1
|
Zhang S, Zhou H, Liu Y, Chen G, Li Q, Xu Y, Zheng R, Li S, Chen X, Zhao L. A stimuli-responsive immunostimulant to activate chemo-immunotherapeutic effects by inducing DNA damage and STING activation. J Colloid Interface Sci 2025; 688:664-676. [PMID: 40022787 DOI: 10.1016/j.jcis.2025.02.179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
The integration of chemotherapy with systemic immune activation represents a promising approach to eradicate metastatic tumors. In this study, a stimuli-responsive immunostimulant nanoplatform (denoted as MV@Lip) was developed to synergistically activate antitumor immunity via chemotherapy-induced DNA damage and subsequent activation of the stimulator of interferon genes (STING) signaling pathway. The MV@Lip system encapsulates the chemotherapeutic agent mitoxantrone (Mit) and the STING agonist vadimezan (Vad) within a redox-responsive liposomal carrier. MV@Lip was indicated to enhance drug stability, while simultaneously promoting efficient co-delivery of both agents into tumor cells and suppressing tumor proliferation. Mechanistically, MV@Lip exerts its therapeutic efficacy through inducing DNA damage and triggering immunogenic cell death (ICD) to enhance tumor immunogenicity by releasing damage-associated molecular patterns (DAMPs). Concurrently, the released Vadimezan could activate the STING pathway, amplifying innate immune responses through the production of proinflammatory factors and the recruitment of immune effector cells. This concerted action facilitates the infiltration and activation of natural killer (NK) cells and T lymphocytes in the tumor microenvironment, ultimately leading to the suppression of both primary and metastatic tumors. These findings provide a compelling basis for advancing chemotherapeutic combinations as immune-stimulating strategies in the treatment of metastatic malignancies.
Collapse
Affiliation(s)
- Shuiying Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Hangyu Zhou
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yixin Liu
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Guangmiao Chen
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Qiuyuan Li
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Youqin Xu
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Rongrong Zheng
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Shiying Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.
| | - Linping Zhao
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
2
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
3
|
Shaik R, Chittepu SM, Tarapatla M, Begum F, Vempati S, Royyala A. Chemoimmunotherapy synergism: mechanisms and clinical applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04125-8. [PMID: 40220027 DOI: 10.1007/s00210-025-04125-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
Chemoimmunotherapy, combining chemotherapy and immunotherapy, has emerged as a promising strategy for treating various cancers. This approach leverages the complementary mechanisms of both modalities to enhance tumor eradication. Recent advances have shed new light on the synergistic interactions between chemotherapy and immunotherapy, revealing complex mechanisms that contribute to improved clinical outcomes. Chemotherapy induces immunogenic cell death, releasing tumor antigens and damage-associated molecular patterns (DAMPs) that stimulate immune responses. It also modulates the tumor microenvironment, enhancing immune cell infiltration and reducing immunosuppressive elements. Concurrently, immunotherapy, particularly immune checkpoint inhibitors, activates the immune system to more effectively target and destroy cancer cells. Clinical evidence demonstrates significant benefits of chemoimmunotherapy in various cancers, including non-small-cell lung cancer, triple-negative breast cancer, and melanoma. Recent trials, such as KEYNOTE- 189 and IMpassion130, have shown improved overall survival and progression-free survival compared to chemotherapy alone. Emerging biomarkers, including tumor mutational burden, Programmed Death Ligand- 1 (PD-L1) expression, and immune cell infiltration patterns, are refining patient selection and response prediction. Novel strategies, such as nanoparticle-based drug delivery systems and personalized medicine approaches, are being explored to optimize chemoimmunotherapy combinations. However, challenges remain, including managing treatment-related toxicities, determining optimal dosing and sequencing, and addressing potential resistance mechanisms. Ongoing research focuses on elucidating the complex interplay between chemotherapy-induced immunomodulation and immune checkpoint inhibition to further improve treatment efficacy and patient outcomes. This review provides a comprehensive update on the mechanisms, clinical applications, and future directions of chemoimmunotherapy, highlighting its potential to revolutionize cancer treatment strategies. Clinical trial number: not applicable.
