with mRNA delivery by incorporating a lymph node-targeting ligand and a STING pathway agonist onto highly biocompatible SAP, and Man-MPm holds great potential for developing mRNA tumor vaccines.
Collapse
Affiliation(s)
- Yang Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Ye Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Shengjie Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Wei Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Siai Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Hua Liu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Yajing Guo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Sheng Hong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Feiqing Ding
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangming 518107, China
| |
Collapse
|
8
|
Guo C, Lin L, Wang Y, Jing J, Gong Q, Luo K. Nano drug delivery systems for advanced immune checkpoint blockade therapy. Theranostics 2025; 15:5440-5480. [PMID: 40303342 PMCID: PMC12036873 DOI: 10.7150/thno.112475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been widely utilized in the first-line therapy of various types of cancer. However, immune-related adverse events (irAEs) and resistance to ICIs remain intractable challenges for immune checkpoint blockade (ICB) therapy during clinic treatment. Nano drug delivery systems (NDDSs) have shown promising potential to improve anticancer efficacy and reduce side effects of small molecular drugs. The combination of nanotechnology and ICB provides new opportunities to overcome the challenges of immunotherapy. Nanoplatforms have been employed for direct delivery of ICIs, and they are preferred vehicles for combination therapy of ICIs and other therapeutic agents. In this review, the strategies of using NDDSs for advancing ICB therapy in recent years are surveyed, emphasizing the employment of NDDSs for combination treatment by ICIs and other agents to manipulate antitumor immunity. Analysis of current strategies for applying NDDSs for ICB leads to future research directions and development trends.
Collapse
Affiliation(s)
- Chenqi Guo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Lin
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yihan Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- West China School of Medicine, Chengdu 610041, China
| | - Jing Jing
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
9
|
Tian M, Wu D, Gou X, Li R, Zhang X. Genetic modulation of rare earth nanoparticle biotransformation shapes biological outcomes. Nat Commun 2025; 16:3429. [PMID: 40210885 PMCID: PMC11985947 DOI: 10.1038/s41467-025-58520-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/21/2025] [Indexed: 04/12/2025] Open
Abstract
The biotransformation of nanoparticles plays a crucial role in determining their biological fate and responses. Although a few engineering strategies (e.g., surface functionalization and shape control) have been employed to regulate the fate of nanoparticles, the genetic control of nanoparticle biotransformation remains an unexplored avenue. Herein, we utilized a CRISPR-based genome-scale knockout approach to identify genes involved in the biotransformation of rare earth oxide (REO) nanoparticles. We found that the biotransformation of REOs in lysosomes could be genetically controlled via SMPD1. Specifically, suppression of SMPD1 inhibited the transformation of La2O3 into sea urchin-shaped structures, thereby protecting against lysosomal damage, proinflammatory cytokine release, pyroptosis and RE-induced pneumoconiosis. Overall, our study provides insight into how to control the biological fate of nanomaterials.
Collapse
Affiliation(s)
- Mingming Tian
- School of Ecology and Environmental Science, Yunnan University & Yunnan Key Laboratory for Plateau Mountain Ecology and Restoration of Degraded Environments, Kunming, Yunnan, China
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, Nanjing, P. R. China
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, P. R. China
| | - Di Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow Medical College, Soochow University, Suzhou, P. R. China
| | - Xiao Gou
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, Nanjing, P. R. China
| | - Ruibin Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow Medical College, Soochow University, Suzhou, P. R. China.
- Nanotechnology Centre, Centre for Energy and Environmental Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, Czech Republic.
| | - Xiaowei Zhang
- School of Ecology and Environmental Science, Yunnan University & Yunnan Key Laboratory for Plateau Mountain Ecology and Restoration of Degraded Environments, Kunming, Yunnan, China.
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, Nanjing, P. R. China.
| |
Collapse
|
10
|
Gao F, Qiu X, Baddi S, He S, Wang S, Zhao C, Dou X, Feng C. Chiral Nanofibers of Camptothecin Trigger Pyroptosis for Enhanced Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202423446. [PMID: 39803865 DOI: 10.1002/anie.202423446] [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: 12/02/2024] [Indexed: 01/22/2025]
Abstract
Camptothecin (CPT), a chemotherapeutic agent, demonstrates significant potential in cancer therapy. However, as a drug, CPT molecule suffers from poor water solubility, limited bioavailability, and insufficient immune response. Herein, we construct CPT nanofibers (CNF) with a right-handed chiral property via supramolecular self-assembly, which significantly overcomes the solubility barriers associated with bioavailability and improves tumor immune prognosis. The CNF exhibits high chiral asymmetry factor (gabs) (~0.11) and remarkable structure stability under pH 6.5 condition. By formulating chiral CNF with mitochondrial-targeted DSPE-PEG-TPP, CNF accumulates specifically in the mitochondria of cancer cells, leading to mitochondrial dysfunction and a 3.42-fold increase in reactive oxygen species (ROS) generation compared to the CPT molecule. This ROS amplification activates the caspase-1/gasdermin D (GSDMD) pathway, inducing pyroptosis that promotes M1 macrophage polarization and enhances CD8+ T-cell-dependent antitumor immunity. Consequently, CNF achieves 1.8-fold greater growth inhibition of distant tumor and reduces tumor metastasis compared to the CPT molecule. Our innovative platform, assembling CPT molecules into chiral CNF structure, is highly anticipated to overcome the current clinical limitations of CPT molecules and offer a new direction for the development of next-generation immunotherapy strategies.
Collapse
Affiliation(s)
- Fengli Gao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaxin Qiu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Sravan Baddi
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Sijia He
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuting Wang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Changli Zhao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoqiu Dou
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chuanliang Feng
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
11
|
Li M, Zhang H, Xiong P, He Y, Zhou W, Wu C, Liao X, Zhang W, Yang H, Liu Y. DNA origami-based composite nanosandwich for iteratively potentiated chemo-immunotherapy. J Control Release 2025; 379:452-465. [PMID: 39809421 DOI: 10.1016/j.jconrel.2025.01.031] [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: 08/25/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Developing effective nanoplatforms for chemo-immunotherapy to achieve enhanced tumor suppression and systemic antitumor immunity has recently received extensive attention. Herein, we formulated a multifunctional DNA sandwich nanodevice, DSWAC/siPD-L1, based on triangular DNA origami, to implement enhanced cancer chemo-immunotherapy. Taking advantage of the tumor-targeting ability of the AS1411 aptamer, DSWAC/siPD-L1 efficiently delivered doxorubicin (DOX), CpG, and siPD-L1 into tumor cells. Moreover, the sandwich cavity spatially protects siPD-L1 from degradation, and the featured design of the DNA/RNA duplex linkers ensures effective intracellular release of siPD-L1. The pH-responsive release of cytotoxic DOX induces apoptosis and initial mild immunogenic cell death of tumor cells, presenting antigens to enhance the maturation of dendritic cells (DCs) with the assistance of the immune adjuvant CpG, thereby activating cytotoxic T lymphocytes to amplify antitumor immunity. Simultaneously, siPD-L1 downregulated the endogenous expression of PD-L1 to inhibit adaptive tumor immune escape. DSWAC/siPD-L1 initiated the iterative revolution of the cancer-immunity cycle, leading to the inhibition of primary and metastatic tumors, as demonstrated by DC maturation and T-cell infiltration in established subcutaneous primary tumor model and metastatic lung tumor model. Furthermore, the superior antitumor effect of DSWAC/siPD-L1 resulted in approximately 91 % inhibition of primary tumor growth and 93 % prevention of lung metastasis. Collectively, this study describes a siPD-L1-based sandwich DNA nanodevice functionalized with AS1411/CpG for enhanced cancer chemo-immunotherapy, inspiring the creation of more innovative drug nanocarriers and the exploitation of novel cancer therapies.
Collapse
Affiliation(s)
- Mengyue Li
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hanxi Zhang
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Peizheng Xiong
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China
| | - Yuhan He
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Wanyi Zhou
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Chunhui Wu
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, PR China.
| | - Wei Zhang
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| | - Hong Yang
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| | - Yiyao Liu
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China; Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, PR China; Department of Urology, Deyang People's Hospital, Deyang 618099, Sichuan, PR China.
| |
Collapse
|
12
|
Chen C, Zhang H, Han J, Yang L, Li S, Jia Q, Fang J, Ling P, Wang S. Synthesis and antitumor activity of ultra-low molecular weight hyaluronic acid-decorated camptothecin conjugates. Carbohydr Polym 2025; 351:123144. [PMID: 39778985 DOI: 10.1016/j.carbpol.2024.123144] [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: 07/05/2024] [Revised: 11/16/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Camptothecin (CPT) exhibits potent anticancer activity, but its clinical application is limited by poor solubility and severe side effects. Hyaluronic acid (HA) is gaining attention in drug delivery systems due to its excellent biocompatibility and tumor-targeting properties. In this study, we conjugated CPT to the reducing end of ultra-low molecular weight HA to create a series of HA-decorated CPT conjugates. These novel conjugates offer significant advantages over traditional HA-drug formulations, including well-defined structures and consistent drug-loading rates. In vitro studies demonstrated that these HA-decorated conjugates exhibited enhanced anti-proliferative and targeting effects towards various tumor cells. Furthermore, in vivo studies showed that HA-CPT nanoparticles significantly inhibited tumor growth with minimal side effects, as evidenced by stable body weight and histological analyses in treated mice. The approach of using structurally well-defined HA as a carrier for site-specific drug modification expands the potential applications of HA and enhances the therapeutic efficacy of conventional drugs.