Collapse
Affiliation(s)
- Rahaman Shaik
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Sai Manasa Chittepu
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad, 501510, Telangana, India
| | - Meghana Tarapatla
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad, 501510, Telangana, India
| | - Fathima Begum
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad, 501510, Telangana, India
| | - Srujan Vempati
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad, 501510, Telangana, India
| | - Abhistika Royyala
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad, 501510, Telangana, India
| |
Collapse
|
4
|
Liu Z, Huang Y, Sun R, Li N, Xu F, Zhang W, Zhang W. Polyethylene Glycolated Shikonin-Functionalized Carbon Nanotubes as an Effective Anticancer System Combining Chemotherapy and Photothermal Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:6236-6246. [PMID: 40015981 DOI: 10.1021/acs.langmuir.4c05257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Shikonin (SHK) is a potential chemotherapy drug for the treatment of breast cancer. However, the poor water solubility, as well as the lack of tumor targeting, restricts the practical application of SHK molecules. In addition, chemotherapy alone is no longer sufficient for current clinical treatment, and it needs to be combined with other therapeutical methods. Here, using our previously developed shikonin-disulfide bond-poly(ethylene glycol) compound (referred to as SHK-SS-PEG) as a chemotherapy molecule that responds to reduced glutathione (GSH), a SHK-SS-PEG-functionalized multiwall carbon nanotubes (MWCNTs) system (SHK-SS-PEG/MWCNTs) was prepared by physical interaction. Such a system exhibits both chemotherapy and photothermal therapy for breast cancer. The physical adsorption of SHK-SS-PEG on the MWCNTs causes them to have good dispersion and storage stability for up to 3 months in aqueous solution and possess excellent photothermal conversion performance of MWCNTs. In addition, the drug release rate of SHK-SS-PEG/MWCNTs in MCF-7 cells as triggered by a near-infrared (NIR) laser was increased by 2-fold compared with that of nonirradiated cells. In vitro antitumor studies demonstrated that SHK-SS-PEG/MWCNTs had a combined effect of chemotherapy and photothermal therapy. The mortality of MCF-7 cells treated with SHK-SS-PEG/MWCNTs ([MWCNTs] = 75 μg/mL) under 808 nm NIR laser irradiation was as high as 87.78%, which was 34.39% and 28.31% higher than that of the SHK-SS-PEG/MWCNTs group without NIR laser irradiation and the MWCNTs group irradiated with NIR laser at the same equivalent concentration, respectively. These results indicated that SHK-SS-PEG/MWCNTs combined with an 808 nm NIR laser were an ideal antitumor system.
Collapse
Affiliation(s)
- Zhiyuan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yaxuan Huang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Rui Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Nan Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Fengyun Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Wei Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Wenke Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
5
|
Lei L, Song Y, Yang L, Wang Y, Xia X, Zhang Y, Zhang X, Zhang X, Duggal I, He B, Peppas NA, Cao J, Gao H. Triethylamine-mediated protonation-deprotonation unlocks dual-drug self assembly to suppress breast cancer progression and metastasis. Proc Natl Acad Sci U S A 2025; 122:e2416796122. [PMID: 39874295 PMCID: PMC11804646 DOI: 10.1073/pnas.2416796122] [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: 08/20/2024] [Accepted: 12/26/2024] [Indexed: 01/30/2025] Open
Abstract
Carrier-free nanomedicines exhibited significant potential in elevating drug efficacy and safety for tumor management, yet their self assembly typically relied on chemical modifications of drugs or the incorporation of surfactants, thereby compromising the drug's inherent pharmacological activity. To address this challenge, we proposed a triethylamine (TEA)-mediated protonation-deprotonation strategy that enabled the adjustable-proportion self assembly of dual drugs without chemical modification, achieving nearly 100% drug loading capacity. Molecular dynamic simulations, supported by experiment evidence, elucidated the underlying self-assembly mechanism. Specifically, TEA facilitated the deprotonation of Doxorubicin (Dox) and α-Tocopherol succinate (α-tos), causing Dox to transition from a hydrophilic to a hydrophobic state, while simultaneously increasing the hydrophilicity of α-tos. This allowed for a fine-tuned balance between the hydrophilic and hydrophobic properties of the two compounds, enabling their precise self assembly into a carrier-free nanomedicine (DT) with a tailored drug ratio. The engineered DT demonstrated the ability to accumulate at the tumor sites and release its therapeutic drugs in a controlled manner. The combination of Dox and α-tos synergistically generated reactive oxygen species and modulated the expression of tumor matrix metalloproteinase-9, leading to superior antitumor efficacy without significant metastasis, while maintaining excellent safety profiles. Our findings provided unique perspectives on the design of carrier-free nanomedicine for cancer therapy, thereby laying a solid foundation for its potential clinical translation.
Collapse
Affiliation(s)
- Lei Lei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| | - Yujun Song
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| | - Lianyi Yang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Yazhen Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Xue Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| | - Yiwei Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| | - Xiaoxian Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Xuequan Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Ishaan Duggal
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX78712
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Nicholas A. Peppas
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX78712
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX78712
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX78712
- Department of Surgery and Perioperative Care, Dell Medical School, Austin, TX78712
- Department of Pediatrics, Dell Medical School, Austin, TX78712
| | - Jun Cao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| |
Collapse
|
6
|
Lou H, Shang X, Wang Z, Xiang S, Qiu Y, Wang C, Yu F. Chitosan-derived drug free "artificial beacon" simulating immunogenetic cell death cascade effector to initiate immune response for cancer therapy. Int J Biol Macromol 2025; 287:138434. [PMID: 39645118 DOI: 10.1016/j.ijbiomac.2024.138434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Immunogenetic cell death (ICD) is widely participated in tumor immune therapy. However, the stress responses triggered by individual ICD inducers are typically not strong enough to effectively kickstart an ICD effect and successful ICD necessitates a high level of ICD stimulus, which may be linked to dose-related toxicity. In this research, we developed a drug-free "artificial beacon" ATP/CSO@ECM that mimics the ICD cascade system to kickstart an immune response with cationic chitosan (CSO) as a bridge, which participated in integrating tumor antigens and functional damage-associated molecular patterns (DAMPs) into one effector by electrostatic interaction. This beacon is made up of ATP/CSO nanocomplexes covered by an engineered cell membrane (ECM), which is verified to enrich with high mobility group box 1 (HMGB1) and calreticulin (CRT). When exposed to the acidic tumor environment, the ATP/CSO@ECM underwent a morphological change by proton buffering capability of CSO. This resulted in the release of simulated DAMPs and the adjuvant CSO, all of which collaborated to activate dendritic cells and ultimately prolong the effectiveness of immunotherapy. This chitosan-derived "artificial beacon" ATP/CSO@ECM fully mobilizes the function of CSO and avoids the insufficient ICD effect by concentrating signaling molecules, providing a hopeful strategy for using the ICD process in targeted cancer therapy.
Collapse
Affiliation(s)
- Haiya Lou
- Department of Diagnostic Ultrasound and Echocardiography, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Xuwei Shang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Zixu Wang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Shanshan Xiang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yihe Qiu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China.