Collapse
Affiliation(s)
- Changsheng Chen
- National Glycoengineering Research Center and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Henan Zhang
- National Glycoengineering Research Center and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Jingjun Han
- National Glycoengineering Research Center and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Lin Yang
- National Glycoengineering Research Center and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Shuang Li
- National Glycoengineering Research Center and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Qingwen Jia
- Shandong Freda Pharmaceutical Group Co. Ltd, Jinan, Shandong 250101, China
| | - Junqiang Fang
- National Glycoengineering Research Center and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China
| | - Peixue Ling
- National Glycoengineering Research Center and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China.
| | - Shuaishuai Wang
- National Glycoengineering Research Center and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong 266237, China; Suzhou Research Institute of Shandong University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
13
|
Du L, Min H, Meng Y, Zhang K, Wang L, Zhang Y, Sun P, Zhang W, Qi Y, Wu G. LRG1-Targeted Camptothecin Nanomicelles with Simultaneous Delivery of Olaparib for Enhanced Colorectal Cancer Chemotherapy. NANO LETTERS 2025; 25:3130-3140. [PMID: 39947227 DOI: 10.1021/acs.nanolett.4c05354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Camptothecin (CPT), an effective topoisomerase I inhibitor used in colorectal cancer chemotherapy, often faces limitations due to severe toxicities. Addressing this, we developed OCENM, a nanomedicine featuring the CPT-ET conjugate─comprising a cathepsin B-sensitive linker and CPT linked to the ET peptide targeting leucine-rich alpha-2-glycoprotein 1 (LRG1) and encapsulating Olaparib, a potent poly ADP-ribose polymerase (PARP) inhibitor. OCENM aims for precise CPT delivery to colorectal cancer sites overexpressing LRG1, while Olaparib disrupts compromised DNA repair pathways, enhancing DNA damage and promoting increased tumor cell apoptosis. Our results show OCENM accumulates preferentially in colorectal cancer models through LRG1 targeting and triggers synergistic tumor cell apoptosis through the dual action of enhanced DNA damage and inhibited repair mechanisms. This study not only highlights the potential of LRG1 as a strategic target for nanomedicine delivery but also underscores the therapeutic promise of combining CPT with Olaparib for colorectal cancer treatment.
Collapse
Affiliation(s)
- Lin Du
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
| | - Huan Min
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yongkang Meng
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Kejuan Zhang
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Longdi Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
| | - Yana Zhang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
| | - Peichun Sun
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Wei Zhang
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Yingqiu Qi
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Gang Wu
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| |
Collapse
|
14
|
Wang Y, Li H, Lin J, Li Y, Zhang K, Li H, Fu Q, Jiang Y. Engineering nanozyme immunomodulator with magnetic targeting effect for cascade-enzyodynamic and ultrasound-reinforced metallo-immunotherapy in prostate carcinoma. Nat Commun 2025; 16:1876. [PMID: 39987131 PMCID: PMC11846840 DOI: 10.1038/s41467-025-57190-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
Conventional immunotherapy exhibits low response rates due to the immunosuppressive tumor microenvironment (TME). To overcome this limitation, this study introduces ZFPG nanoparticles (ZFPG NPs) with ZnFe2O4@Pt cores and glucose oxidase (GOx) shells. The ZFPG NPs possess five-enzyme activities, good sonosensitivity, and remarkable magnetic targeting properties, which facilitate sono-metallo-immunotherapy for prostate cancer treatment in male mice. Specifically, the magnetic targeting ability effectively improves their accumulation in tumors while still showing enrichment in the liver and kidneys. The multienzyme cascade catalysis and sonosensitivity of these NPs effectively deplete glutathione and glucose, and enhance the generation and utilization of H2O2, thereby inducing multiple ROS bursts. Furthermore, these comprehensive effects up-regulate the HMOX1 to promote the Fe2+ and lipid peroxides accumulation, thereby inducing immunogenic ferroptosis. This strategy facilitates anti-tumor immunity by ameliorating the immunosuppressive TME and inhibiting lung metastatic progression. This joint warfare strategy offers a powerful solution to address conventional immunotherapy limitations.
Collapse
Affiliation(s)
- Yandong Wang
- Key Laboratory for Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, P. R. China
| | - Haodong Li
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, P. R. China
| | - Junyang Lin
- Department of Urology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, P. R. China
| | - Yutang Li
- Key Laboratory for Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, P. R. China
| | - Keqin Zhang
- Department of Urology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, P. R. China
| | - Hui Li
- Key Laboratory for Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, P. R. China
| | - Qiang Fu
- Department of Urology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, P. R. China.
- Key Laboratory of Urinary Diseases in Universities of Shandong, Shandong First Medical University, Jinan, P. R. China.
| | - Yanyan Jiang
- Key Laboratory for Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, P. R. China.
| |
Collapse
|
15
|
Wu S, Lu J. Liposome-Enabled Nanomaterials for Muscle Regeneration. SMALL METHODS 2025:e2402154. [PMID: 39967365 DOI: 10.1002/smtd.202402154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/08/2025] [Indexed: 02/20/2025]
Abstract
Muscle regeneration is a vital biological process that is crucial for maintaining muscle function and integrity, particularly for the treatment of muscle diseases such as sarcopenia and muscular dystrophy. Generally, muscular tissues can self-repair and regenerate under various conditions, including acute or chronic injuries, aging, and genetic mutation. However, regeneration becomes challenging beyond a certain threshold, particularly in severe muscle injuries or progressive diseases. In recent years, liposome-based nanotechnologies have shown potential as promising therapeutic strategies for muscle regeneration. Liposomes offer an adaptable platform for targeted drug delivery due to their cell membrane-like structure and excellent biocompatibility. They can enhance drug solubility, stability, and targeted delivery while minimizing systemic side effects by different mechanisms. This review summarizes recent advancements, discusses current applications and mechanisms, and highlights challenges and future directions for possible clinical translation of liposome-based nanomaterials in the treatment of muscle diseases. It is hoped this review offers new insights into the development of liposome-enabled nanomedicine to address current limitations.
Collapse
Affiliation(s)
- Shuang Wu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
- Clinical and Translational Oncology Program, The University of Arizona Cancer Center, Tucson, AZ, 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
16
|
Qin J, Liu J, Wei Z, Li X, Chen Z, Li J, Zheng W, Liu H, Xu S, Yong T, Zhao B, Gou S, Ju S, Teng GJ, Yang X, Gan L. Targeted intervention in nerve-cancer crosstalk enhances pancreatic cancer chemotherapy. NATURE NANOTECHNOLOGY 2025; 20:311-324. [PMID: 39496914 DOI: 10.1038/s41565-024-01803-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 09/04/2024] [Indexed: 11/06/2024]
Abstract
Nerve-cancer crosstalk has gained substantial attention owing to its impact on tumour growth, metastasis and therapy resistance. Effective therapeutic strategies targeting tumour-associated nerves within the intricate tumour microenvironment remain a major challenge in pancreatic cancer. Here we develop Escherichia coli Nissle 1917-derived outer membrane vesicles conjugated with nerve-binding peptide NP41, loaded with the tropomyosin receptor kinase (Trk) inhibitor larotrectinib (Lar@NP-OMVs) for tumour-associated nerve targeting. Lar@NP-OMVs achieve efficient nerve intervention to diminish neurite growth by disrupting the neurotrophin/Trk signalling pathway. Moreover, OMV-mediated repolarization of M2-like tumour-associated macrophages to an M1-like phenotype results in nerve injury, further accentuating Lar@NP-OMV-induced nerve intervention to inhibit nerve-triggered proliferation and migration of pancreatic cancer cells and angiogenesis. Leveraging this strategy, Lar@NP-OMVs significantly reduce nerve infiltration and neurite growth promoted by gemcitabine within the tumour microenvironment, leading to augmented chemotherapy efficacy in pancreatic cancer. This study sheds light on a potential avenue for nerve-targeted therapeutic intervention for enhancing pancreatic cancer therapy.
Collapse
Affiliation(s)
- Jiaqi Qin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoxia Chen
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jianye Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Wenxia Zheng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Haojie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, China
| | - Ben Zhao
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Shanmiao Gou
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenghong Ju
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, China.
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
17
|
Chen H, Tan F, Zhang Y, Xie B, Luo A. Enhancing PARP inhibitor efficacy using reduction-responsive nanoparticles encapsulating NADP. J Mater Chem B 2025; 13:955-964. [PMID: 39624987 DOI: 10.1039/d4tb01797h] [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
Poly(ADP-ribose) polymerase inhibitors (PARPi) have shown success in cancer chemotherapy; however, not all tumors respond effectively to PARPi treatment, even in the presence of BRCA1/2 mutations or homologous recombination (HR) repair defects. NADP+ was recently identified as an endogenous inhibitor of ADP-ribosylation with the potential to sensitize cancer cells to PARPi, yet its lack of membrane permeability poses a significant challenge to its clinical application. In this study, we developed reduction-responsive nanoparticles (NPs) containing disulfide bonds, which can be cleaved in the reductive environment of tumor cells. These NPs encapsulate NADP+ and the commercially available PARP inhibitor olaparib. The uptake of these NPs significantly increases the intracellular concentration of NADP+, which negatively regulates DNA damage-induced PARylation and impairs DNA damage repair. The combined effects of elevated NADP+ levels and olaparib synergistically suppress tumor cell growth. Overall, our study offers a promising strategy for the clinical application of NADP+.
Collapse
Affiliation(s)
- Hao Chen
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Fan Tan
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Yukui Zhang
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
- National Chromatographic Research and Analysis Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Bingteng Xie
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Aiqin Luo
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
18
|
Zhang Z, Hu Y, Ding Y, Zhang X, Dong X, Xie L, Yang Z, Hu ZW. Dual-Enzyme-Instructed Peptide Self-Assembly to Boost Immunogenic Cell Death by Coordinating Intracellular Calcium Overload and Chemotherapy. ACS NANO 2025; 19:488-503. [PMID: 39754594 DOI: 10.1021/acsnano.4c10119] [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: 01/06/2025]
Abstract
The concept of immunogenic cell death (ICD) induced by chemotherapy as a potential synergistic modality for cancer immunotherapy has been widely discussed. Unfortunately, most chemotherapeutic agents failed to dictate effective ICD responses due to their defects in inducing potent ICD signaling. Here, we report a dual-enzyme-instructed peptide self-assembly platform of CPMC (CPT-GFFpY-PLGVRK-Caps) that cooperatively utilizes camptothecin (CPT) and capsaicin (Caps) to promote ICD and engage systemic adaptive immunity for tumor rejection. Although CPT and Caps respectively prevent tumor progression by inhibiting type-I DNA topoisomerase and activating transient receptor potential cation channel subfamily V member 1 (TRPV1) for intracellular calcium overload, neither alone effectively stimulates sufficient ICD signaling to meet immunotherapeutic needs. CPMC, sequentially allowing an active Caps derivative of VRK-Caps and CPT to release extracellularly and intracellularly, can synergize two distinct apoptosis pathways stimulated by Caps and CPT to increase tumor immunogenicity and elicit systemic T-cell-based immunity. Consequently, CPMC facilitates the generation of improved tumor-specific cytotoxic T-cell responses and sustained immunological memory, successfully suppressing both primary and distant tumors. Moreover, CPMC can render tumors susceptible to PD-L1 blockade and synergize with an antiprogrammed cell death-ligand 1 (aPDL1) antibody for tumor inhibition. Combining two cancer chemotherapeutic drugs with low ICD-stimulating capacity using a peptide self-assembly strategy was demonstrated to boost ICD responses and potentiate cancer immunotherapy.
Collapse
Affiliation(s)
- Zhenghao Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Yuhan Hu
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Yinghao Ding
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Xiangyang Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Xiao Dong
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Limin Xie
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Zhimou Yang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Zhi-Wen Hu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
19
|
Han C, Xiao S, Xing Z, Xu X, Wang M, Han X, Adeli M, Qiu L, Ye L, Cheng C. NADPH Oxidases-Inspired Reactive Oxygen Biocatalysts with Electron-Rich Pt Sites to Potently Amplify Immune Checkpoint Blockade Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407644. [PMID: 39400421 DOI: 10.1002/adma.202407644] [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: 05/29/2024] [Revised: 07/19/2024] [Indexed: 10/15/2024]
Abstract
Clinical immune checkpoint blockade (ICB)-based immunotherapy of malignant tumors only elicits durable responses in a minority of patients, primarily due to the highly immunosuppressive tumor microenvironment. Although inducing immunogenic cell death (ICD) through reactive oxygen biocatalyst represents an attractive therapeutic strategy to amplify ICB, currently reported biocatalysts encounter insurmountable challenges in achieving high ROS-generating activity to induce potent ICD. Here, inspired by the natural catalytic characteristics of NADPH oxidases, the design of efficient, robust, and electron-rich Pt-based redox centers on the non-stoichiometric W18O49 substrates (Pt─WOx) to serve as bioinspired reactive oxygen biocatalysts to potently activate the ICD, which eventually enhance cancer immune responses and amplifies the ICB-based immunotherapy is reported. These studies demonstrate that the Pt─WOx exhibits rapid electron transfer capability and can promote the formation of electron-rich and low oxophilic Pt redox centers for superior reactive oxygen biocatalysis, which enables the Pt─WOx-based inducers to trigger endoplasmic reticulum stress directly and stimulate immune responses potently for amplifying the anti-PD-L1-based ICB therapy. This bioinspired design provides a straightforward strategy to engineer efficient, robust, and electron-rich reactive oxygen biocatalysts and also opens up a new avenue to create efficient ICD inducers for primary/metastatic tumor treatments.
Collapse
Affiliation(s)
- Chuyi Han
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Sutong Xiao
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Zhenyu Xing
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xiaohui Xu
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xianglong Han
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Mohsen Adeli
- Institute of Chemistry and Biochemistry, Freie Universitat Berlin, Takustr. 3, 14195, Berlin, Germany
- Department of Organic Chemistry, Faculty of Chemistry, Lorestan University, Khorramabad, 68137-17133, Iran
| | - Li Qiu
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ling Ye
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chong Cheng
- Department of Endodontics, Department of Orthodontics, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
20
|
Zhang N, Song J, Han Y. Research Progress of Phospholipid Vesicles in Biological Field. Biomolecules 2024; 14:1628. [PMID: 39766335 PMCID: PMC11726895 DOI: 10.3390/biom14121628] [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: 11/08/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
Due to their high biocompatibility, biodegradability, and facile surface functionalization, phospholipid vesicles as carriers have garnered significant attention in the realm of disease diagnosis and treatment. On the one hand, phospholipid vesicles can function as probes for the detection of various diseases by encapsulating nanoparticles, thereby enabling the precise localization of pathological changes and the monitoring of disease progression. On the other hand, phospholipid vesicles possess the capability to selectively target and deliver therapeutic agents, including drug molecules, genes and immune modulators, to affected sites, thereby enhancing the sustained release of these agents and improving therapeutic efficacy. Recent advancements in nanotechnology have led to an increased focus on the application of phospholipid vesicles in drug delivery, biological detection, gene therapy, and cell mimics. This review aims to provide a concise overview of the structure, characteristics, and preparation techniques of phospholipid vesicles of varying sizes. Furthermore, we will summarize the latest research developments regarding their use as nanomedicines and gene carriers in disease treatment. Additionally, we will elucidate the potential of phospholipid vesicles in facilitating the internalization, controlled release, and targeted delivery of therapeutic substrates. Through this review, we aspire to enhance the understanding of the evolution of phospholipid vesicles within the biological field, outline prospective research, and address the forthcoming challenges associated with phospholipid vesicles in disease diagnosis and treatment.
Collapse
Affiliation(s)
- Na Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China;
| | - Jie Song
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China;
| | - Yuchun Han
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, CAS Research/Education Center for Excellence in Molecular Sciences, Beijing National Laboratory for Molecular Sciences (BNLMS), Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
21
|
Yang JB, Xu DZ, Zhang ZH, Zhang X, Ren ZX, Lu ZL, Liu R, Liu Y. Multifunctional System with Camptothecin and [12]aneN 3 Units for Effective In Vivo Anti Pancreatic Cancer through Synergistic Chemotherapy, Gene Therapy, and Photodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67203-67215. [PMID: 39585759 DOI: 10.1021/acsami.4c12936] [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: 11/27/2024]
Abstract
Maximizing drug cargo carrying capacity in blood circulation, controlling the in vivo fate of nanoparticles, and precisely drug release to tumor targets are the main aims of multifunctional nanomedicine-based antitumor therapy. Here we combined macrocyclic polyamine di(triazole-[12]aneN3) (M) and chemical drug camptothecin (CPT, C) through photosensitizer 1,1-dicyano-2-phenyl-2-(4-diphenylamino) phenyl-ethylene (DT) containing the cleavable disulfide (S) linkage as an all-in-one theranostic nanoprodrug, MDTSC. The corresponding compound with carbon chain (C) linkage, MDTCC, was also prepared for a comparison study. MDTSC showed the ability to carry plasmids, including the p53 tumor suppressor gene, to form lipoplexes with a size of ∼150 nm. Further addition of DOPE-PEG2k resulted in the hybrid lipoplexes MDTSC/DOPE-PEG2k@DNA, which showed good stability in blood circulation and good biocompatibility to normal cell lines. Experiments demonstrated that the hybrid lipoplexes were able to realize the successful cellular uptake and endosomal escape, to generate ROS under visible light irradiation as well as to trigger the localized release of CPT and the plasmid encoding p53 in tumor cells. In vitro, the hybrid lipoplexes showed better EGFP expression than the commercial Lipo2000, and markedly reduced tumor cell proliferation and migration rate irrespective of whether the BxPC-3 cell lines were grown on plates or 3D tumor spheroids. In vivo, the hybrid lipoplexes showed effective anticancer activity by reducing the BxPC-3 pancreatic tumor growth by 99% through the synergetic combination of chemotherapy, photodynamic therapy, and gene therapy. This research represented the first example of using a cocktail of three therapeutic approaches to achieve cooperative and effective anti pancreatic cancer treatment in vivo and in vitro.
Collapse
Affiliation(s)
- Jing-Bo Yang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- China National Institute for Food and Drug Control, Institute of Chemical Drug Control, HuaTuo Road 29, Beijing 100050, China
| | - De-Zhong Xu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Zi-Han Zhang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Xi Zhang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Zhi-Xuan Ren
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Zhong-Lin Lu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Rui Liu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education; College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Yang Liu
- China National Institute for Food and Drug Control, Institute of Chemical Drug Control, HuaTuo Road 29, Beijing 100050, China
| |
Collapse
|
22
|
Chen YY, Zeng XT, Gong ZC, Zhang MM, Wang KQ, Tang YP, Huang ZH. Euphorbia Pekinensis Rupr. sensitizes colorectal cancer to PD-1 blockade by remodeling the tumor microenvironment and enhancing peripheral immunity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156107. [PMID: 39368338 DOI: 10.1016/j.phymed.2024.156107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/28/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Immune checkpoint blockade, such as monoclonal antibodies targeting programmed cell death protein 1 (PD-1), has been a major breakthrough in the treatment of several cancers, but has limited effect in colorectal cancer (CRC), which is a highly prevalent cancer worldwide. Current chemotherapy-based strategies to boost PD-1 response have many limitations. And the role of peripheral immunity in boosting PD-1 response continues to attract attention. Therefore, candidate combinations of PD-1 blockade need to be drugs with multi-targets and multi-modulatory functions. However, it is still unknown whether traditional Chinese medicines with such property can enhance the applicability and efficacy of PD-1 blockade in colorectal cancer. METHODS Euphorbia Pekinensis extract (EP) was prepared and the constituents were analyzed by HPLC. CRC cells were used for in vitro experiments, including cell viability assay, colony formation assay, flow cytometry for 7-AAD staining, western blotting for caspase 3 and caspase 7, HMGB1 and ATP detection. An orthotopic CT26 mouse model was subsequently used to investigate the combination of EP and PD-1 blockade therapy. Tumor volume and tumor weight were assessed, tumor tissues were subjected to histopathological HE staining and TUNEL staining, and tumor-infiltrating immune cells were evaluated by immunofluorescence staining. RNA-sequencing, target prediction and pathway analysis were further employed to explore the mechanism. Molecular docking and cellular thermal shift assay (CETSA) were utilized to verify the direct target of the core component of EP. And, loss-of-function analysis was carried to confirm the upstream-downstream relationship. Flow cytometry was employed to analyze CD8+ T cells in the peripheral blood and spleen. RESULTS The main constituents of EP are diterpenoids and flavonoids. EP dramatically suppresses CRC cell growth and exerts its cytotoxic effect by triggering immunogenic cell death in vitro. Moreover, EP synergizes with PD-1 blockade to inhibit tumorigenesis in tumor-bearing mice. Disruption of ISX nuclear localization by helioscopinolide E is a central mechanism of EP-induced apoptosis in CRC cell. Meanwhile, EP activates immune response by upregulating Phox2b to reshape the immune microenvironment. In addition, EP regulates peripheral immunity by regulating the T cell activation and proliferation, and the ratio of CD8+ T cells in peripheral blood is drastically increased, thereby enhancing the therapeutic efficacy of anti-PD1 immunotherapy. CONCLUSION EP triggers intra-tumor immunogenic cell death and modulates the immunoregulatory signaling to elicit the tumor immunogenicity. Moreover, EP participates in transcriptional activation of immune response-related pathways. Consequently, multiple stimulating functions of EP on macro- and micro-immune potentiates the anti-tumor effect of PD-1 blockade in CRC.