| | - Fangying Yu
- Department of Diagnostic Ultrasound and Echocardiography, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
7
|
Hajimolaali M, Dorkoosh FA, Antimisiaris SG. Review of recent preclinical and clinical research on ligand-targeted liposomes as delivery systems in triple negative breast cancer therapy. J Liposome Res 2024; 34:671-696. [PMID: 38520185 DOI: 10.1080/08982104.2024.2325963] [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: 11/03/2023] [Revised: 02/06/2024] [Accepted: 02/27/2024] [Indexed: 03/25/2024]
Abstract
Triple-negative breast Cancer (TNBC) is one of the deadliest types, making up about 20% of all breast cancers. Chemotherapy is the traditional manner of progressed TNBC treatment; however, it has a short-term result with a high reversibility pace. The lack of targeted treatment limited and person-dependent treatment options for those suffering from TNBC cautions to be the worst type of cancer among breast cancer patients. Consequently, appropriate treatment for this disease is considered a major clinical challenge. Therefore, various treatment methods have been developed to treat TNBC, among which chemotherapy is the most common and well-known approach recently studied. Although effective methods are chemotherapies, they are often accompanied by critical limitations, especially the lack of specific functionality. These methods lead to systematic toxicity and, ultimately, the expansion of multidrug-resistant (MDR) cancer cells. Therefore, finding novel and efficient techniques to enhance the targeting of TNBC treatment is an essential requirement. Liposomes have demonstrated that they are an effective method for drug delivery; however, among a large number of liposome-based drug delivery systems annually developed, a small number have just received authorization for clinical application. The new approaches to using liposomes target their structure with various ligands to increase therapeutic efficiency and diminish undesired side effects on various body tissues. The current study describes the most recent strategies and research associated with functionalizing the liposomes' structure with different ligands as targeted drug carriers in treating TNBCs in preclinical and clinical stages.
Collapse
Affiliation(s)
- Mohammad Hajimolaali
- Department of Pharmacy, Laboratory of Pharmaceutical Technology, University of Patras, Patras, Greece
| | - Farid Abedin Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Sophia G Antimisiaris
- Department of Pharmacy, Laboratory of Pharmaceutical Technology, University of Patras, Patras, Greece
- Institute of Chemical Engineering, Foundation for Research and Technology Hellas, FORTH/ICEHT, Patras, Greece
| |
Collapse
|
8
|
Yang L, Wang Y, Ye X, Liu Q, Qu D, Chen Y. Traditional Chinese medicine-based drug delivery systems for anti-tumor therapies. Chin J Nat Med 2024; 22:1177-1192. [PMID: 39725515 DOI: 10.1016/s1875-5364(24)60746-6] [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: 08/10/2024] [Indexed: 12/28/2024]
Abstract
The treatment of tumors continues to be significantly challenging. The presence of multiple modalities, including surgery, radiation, chemotherapy and immunotherapy, the therapeutic outcomes remain limited and are often associated with adverse effects and inconsistent efficacy across cancer types. Recent studies have highlighted the potential of active components from traditional Chinese medicine (TCM) for their anti-cancer properties, which are attributable to multi-targeted mechanisms and broad pharmacological actions. Despite this potential, TCM-derived compounds are commonly limited by poor water solubility, low bioavailability, and suboptimal targeting. Currently, it is believed that advances in nanotechnology could address these limitations. Nanoparticles (NPs), which possess properties such as enhanced bioavailability, controlled release and precise targeting, have been used to improve the therapeutic efficacy of TCM components in cancer therapy. This review discusses the use of NPs for the delivery of active TCM compounds via organic-inorganic nanocarriers, highlighting innovative strategies that enhance the effectiveness of TCM-based anti-tumor components to provide insights into improving clinical outcomes while advancing the modernization and global application of TCM in oncology.