Collapse
Affiliation(s)
- Yan-Yan Chen
- Wuxi Cancer Institute, Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiao-Tao Zeng
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, China
| | - Zhi-Cheng Gong
- Wuxi Cancer Institute, Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Mei-Mei Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, China
| | - Kai-Qing Wang
- Wuxi Cancer Institute, Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, China.
| | - Zhao-Hui Huang
- Wuxi Cancer Institute, Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
23
|
Chen L, Yin Q, Zhang H, Zhang J, Yang G, Weng L, Liu T, Xu C, Xue P, Zhao J, Zhang H, Yao Y, Chen X, Sun S. Protecting Against Postsurgery Oral Cancer Recurrence with an Implantable Hydrogel Vaccine for In Situ Photoimmunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309053. [PMID: 39467056 PMCID: PMC11633475 DOI: 10.1002/advs.202309053] [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: 11/23/2023] [Revised: 08/20/2024] [Indexed: 10/30/2024]
Abstract
Oral squamous cell carcinoma (OSCC) often recurs aggressively and metastasizes despite surgery and adjuvant therapy, driven by postoperative residual cancer cells near the primary tumor site. An implantable in situ vaccine hydrogel was designed to target residual OSCC cells post-tumor removal. This hydrogel serves as a reservoir for the sustained localized release of δ-aminolevulinic acid (δ-ALA), enhancing protoporphyrin IX-mediated photodynamic therapy (PDT), and a polydopamine-hyaluronic acid composite for photothermal therapy (PTT). Additionally, immune adjuvants, including anti-CD47 antibodies (aCD47) and CaCO3 nanoparticles, are directly released into the resected tumor bed. This approach induces apoptosis of residual OSCC cells through sequential near-infrared irradiation, promoting calcium interference therapy (CIT). The hydrogel further stimulates immunogenic cell death (ICD), facilitating the polarization of tumor-associated macrophages from the M2 to the M1 phenotype. This facilitates phagocytosis, dendritic cell activation, robust antigen presentation, and cytotoxic T lymphocyte-mediated cytotoxicity. In murine OSCC models, the in situ vaccine effectively prevents local recurrence, inhibits orthotopic OSCC growth and pulmonary metastases, and provides long-term protective immunity against tumor rechalle nge. These findings support postoperative in situ vaccination with a biocompatible hydrogel implant as a promising strategy to minimize residual tumor burden and reduce recurrence risk after OSCC resection.
Collapse
Affiliation(s)
- Lan Chen
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Qiqi Yin
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Handan Zhang
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Jie Zhang
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Guizhu Yang
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Lin Weng
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Tao Liu
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Chenghui Xu
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Pengxin Xue
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Jinchao Zhao
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Han Zhang
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Yanli Yao
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Xin Chen
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Shuyang Sun
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| |
Collapse
|
24
|
Xie Q, Liu X, Liu R, Pan J, Liang J. Cellular mechanisms of combining innate immunity activation with PD-1/PD-L1 blockade in treatment of colorectal cancer. Mol Cancer 2024; 23:252. [PMID: 39529058 PMCID: PMC11555832 DOI: 10.1186/s12943-024-02166-w] [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: 10/03/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
PD-1/PD-L1 blockade therapies have displayed extraordinary clinical efficacy for melanoma, renal, bladder and lung cancer; however, only a minority of colorectal cancer (CRC) patients benefit from these treatments. The efficacy of PD-1/PD-L1 blockade in CRC is limited by the complexities of tumor microenvironment. PD-1/PD-L1 blockade immunotherapy is based on T cell-centered view of tumor immunity. However, the onset and maintenance of T cell responses and the development of long-lasting memory T cells depend on innate immune responses. Acknowledging the pivotal role of innate immunity in anti-tumor immune response, this review encapsulates the employment of combinational therapies those involve PD-1/PD-L1 blockade alongside the activation of innate immunity and explores the underlying cellular mechanisms, aiming to harnessing innate immune responses to induce long-lasting tumor control for CRC patients who received PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Xiaolin Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Rengyun Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
| |
Collapse
|
25
|
Luo Z, Huang Y, Chen S, Zhang B, Huang H, Dabiri S, Chen Y, Zhang A, Andreas AR, Li S. Delivery of PARP inhibitors through 2HG-incorporated liposomes for synergistically targeting DNA repair in cancer. Cancer Lett 2024; 604:217268. [PMID: 39321912 DOI: 10.1016/j.canlet.2024.217268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/15/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
PARP inhibitors (PARPi) benefit only a small subset of patients with DNA homologous recombination (HR) defects. In addition, long-term administration of a PARPi can lead to the development of drug resistance. 2-Hydroxyglutarate (2HG) has long been known as an oncometabolite but is capable of inducing an HR defect, which makes tumor cells exquisitely sensitive to PARPi. To facilitate the translation of this discovery to the treatment of both HR-deficient and HR-proficient tumors, a liposomal formulation was developed for codelivery of 2HG and veliparib, a PARPi. A sequential loading protocol was developed such that the initial loading of 2HG into liposomes greatly facilitated the subsequent, pH gradient-driven remote loading of veliparib. The liposomes co-loaded with veliparib and 2HG exhibited favorable stability, slow kinetics of drug release, and targeted delivery to the tumor. Furthermore, the veliparib/2HG liposomes demonstrated enhanced anti-tumor activity in both PARPi-resistant BRCA mutant cancer and BRCA wildtype cancer by synergistically enhancing the defect in DNA repair. Moreover, combination of veliparib and 2HG via liposomal co-delivery also augmented the function of cytotoxic T cells by activating the STING pathway and downregulating PD-L1 expression via 2HG-induced hypermethylation.
Collapse
Affiliation(s)
- Zhangyi Luo
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Shangyu Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Bei Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Haozhe Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Sheida Dabiri
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Yuang Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Anju Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Alexis R Andreas
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh, School of Pharmacy, Pittsburgh, PA, USA.
| |
Collapse
|
26
|
Liu Q, Yang Y, Pan M, Shi K, Mo D, Li Y, Wang M, Guo L, Qian Z. Camptothecin multifunctional nanoparticles effectively achieve a balance between the efficacy of breast cancer treatment and the preservation of intestinal homeostasis. Bioact Mater 2024; 41:413-426. [PMID: 39184827 PMCID: PMC11342206 DOI: 10.1016/j.bioactmat.2024.07.032] [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: 06/12/2024] [Revised: 07/27/2024] [Accepted: 07/27/2024] [Indexed: 08/27/2024] Open
Abstract
Camptothecin (CPT) exhibits potent antitumor activity; however, its clinical application is limited by significant gastrointestinal adverse effects (GAEs). Although the severity of GAEs associated with CPT derivatives has decreased, the incidence rate of these adverse effects has remained high. CPT multifunctional nanoparticles (PCRHNs) have the potential to increase the efficacy of CPT while reducing side effects in major target organs; however, the impact of PCRHNs on the GAEs from CPT remains uncertain. Here, we investigated the therapeutic effects of PCRHNs and different doses of CPT and examined their impacts on the intestinal barrier and the intestinal microbiota. We found that the therapeutic efficacy of PCRHNs + Laser treatment was superior to that of high-dose CPT, and PCRHNs + Laser treatment also provided greater benefits by helping maintain intestinal barrier integrity, intestinal microbiota diversity, and intestinal microbiota abundance. In summary, compared to high-dose CPT treatment, PCRHNs + Laser treatment can effectively balance therapeutic effects and GAEs. A high dose of CPT promotes the enrichment of the pathogenic bacteria Escherichia-Shigella, which may be attributed to diarrhea caused by CPT, thus leading to a reduction in microbial burden; additionally, Escherichia-Shigella rapidly grows and occupies niches previously occupied by other bacteria that are lost due to diarrhea. PCRHNs + Laser treatment increased the abundance of Lactobacillus (probiotics), possibly due to the photothermal effect of the PCRHNs. This effect increased catalase activity, thus facilitating the conversion of hydrogen peroxide into oxygen within tumors and increasing oxygen levels in the body, which is conducive to the growth of facultative anaerobic bacteria.
Collapse
Affiliation(s)
- Qingya Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yun Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Pan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kun Shi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dong Mo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yicong Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linfeng Guo
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
27
|
Wang N, Liu Y, Peng D, Zhang Q, Zhang Z, Xu L, Yin L, Zhao X, Lu Z, Peng J. Copper-Based Composites Nanoparticles Improve Triple-Negative Breast Cancer Treatment with Induction of Apoptosis-Cuproptosis and Immune Activation. Adv Healthc Mater 2024; 13:e2401646. [PMID: 39001628 DOI: 10.1002/adhm.202401646] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/05/2024] [Indexed: 11/12/2024]
Abstract
The synergistic effect of apoptosis and cuproptosis, along with the activation of the immune system, presents a promising approach to enhance the efficacy against triple-negative breast cancer (TNBC). Here, two prodrugs are synthesized: a reactive oxygen species (ROS)-responsive prodrug PEG-TK-DOX and a glutathione (GSH)-responsive prodrug PEG-DTPA-SS-CPT. These prodrugs are self-assembled and chelated Cu2+ to prepare nanoparticle PCD@Cu that simultaneously loaded doxorubicin (DOX), camptothecin (CPT), and Cu2+. The elevated levels of ROS and GSH in TNBC cells disrupted the PCD@Cu structure, leading to the release of Cu+, DOX, and CPT and the depletion of GSH. DOX and CPT triggered apoptosis with immunogenic cell death (ICD) in TNBC cells. Simultaneously, PCD@Cu downregulated the expression of copper transporting ATPase 2 (ATP7B), causing a significant accumulation of copper ions in TNBC cells. This further induced the aggregation of lipoylated dihydrolipoamide S-acetyltransferase (DLAT) and downregulation of iron-sulfur (Fe-S) cluster proteins, ultimately leading to cuproptosis and ICD in TNBC. In vitro and in vivo experiments confirmed that PCD@Cu induced apoptosis and cuproptosis in TNBC and activated the immune system, demonstrating strong anti-tumor capabilities. Moreover, PCD@Cu exhibited an excellent biosafety profile. Overall, this study provides a promising strategy for effective TNBC therapy.
Collapse
Affiliation(s)
- Ning Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yichao Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Dezhou Peng
- School of Materials Science and Engineering, Jilin University, Changchun, 130012, China
| | - Qiyu Zhang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Zhibo Zhang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Xuerong Zhao
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Zhi Lu
- Department of Nuclear Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| |
Collapse
|
28
|
Huang H, Tong QS, Chen Y, Liu XY, Liu R, Shen S, Du JZ, Wang J. PAMAM-Based Polymeric Immunogenic Cell Death Inducer To Potentiate Cancer Immunotherapy. J Am Chem Soc 2024; 146:29189-29198. [PMID: 39387453 DOI: 10.1021/jacs.4c11636] [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: 10/15/2024]
Abstract
Immunogenic cell death (ICD) has been widely employed to potentiate cancer immunotherapy due to its capability to activate the anticancer immune response. Although various ICD inducers have been described, the development of synthetic materials with intrinsic ICD-inducing competency has rarely been reported. Herein, we identify a derivative of the fourth generation polyamidoamine (PAMAM) modified with multiple seven-membered heterocyclic rings, G4P-C7A, as a robust ICD inducer. G4P-C7A evokes characteristic release of damage-associated molecular patterns in tumor cells and induces efficient dendritic cell maturation. Mechanistic studies suggest that G4P-C7A can selectively accumulate in the endoplasmic reticulum and mitochondria to generate reactive oxygen species. G4P-C7A-treated tumor cells can work as potent vaccines to protect against secondary tumor implantation. Either local or systemic injection of G4P-C7A alone can effectively inhibit tumor growth by eliciting robust antitumor immune response. The combination of G4P-C7A with immunotherapeutic antibodies such as anti-PD1 (aPD-1) and anti-CD47 (aCD47) further potentiates the antitumor effect in either CT26 or 4T1 tumor model. This study offers a simple but effective strategy to induce ICD to boost cancer immunotherapy.
Collapse
Affiliation(s)
- Hua Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Qi-Song Tong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yang Chen
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xiao-Yue Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Rong Liu
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
29
|
Chen Y, Wang Z, Zhang C, Su Y, Zhou T, Hu K. Revealing the mechanism of natural product-induced immunogenic cell death: opening a new chapter in tumor immunotherapy. Front Immunol 2024; 15:1470071. [PMID: 39445013 PMCID: PMC11496055 DOI: 10.3389/fimmu.2024.1470071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
This review underscores the role of natural products in inducing immunogenic cell death (ICD) as a key strategy in tumor immunotherapy. It reveals that natural products can activate ICD through multiple pathways-apoptosis, autophagy, pyroptosis, and necroptosis-leading to the release of danger-associated molecular patterns (DAMPs), dendritic cell activation, and improved antigen presentation, which together stimulate a potent anti-tumor immune response. The study also demonstrates the enhanced therapeutic potential of combining natural products with immune checkpoint inhibitors. With a focus on translating preclinical findings into clinical practice, this review consolidates recent discoveries and suggests future research paths, offering both theoretical insights and practical guidance for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Yukun Chen
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenzhi Wang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chi Zhang
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yisa Su
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tian Zhou
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kaiwen Hu
- Department of Oncology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
30
|
Wang Z, Li W, Lu J. Sphingomyelin-derived epacadostat nanovesicle enhances IDO1 inhibition for improved melanoma combination immunotherapy. Genes Immun 2024; 25:440-442. [PMID: 38858600 PMCID: PMC11479849 DOI: 10.1038/s41435-024-00281-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Affiliation(s)
- Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.
- NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, AZ, 85721, USA.
- BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA.
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
31
|
Tanita K, Koseki Y, Kumar S, Taemaitree F, Mizutani A, Nakatsuji H, Suzuki R, Dao ATN, Fujishima F, Tada H, Ishida T, Saijo K, Ishioka C, Kasai H. Carrier-free nano-prodrugs for minimally invasive cancer therapy. NANOSCALE 2024; 16:15256-15264. [PMID: 39073351 DOI: 10.1039/d4nr01763c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
An anticancer nanodrug with few side effects that does not require the use of a nanocarrier, polyethylene glycol, or other additives has been developed. We have fabricated nano-prodrugs (NPDs) composed only of homodimeric prodrugs of the anticancer agent SN-38, which contains a disulfide bond. The prodrugs are stable against hydrolysis but selectively release SN-38 when the disulfide bond is cleaved by glutathione, which is present in high concentrations in cancer cells. The best-performing NPDs showed good dispersion stability in nanoparticle form, and animal experiments revealed that they possess much higher antitumor activity than irinotecan, a clinically applied prodrug of SN-38. This performance was achieved by improving tumor accumulation due to the size effect and targeted drug release mechanism. The present study provides an insight into the development of non-invasive NPDs with high pharmacological activity, and also offers new possibilities for designing prodrug molecules that can release drugs in response to various kinds of triggers.
Collapse
Affiliation(s)
- Keita Tanita
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.
| | - Yoshitaka Koseki
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.
- Research Institute for Electronic Science, Hokkaido University, N20W10, Kita-ku, Sapporo, 001-0020, Japan
| | - Sanjay Kumar
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.
| | - Farsai Taemaitree
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.
- Research Institute for Electronic Science, Hokkaido University, N20W10, Kita-ku, Sapporo, 001-0020, Japan
| | - Asuka Mizutani
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.
| | - Hirotaka Nakatsuji
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.
| | - Ryuju Suzuki
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.
| | - Anh Thi Ngoc Dao
- Graduate School of Engineering, Nagasaki University, 1-14 Bunkyo, Nagasaki, 852-8521, Japan
| | - Fumiyoshi Fujishima
- Department of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Hiroshi Tada
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Ken Saijo
- Department of Medical Oncology, Tohoku University Hospital, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- Department of Clinical Oncology, Tohoku University Graduate School of Medicine, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Chikashi Ishioka
- Department of Medical Oncology, Tohoku University Hospital, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- Department of Clinical Oncology, Tohoku University Graduate School of Medicine, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Hitoshi Kasai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.
| |
Collapse
|
32
|
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
|
33
|
Xu S, Lan H, Huang C, Ge X, Zhu J. Mechanisms and emerging strategies for irinotecan-induced diarrhea. Eur J Pharmacol 2024; 974:176614. [PMID: 38677535 DOI: 10.1016/j.ejphar.2024.176614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Irinotecan (also known as CPT-11) is a topoisomerase I inhibitor first approved for clinical use as an anticancer agent in 1996. Over the past more than two decades, it has been widely used for combination regimens to treat various malignancies, especially in gastrointestinal and lung cancers. However, severe dose-limiting toxicities, especially gastrointestinal toxicity such as late-onset diarrhea, were frequently observed in irinotecan-based therapy, thus largely limiting the clinical application of this agent. Current knowledge regarding the pathogenesis of irinotecan-induced diarrhea is characterized by the complicated metabolism of irinotecan to its active metabolite SN-38 and inactive metabolite SN-38G. A series of enzymes and transporters were involved in these metabolic processes, including UGT1A1 and CYP3A4. Genetic polymorphisms of these metabolizing enzymes were significantly associated with the occurrence of irinotecan-induced diarrhea. Recent discoveries and progress made on the detailed mechanisms enable the identification of potential biomarkers for predicting diarrhea and as such guiding the proper patient selection with a better range of tolerant dosages. In this review, we introduce the metabolic process of irinotecan and describe the pathogenic mechanisms underlying irinotecan-induced diarrhea. Based on the mechanisms, we further outline the potential biomarkers for predicting the severity of diarrhea. Finally, based on the current experimental evidence in preclinical and clinical studies, we discuss and prospect the current and emerging strategies for the prevention of irinotecan-induced diarrhea.
Collapse
Affiliation(s)
- Shengkun Xu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China
| | - Huiyin Lan
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China
| | - Chengyi Huang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China
| | - Xingnan Ge
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
34
|
Wang B, Wang W, Xu Y, Liu R, Li R, Yang P, Zhao C, Dai Z, Wang Y. Manipulating Redox Homeostasis of Cancer Stem Cells Overcome Chemotherapeutic Resistance through Photoactivatable Biomimetic Nanodiscs. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308539. [PMID: 38326103 DOI: 10.1002/smll.202308539] [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: 12/12/2023] [Revised: 01/09/2024] [Indexed: 02/09/2024]
Abstract
Tumor heterogeneity remains a significant obstacle in cancer therapy due to diverse cells with varying treatment responses. Cancer stem-like cells (CSCs) contribute significantly to intratumor heterogeneity, characterized by high tumorigenicity and chemoresistance. CSCs reside in the depth of the tumor, possessing low reactive oxygen species (ROS) levels and robust antioxidant defense systems to maintain self-renewal and stemness. A nanotherapeutic strategy is developed using tumor-penetrating peptide iRGD-modified high-density lipoprotein (HDL)-mimetic nanodiscs (IPCND) that ingeniously loaded with pyropheophorbide-a (Ppa), bis (2-hydroxyethyl) disulfide (S-S), and camptothecin (CPT) by synthesizing two amphiphilic drug-conjugated sphingomyelin derivatives. Photoactivatable Ppa can generate massive ROS which as intracellular signaling molecules effectively shut down self-renewal and trigger differentiation of the CSCs, while S-S is utilized to deplete GSH and sustainably imbalance redox homeostasis by reducing ROS clearance. Simultaneously, the depletion of GSH is accompanied by the release of CPT, which leads to subsequent cell death. This dual strategy successfully disturbed the redox equilibrium of CSCs, prompting their differentiation and boosting the ability of CPT to kill CSCs upon laser irradiation. Additionally, it demonstrated a synergistic anti-cancer effect by concurrently eliminating therapeutically resistant CSCs and bulk tumor cells, effectively suppressing tumor growth in CSC-enriched heterogeneous colon tumor mouse models.
Collapse
Affiliation(s)
- Bo Wang
- Department of Ultrasound, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wuwan Wang
- Department of Ultrasound, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yunxue Xu
- Department of Biomedical Engineering, College of Future Technology, National Biomedical, Imaging Center, Peking University, Beijing, 100871, China
| | - Renfa Liu
- Department of Biomedical Engineering, College of Future Technology, National Biomedical, Imaging Center, Peking University, Beijing, 100871, China
| | - Rui Li
- Department of Biomedical Engineering, College of Future Technology, National Biomedical, Imaging Center, Peking University, Beijing, 100871, China
| | - Peipei Yang
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Chenyang Zhao
- Department of Ultrasound, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Future Technology, National Biomedical, Imaging Center, Peking University, Beijing, 100871, China
| | - Yong Wang
- Department of Ultrasound, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| |
Collapse
|
35
|
Li Z, Yang J, Ren B, Fan Q, Huang L, Guo S, Zhou R, Chen S, Feng J, Yan C, Chen X, Shen Z. Double-Layered Hollow Mesoporous Cuprous Oxide Nanoparticles for Double Drug Sequential Therapy of Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313212. [PMID: 38670140 DOI: 10.1002/adma.202313212] [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: 12/05/2023] [Revised: 04/08/2024] [Indexed: 04/28/2024]
Abstract
Cancer stem cells (CSCs) are one of the determinants of tumor heterogeneity and are characterized by self-renewal, high tumorigenicity, invasiveness, and resistance to various therapies. To overcome the resistance of traditional tumor therapies resulting from CSCs, a strategy of double drug sequential therapy (DDST) for CSC-enriched tumors is proposed in this study and is realized utilizing the developed double-layered hollow mesoporous cuprous oxide nanoparticles (DL-HMCONs). The high drug-loading contents of camptothecin (CPT) and all-trans retinoic acid (ATRA) demonstrate that the DL-HMCON can be used as a generic drug delivery system. ATRA and CPT can be sequentially loaded in and released from CPT3@ATRA3@DL-HMCON@HA. The DDST mechanisms of CPT3@ATRA3@DL-HMCON@HA for CSC-containing tumors are demonstrated as follows: 1) the first release of ATRA from the outer layer induces differentiation from CSCs with high drug resistance to non-CSCs with low drug resistance; 2) the second release of CPT from the inner layer causes apoptosis of non-CSCs; and 3) the third release of Cu+ from DL-HMCON itself triggers the Fenton-like reaction and glutathione depletion, resulting in ferroptosis of non-CSCs. This CPT3@ATRA3@DL-HMCON@HA is verified to possess high DDST efficacy for CSC-enriched tumors with high biosafety.
Collapse
Affiliation(s)
- Zongheng Li
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Jing Yang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Bin Ren
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Qingdeng Fan
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Lin Huang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Shuai Guo
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - RuiLong Zhou
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Sijin Chen
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Jie Feng
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Chenggong Yan
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Zheyu Shen
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai South Road, Baiyun, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
36
|
Han C, Gu Y, Miao R, Han W, Zhang Q, Hu X, Li H, Zhang Y, Chen M. DLPC induces ferroptosis in cancer cells. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1401-1405. [PMID: 38932700 PMCID: PMC11532213 DOI: 10.3724/abbs.2024097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/22/2024] [Indexed: 06/28/2024] Open
Affiliation(s)
- Chunmiao Han
- />State Key Laboratory for ComplexSevereand Rare DiseasesInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and School of Basic MedicinePeking Union Medical CollegeBeijing100005China
| | - Yingying Gu
- />State Key Laboratory for ComplexSevereand Rare DiseasesInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and School of Basic MedicinePeking Union Medical CollegeBeijing100005China
| | - Renling Miao
- />State Key Laboratory for ComplexSevereand Rare DiseasesInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and School of Basic MedicinePeking Union Medical CollegeBeijing100005China
| | - Wanhong Han
- />State Key Laboratory for ComplexSevereand Rare DiseasesInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and School of Basic MedicinePeking Union Medical CollegeBeijing100005China
| | - Qianying Zhang
- />State Key Laboratory for ComplexSevereand Rare DiseasesInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and School of Basic MedicinePeking Union Medical CollegeBeijing100005China
| | - Xiaodi Hu
- />State Key Laboratory for ComplexSevereand Rare DiseasesInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and School of Basic MedicinePeking Union Medical CollegeBeijing100005China
| | - Hu Li
- />State Key Laboratory for ComplexSevereand Rare DiseasesInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and School of Basic MedicinePeking Union Medical CollegeBeijing100005China
| | - Yong Zhang
- />State Key Laboratory for ComplexSevereand Rare DiseasesInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and School of Basic MedicinePeking Union Medical CollegeBeijing100005China
| | - Meihong Chen
- />State Key Laboratory for ComplexSevereand Rare DiseasesInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and School of Basic MedicinePeking Union Medical CollegeBeijing100005China
| |
Collapse
|
37
|
Zhong D, Hou X, Pan D, Li Z, Gong Q, Luo K. Bioorthogonal In Situ Polymerization of Dendritic Agents for Hijacking Lysosomes and Enhancing Antigen Presentation in Cancer Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403588. [PMID: 38490170 DOI: 10.1002/adma.202403588] [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: 03/10/2024] [Revised: 03/13/2024] [Indexed: 03/17/2024]
Abstract
A low-generation lysine dendrimer, SPr-G2, responds to intracellular glutathione to initiate bioorthogonal in situ polymerization, resulting in the formation of large assemblies in mouse breast cancer cells. The intracellular large assemblies of SPr-G2 can interact with lysosomes to induce lysosome expansion and enhance lysosomal membrane permeabilization, leading to major histocompatibility complex class I upregulation on tumor cell surfaces and ultimately tumor cell death. Moreover, the use of the SPr-G2 dendrimer to conjugate the chemotherapeutic drug, camptothecin (CPT), can boost the therapeutic potency of CPT. Excellent antitumor effects in vitro and in vivo are obtained from the combinational treatment of the SPr-G2 dendrimer and CPT. This combinational effect also enhances antitumor immunity through promoting activation of cytotoxic T cells in tumor tissues and maturation of dendritic cells. This study can shed new light on the development of peptide dendritic agents for cancer therapy.
Collapse
Affiliation(s)
- Dan Zhong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xingyu Hou
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiqian Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361000, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
38
|
Huang PY, Liang SY, Xiang Y, Li MR, Wang MR, Liu LH. Endoplasmic Reticulum-Targeting Self-Assembly Nanosheets Promote Autophagy and Regulate Immunosuppressive Tumor Microenvironment for Efficient Photodynamic Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311056. [PMID: 38377262 DOI: 10.1002/smll.202311056] [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: 11/29/2023] [Revised: 02/07/2024] [Indexed: 02/22/2024]
Abstract
The poor efficiency and low immunogenicity of photodynamic therapy (PDT), and the immunosuppressive tumor microenvironment (ITM) lead to tumor recurrence and metastasis. In this work, TCPP-TER-Zn@RSV nanosheets (TZR NSs) that co-assembled from the endoplasmic reticulum (ER)-targeting photosensitizer TCPP-TER-Zn nanosheets (TZ NSs for short) and the autophagy promoting and indoleamine-(2, 3)-dioxygenase (IDO) inhibitor-like resveratrol (RSV) are fabricated to enhance antitumor PDT. TZR NSs exhibit improved therapeutic efficiency and amplified immunogenic cancer cell death (ICD) by ER targeting PDT and ER autophagy promotion. TZR NSs reversed the ITM with an increase of CD8+ T cells and reduce of immunosuppressive Foxp3 regulatory T cells, which effectively burst antitumor immunity thus clearing residual tumor cells. The ER-targeting TZR NSs developed in this paper presents a simple but valuable reference for high-efficiency tumor photodynamic immunotherapy.
Collapse
Affiliation(s)
- Pei-Ying Huang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Shi-Yu Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yun Xiang
- Department of Ultrasonography, Third Affiliated Hospital of Southern Medical University, Academy of Orthopedics, Guangzhou, Guangdong Province, 510515, P. R. China
| | - Ming-Rui Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Mei-Rong Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Li-Han Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
39
|
Chen X, Tian J, Zhao C, Wu Y, Li J, Ji Z, Lian D, Jia Z, Chen X, Zhou Z, Zhu B, Hua Z. Resveratrol, a novel inhibitor of fatty acid binding protein 5, inhibits cervical cancer metastasis by suppressing fatty acid transport into nucleus and downstream pathways. Br J Pharmacol 2024; 181:1614-1634. [PMID: 38158217 DOI: 10.1111/bph.16308] [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: 07/27/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND PURPOSE Because of cervical cancer (CC) metastasis, the prognosis of diagnosed patients is poor. However, the molecular mechanisms and therapeutic approach for metastatic CC remain elusive. EXPERIMENTAL APPROACH In this study, we first evaluated the effect of resveratrol (RSV) on CC cell migration and metastasis. Via an activity-based protein profiling (ABPP) approach, a photoaffinity probe of RSV (RSV-P) was synthesized, and the protein targets of RSV in HeLa cells were identified. Based on target information and subsequent in vivo and in vitro validation experiments, we finally elucidated the mechanism of RSV corresponding to its antimetastatic activity. KEY RESULTS The results showed that RSV concentration-dependently suppressed CC cell migration and metastasis. A list of proteins was identified as the targets of RSV, through the ABPP approach with RSV-P, among which fatty acid binding protein 5 (FABP5) attracted our attention based on The Cancer Genome Atlas (TCGA) database analysis. Subsequent knockout and overexpression experiments confirmed that RSV directly interacted with FABP5 to inhibit fatty acid transport into the nucleus, thereby suppressing downstream matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9) expression, thus inhibiting CC metastasis. CONCLUSIONS AND IMPLICATIONS Our study confirmed the key role of FABP5 in CC metastasis and provided important target information for the design of therapeutic lead compounds for metastatic CC.
Collapse
Affiliation(s)
- Xiao Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Jing Tian
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Chunyuan Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yanhui Wu
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiahuang Li
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zehan Ji
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Danchen Lian
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhibo Jia
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Xingyu Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zixin Zhou
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Bo Zhu
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zichun Hua
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
40
|
Yang P, Xu Y, Zhi X, Li R, Wang B, Liu R, Dai Z, Qian L. Photodynamically Tumor Vessel Destruction Amplified Tumor Targeting of Nanoparticles for Efficient Chemotherapy. ACS NANO 2024; 18:12933-12944. [PMID: 38712906 DOI: 10.1021/acsnano.4c00833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Efficient tumor-targeted drug delivery is still a challenging and currently unbreakable bottleneck in chemotherapy for tumors. Nanomedicines based on passive or active targeting strategy have not yet achieved convincing chemotherapeutic benefits in the clinic due to the tumor heterogeneity. Inspired by the efficient inflammatory-cell recruitment to acute clots, we constructed a two-component nanosystem, which is composed of an RGD-modified pyropheophorbide-a (Ppa) micelle (PPRM) that mediates the tumor vascular-targeted photodynamic reaction to activate local coagulation and subsequently transmits the coagulation signals to the circulating clot-targeted CREKA peptide-modified camptothecin (CPT)-loaded nanodiscs (CCNDs) for amplifying tumor targeting. PPRM could effectively bind with the tumor vasculature and induce sufficient local thrombus by a photodynamic reaction. Local photodynamic reaction-induced tumor target amplification greatly increased the tumor accumulation of CCND by 4.2 times, thus significantly enhancing the chemotherapeutic efficacy in the 4T1 breast tumor model. In other words, this study provides a powerful platform to amplify tumor-specific drug delivery by taking advantage of the efficient crosstalk between the PPRM-activated coagulation cascade and clot-targeted CCND.
Collapse
Affiliation(s)
- Peipei Yang
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing 100050, China
| | - Yunxue Xu
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Xin Zhi
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing 100050, China
| | - Rui Li
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Bo Wang
- Cancer Hospital Chinese Academy of Medical Sciences, No.17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Renfa Liu
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Linxue Qian
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing 100050, China
| |
Collapse
|
41
|
Gu L, Kong X, Li M, Chen R, Xu K, Li G, Qin Y, Wu L. Molecule engineering strategy of toll-like receptor 7/8 agonists designed for potentiating immune stimuli activation. Chem Commun (Camb) 2024; 60:5474-5485. [PMID: 38712400 DOI: 10.1039/d4cc00792a] [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: 05/08/2024]
Abstract
Toll-like receptor 7/8 (TLR-7/8) agonists serve as a promising class of pattern recognition receptors that effectively evoke the innate immune response, making them promising immunomodulatory agents for tumor immunotherapy. However, the uncontrollable administration of TLR-7/8 agonists frequently leads to the occurrence of severe immune-related adverse events (irAEs). Thus, it is imperative to strategically design tumor-microenvironment-associated biomarkers or exogenous stimuli responsive TLR-7/8 agonists in order to accurately evaluate and activate innate immune responses. No comprehensive elucidation has been documented thus far regarding TLR-7/8 immune agonists that are specifically engineered to enhance immune activation. In this feature article, we provide an overview of the advancements in TLR-7/8 agonists, aiming to enhance the comprehension of their mechanisms and promote the clinical progression through nanomedicine strategies. The current challenges and future directions of cancer immunotherapy are also discussed, with the hope that this work will inspire researchers to explore innovative applications for triggering immune responses through TLR-7/8 agonists.
Collapse
Affiliation(s)
- Liuwei Gu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Xiaojie Kong
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Mengyan Li
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Rui Chen
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Ke Xu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Guo Li
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Yulin Qin
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| | - Li Wu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, P. R. China.
| |
Collapse
|
42
|
Zhou J, Meng N, Lu L, Lu J, Wu S, Ding Y, Wu S, Bao Y, Xu Q, Chen R, Wang J, Xie C, Wu J, Lu W. A novel peptide-drug conjugate for glioma-targeted drug delivery. J Control Release 2024; 369:722-733. [PMID: 38583575 DOI: 10.1016/j.jconrel.2024.04.011] [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: 12/19/2023] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
The existence of the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) greatly limits the application of chemotherapy in glioma. To address this challenge, an optimal drug delivery system must efficiently cross the BBB/BBTB and specifically deliver therapeutic drugs into glioma cells while minimizing systemic toxicity. Here we demonstrated that glucose-regulated protein 78 (GRP78) and dopamine receptor D2 were highly expressed in patient-derived glioma tissues, and dopamine receptors were highly expressed on the BBB. Subsequently, we synthesized a novel "Y"-shaped peptide and compared the effects of different linkers on the receptor affinity and targeting ability of the peptide. A peptide-drug conjugate (pHA-AOHX-VAP-doxorubicin conjugate, pHA-AOHX-VAP-DOX) with a better affinity for glioma cells and higher solubility was derived for glioma treatment. pHA-AOHX-VAP-DOX could cross both BBB and BBTB via dopamine receptor and GRP78 receptor, and finally target glioma cells, significantly prolonging the survival time of nude mice bearing intracranial glioma. Furthermore, pHA-AOHX-VAP-DOX significantly reduced the toxicity of DOX and increased the maximum tolerated dose (MTD). Collectively, this work paves a new avenue for overcoming multiple barriers and effectively delivering chemotherapeutic agents to glioma cells while providing key evidence to identify potential receptors for glioma-targeted drug delivery.
Collapse
Affiliation(s)
- Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Nana Meng
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, and Institutes of Integrative Medicine of Fudan University, Shanghai 200040, China
| | - Jiasheng Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Yuan Ding
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Shuai Wu
- Glioma Surgery Division, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yanning Bao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Ruohan Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Jinsong Wu
- Glioma Surgery Division, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China; Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| |
Collapse
|
43
|
Zhang L, Yang J, Huang J, Yu Y, Ding J, Karges J, Xiao H. Development of tumor-evolution-targeted anticancer therapeutic nanomedicineEVT. Chem 2024; 10:1337-1356. [DOI: 10.1016/j.chempr.2023.12.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
|
44
|
Fan Y, Chen A, Zhu J, Liu R, Mei Y, Li L, Sha X, Wang X, Ren W, Wang L, Liu B. Engineered lactococcus lactis intrapleural therapy promotes regression of malignant pleural effusion by enhancing antitumor immunity. Cancer Lett 2024; 588:216777. [PMID: 38432582 DOI: 10.1016/j.canlet.2024.216777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/14/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Intrapleural immunotherapies have emerged as a prominent field in treating malignant pleural effusion (MPE). Among these, bacteria-based intrapleural therapy has exerted an anti-MPE effect by immuno-stimulating or cytotoxic properties. We previously engineered a probiotic Lactococcus lactis (FOLactis) expressing a fusion protein of Fms-like tyrosine kinase 3 and co-stimulator OX40 ligands. FOLactis activates tumor antigen-specific immune responses and displays systemic antitumor efficacy via intratumoral delivery. However, no available lesions exist in the pleural cavity of patients with MPE for intratumoral administration. Therefore, we further optimize FOLactis to treat MPE through intrapleural injection. Intrapleural administration of FOLactis (I-Pl FOLactis) not only distinctly suppresses MPE and pleural tumor nodules, but also significantly extends noticeable survival in MPE-bearing murine models. The proportion of CD103+ dendritic cells (DCs) in tumor-draining lymph nodes increases three-fold in FOLactis group, compared to the wild-type bacteria group. The enhanced DCs recruitment promotes the infiltration of effector memory T and CD8+ T cells, as well as the activation of NK cells and the polarization of macrophages to M1. Programmed death 1 blockade antibody combination further enhances the antitumor efficacy of I-Pl FOLactis. In summary, we first develop an innovative intrapleural strategy based on FOLactis, exhibiting remarkable efficacy and favorable biosafety profiles. These findings suggest prospective clinical translation of engineered probiotics for managing MPE through direct administration into the pleural cavity.
Collapse
Affiliation(s)
- Yue Fan
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China; The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing, 210008, China
| | - Aoxing Chen
- The Clinical Cancer Institute of Nanjing University, Nanjing, China; Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, China
| | - Junmeng Zhu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Rui Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China; The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yi Mei
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lin Li
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, China; Department of Pathology, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Xiaoxuan Sha
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xiaonan Wang
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China; The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing, 210008, China
| | - Wei Ren
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lifeng Wang
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China.
| | - Baorui Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China.
| |
Collapse
|
45
|
Zhang MR, Fang LL, Guo Y, Wang Q, Li YJ, Sun HF, Xie SY, Liang Y. Advancements in Stimulus-Responsive Co-Delivery Nanocarriers for Enhanced Cancer Immunotherapy. Int J Nanomedicine 2024; 19:3387-3404. [PMID: 38617801 PMCID: PMC11012697 DOI: 10.2147/ijn.s454004] [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: 12/08/2023] [Accepted: 03/14/2024] [Indexed: 04/16/2024] Open
Abstract
Cancer immunotherapy has emerged as a novel therapeutic approach against tumors, with immune checkpoint inhibitors (ICIs) making significant clinical practice. The traditional ICIs, PD-1 and PD-L1, augment the cytotoxic function of T cells through the inhibition of tumor immune evasion pathways, ultimately leading to the initiation of an antitumor immune response. However, the clinical implementation of ICIs encounters obstacles stemming from the existence of an immunosuppressive tumor microenvironment and inadequate infiltration of CD8+T cells. Considerable attention has been directed towards advancing immunogenic cell death (ICD) as a potential solution to counteract tumor cell infiltration and the immunosuppressive tumor microenvironment. This approach holds promise in transforming "cold" tumors into "hot" tumors that exhibit responsiveness to antitumor. By combining ICD with ICIs, a synergistic immune response against tumors can be achieved. However, the combination of ICD inducers and PD-1/PD-L1 inhibitors is hindered by issues such as poor targeting and uncontrolled drug release. An advantageous solution presented by stimulus-responsive nanocarrier is integrating the physicochemical properties of ICD inducers and PD-1/PD-L1 inhibitors, facilitating precise delivery to specific tissues for optimal combination therapy. Moreover, these nanocarriers leverage the distinct features of the tumor microenvironment to accomplish controlled drug release and regulate the kinetics of drug delivery. This article aims to investigate the advancement of stimulus-responsive co-delivery nanocarriers utilizing ICD and PD-1/PD-L1 inhibitors. Special focus is dedicated to exploring the advantages and recent advancements of this system in enabling the combination of ICIs and ICD inducers. The molecular mechanisms of ICD and ICIs are concisely summarized. In conclusion, we examine the potential research prospects and challenges that could greatly enhance immunotherapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Meng-Ru Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
- Department of Clinical Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Lin-Lin Fang
- RemeGen Co., Ltd, YanTai, ShanDong, 264000, People’s Republic of China
| | - Yang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Qin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - You-Jie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Hong-Fang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Shu-Yang Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| |
Collapse
|
46
|
Wang Z, Li W, Jiang Y, Tran TB, Chung J, Kim M, Scott AJ, Lu J. Camptothesome-based combination nanotherapeutic regimen for improved colorectal cancer immunochemotherapy. Biomaterials 2024; 306:122477. [PMID: 38309054 PMCID: PMC10922823 DOI: 10.1016/j.biomaterials.2024.122477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 02/05/2024]
Abstract
Camptothesome is a sphingomyelin-conjugated camptothecin (SM-CSS-CPT) nanovesicle that fortified the therapeutic delivery of CPT in diverse cancer types. To mitigate the Camptothesome-induced IDO1 negative feedback mechanism, we had co-encapsulated, indoximod (IND, IDO1 inhibitor) into Camptothesome using doxorubicin-derived IND (DOX-IND). To maximize the therapeutic potential of DOX-IND/Camptothesome, herein, we first dissected the synergistic drug ratio (DOX-IND/SM-CSS-CPT) via systematical in vitro screening. DOX-IND/Camptothesome with optimal drug ratio synchronized in vivo drug delivery with significantly higher tumor uptake compared to free drugs. This optimum DOX-IND/Camptothesome outperformed the combination of Camptothesome, Doxil and IND or other IDO1 inhibitors (BMS-986205 or epacadostat) in treating mice bearing late-stage MC38 tumors, and combination with immune checkpoint blockade (ICB) enabled it to eradicate 60 % of large tumors. Further, this optimized co-delivery Camptothesome beat Folfox and Folfiri, two first-line combination chemotherapies for colorectal cancer in antitumor efficacy and exhibited no side effects as compared to the severe systemic toxicities associated with Folfox and Folfiri. Finally, we demonstrated that the synergistic DOX-IND/Camptothesome was superior to the combined use of Onivyde + Doxil + IND in curbing the advanced orthotopic CT26-Luc tumors and eliminated 40 % tumors with complete metastasis remission when cooperated with ICB, eliciting stronger anti-CRC immune responses and greater reversal of immunosuppression. These results corroborated that with precise optimal synergistic drug ratio, the therapeutic potential of DOX-IND/Camptothesome can be fully unleased, which warrants further clinical investigation to benefit the cancer patients.
Collapse
Affiliation(s)
- Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Yanhao Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Tuyen Ba Tran
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Jinha Chung
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Minhyeok Kim
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Aaron James Scott
- Clinical and Translational Oncology Program, The University of Arizona Cancer Center, Tucson, AZ, 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, United States; Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85721, United States; Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, 85721, United States.
| |
Collapse
|
47
|
Xia Y, Li X, Bie N, Pan W, Miao YR, Yang M, Gao Y, Chen C, Liu H, Gan L, Guo AY. A method for predicting drugs that can boost the efficacy of immune checkpoint blockade. Nat Immunol 2024; 25:659-670. [PMID: 38499799 DOI: 10.1038/s41590-024-01789-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 02/13/2024] [Indexed: 03/20/2024]
Abstract
Combination therapy is a promising therapeutic strategy to enhance the efficacy of immune checkpoint blockade (ICB); however, predicting drugs for effective combination is challenging. Here we developed a general data-driven method called CM-Drug for screening compounds that can boost ICB treatment efficacy based on core and minor gene sets identified between responsive and nonresponsive samples in ICB therapy. The CM-Drug method was validated using melanoma and lung cancer mouse models, with combined therapeutic efficacy demonstrated in eight of nine predicted compounds. Among these compounds, taltirelin had the strongest synergistic effect. Mechanistic analysis and experimental verification demonstrated that taltirelin can stimulate CD8+ T cells and is mediated by the induction of thyroid-stimulating hormone. This study provides an effective and general method for predicting and evaluating drugs for combination therapy and identifies candidate compounds for future ICB combination therapy.
Collapse
Affiliation(s)
- Yun Xia
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Pan
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Ru Miao
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Yang
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Gao
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hanqing Liu
- Department of Breast and Thyroid Surgery, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
| | - An-Yuan Guo
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
48
|
Geng S, Guo P, Li X, Shi Y, Wang J, Cao M, Zhang Y, Zhang K, Li A, Song H, Zhang Z, Shi J, Liu J, Yang Y. Biomimetic Nanovehicle-Enabled Targeted Depletion of Intratumoral Fusobacterium nucleatum Synergizes with PD-L1 Blockade against Breast Cancer. ACS NANO 2024; 18:8971-8987. [PMID: 38497600 DOI: 10.1021/acsnano.3c12687] [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: 03/19/2024]
Abstract
Immune checkpoint blockade (ICB) therapy has been approved for breast cancer (BC), but clinical response rates are limited. Recent studies have shown that commensal microbes colonize a variety of tumors and are closely related to the host immune system response. Here, we demonstrated that Fusobacterium nucleatum (F.n), which is prevalent in BC, creates an immunosuppressive tumor microenvironment (ITME) characterized by a high-influx of myeloid cells that hinders ICB therapy. Administering the antibiotic metronidazole in BC can deplete F.n and remodel the ITME. To prevent an imbalance in the systemic microbiota caused by antibiotic administration, we designed a biomimetic nanovehicle for on-site antibiotic delivery inspired by F.n homing to BC. Additionally, ferritin-nanocaged doxorubicin was coloaded into this nanovehicle, as immunogenic chemotherapy has shown potential for synergy with ICB. It has been demonstrated that this biomimetic nanovehicle can be precisely homed to BC and efficiently eliminate intratumoral F.n without disrupting the diversity and abundance of systemic microbiota. This ultimately remodels the ITME, improving the therapeutic efficacy of the PD-L1 blocker with a tumor inhibition rate of over 90% and significantly extending the median survival of 4T1 tumor-bearing mice.
Collapse
Affiliation(s)
- Shizhen Geng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Pengke Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xinling Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yaru Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jing Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Mengnian Cao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yunya Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Airong Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Haiwei Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yiling Yang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
49
|
Zuo S, Liu T, Li L, Xu H, Guo J, Wang Q, Yang Y, He Z, Sun J, Sun B. Tetrasulfide bond boosts the anti-tumor efficacy of dimeric prodrug nanoassemblies. Cell Rep Med 2024; 5:101432. [PMID: 38387464 PMCID: PMC10982979 DOI: 10.1016/j.xcrm.2024.101432] [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: 08/29/2023] [Revised: 12/11/2023] [Accepted: 01/25/2024] [Indexed: 02/24/2024]
Abstract
Dimeric prodrug nanoassemblies (DPNAs) stand out as promising strategies for improving the efficiency and safety of chemotherapeutic drugs. The success of trisulfide bonds (-SSS-) in DPNAs makes polysulfide bonds a worthwhile focus. Here, we explore the comprehensive role of tetrasulfide bonds (-SSSS-) in constructing superior DPNAs. Compared to trisulfide and disulfide bonds, tetrasulfide bonds endow DPNAs with superlative self-assembly stability, prolonged blood circulation, and high tumor accumulation. Notably, the ultra-high reduction responsivity of tetrasulfide bonds make DPNAs a highly selective "tumor bomb" that can be ignited by endogenous reducing agents in tumor cells. Furthermore, we present an "add fuel to the flames" strategy to intensify the reductive stress at tumor sites by replenishing exogenous reducing agents, making considerable progress in selective tumor inhibition. This work elucidates the crucial role of tetrasulfide bonds in establishing intelligent DPNAs, alongside the combination methodology, propelling DPNAs to new heights in potent cancer therapy.
Collapse
Affiliation(s)
- Shiyi Zuo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Tian Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Lingxiao Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Hezhen Xu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Jiayu Guo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Qing Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Yinxian Yang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China.
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China.
| |
Collapse
|
50
|
Chen X, Ma Z, Yi Z, Wu E, Shang Z, Tuo B, Li T, Liu X. The effects of metabolism on the immune microenvironment in colorectal cancer. Cell Death Discov 2024; 10:118. [PMID: 38453888 PMCID: PMC10920911 DOI: 10.1038/s41420-024-01865-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Colorectal cancer (CRC) is a malignancy that is widely prevalent worldwide. Due to its unsatisfactory treatment outcome and extremely poor prognosis, many studies on the molecular mechanisms and pathological mechanisms of CRC have been published in recent years. The tumor microenvironment (TME) is an extremely important feature of tumorigenesis and one of the hallmarks of tumor development. Metabolic reprogramming is currently a hot topic in tumor research, and studies on this topic have provided important insights into CRC development. In particular, metabolic reprogramming in cancer causes changes in the composition of energy and nutrients in the TME. Furthermore, it can alter the complex crosstalk between immune cells and associated immune factors, such as associated macrophages and T cells, which play important immune roles in the TME, in turn affecting the immune escape of tumors by altering immune surveillance. In this review, we summarize several metabolism-related processes affecting the immune microenvironment of CRC tumors. Our results showed that the immune microenvironment is regulated by metabolic reprogramming and influences the development of CRC.
Collapse
Affiliation(s)
- Xingzhao Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zhiqiang Yi
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Enqin Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zhengye Shang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Dalian Road 149, Zunyi, 563000, China.
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| |
Collapse
|