Collapse
Affiliation(s)
- Ling Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| | - Yani Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xietao Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Qiaoming Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Ding Qu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China; Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
9
|
Liu S, Guo H, Li D, Wang C. Immunologically effective biomaterials enhance immunotherapy of prostate cancer. J Mater Chem B 2024; 12:9821-9834. [PMID: 39239675 DOI: 10.1039/d3tb03044j] [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: 09/07/2024]
Abstract
Prostate cancer (PCa) is one of the most common malignant neoplasms affecting the male population. The onset of the disease is insidious and often associated with severe consequences, such as bone metastases at the time of initial diagnosis. Once it advances to metastatic castration-resistant PCa (mCRPC), conventional treatment methods become ineffective. As research on the mechanism of tumor therapy advances, immunotherapy has been evolving rapidly. However, PCa is a solid tumor type that primarily faces the challenges of poor immunogenicity and inhibitory tumor microenvironment (TME). Fortunately, the extensive use of biomaterials has led to continuous advancement in PCa immunotherapy. These innovative materials aim to address intractable issues, such as immune escape and immune desert, to inhibit tumor progression and metastasis. This detailed review focuses on the regulation of different aspects of tumor immunity by immunologically effective biomaterials, including modulating adaptive immunity, innate immunity, and the immune microenvironment, to enhance the efficacy of PCa immunotherapy. In addition, this review provides a perspective on the future prospects of immunotherapeutic nanoplatforms based on biomaterials in the treatment of PCa.
Collapse
Affiliation(s)
- Siqi Liu
- Department of Urology, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| | - Hui Guo
- Department of Urology, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| | - Di Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| | - Chunxi Wang
- Department of Urology, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| |
Collapse
|
10
|
Liang J, Tian X, Zhou M, Yan F, Fan J, Qin Y, Chen B, Huo X, Yu Z, Tian Y, Deng S, Peng Y, Wang Y, Liu B, Ma X. Shikonin and chitosan-silver nanoparticles synergize against triple-negative breast cancer through RIPK3-triggered necroptotic immunogenic cell death. Biomaterials 2024; 309:122608. [PMID: 38744189 DOI: 10.1016/j.biomaterials.2024.122608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/21/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Necroptotic immunogenic cell death (ICD) can activate the human immune system to treat the metastasis and recurrence of triple-negative breast cancer (TNBC). However, developing the necroptotic inducer and precisely delivering it to the tumor site is the key issue. Herein, we reported that the combination of shikonin (SHK) and chitosan silver nanoparticles (Chi-Ag NPs) effectively induced ICD by triggering necroptosis in 4T1 cells. Moreover, to address the lack of selectivity of drugs for in vivo application, we developed an MUC1 aptamer-targeted nanocomplex (MUC1@Chi-Ag@CPB@SHK, abbreviated as MUC1@ACS) for co-delivering SHK and Chi-Ag NPs. The accumulation of MUC1@ACS NPs at the tumor site showed a 6.02-fold increase compared to the free drug. Subsequently, upon reaching the tumor site, the acid-responsive release of SHK and Chi-Ag NPs from MUC1@ACS NPs cooperatively induced necroptosis in tumor cells by upregulating the expression of RIPK3, p-RIPK3, and tetrameric MLKL, thereby effectively triggering ICD. The sequential maturation of dendritic cells (DCs) subsequently enhanced the infiltration of CD8+ and CD4+ T cells in tumors, while inhibiting regulatory T cells (Treg cells), resulting in the effective treatment of primary and distal tumor growth and the inhibition of TNBC metastasis. This work highlights the importance of nanoparticles in mediating drug interactions during necroptotic ICD.
Collapse
Affiliation(s)
- Jiahao Liang
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xiangge Tian
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Meirong Zhou
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Fei Yan
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jialong Fan
- College of Biology, Hunan University, Changsha, China
| | - Yan Qin
- College of Biology, Hunan University, Changsha, China
| | - Binlong Chen
- College of Biology, Hunan University, Changsha, China
| | - Xiaokui Huo
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhenlong Yu
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Yan Tian
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Sa Deng
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yulin Peng
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yan Wang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, China.
| | - Xiaochi Ma
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
11
|
Zhang LL, Zhang DJ, Shi JX, Huang MY, Yu JM, Chen XJ, Wei X, Zou L, Lu JJ. Immunogenic cell death inducers for cancer therapy: An emerging focus on natural products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155828. [PMID: 38905847 DOI: 10.1016/j.phymed.2024.155828] [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: 04/01/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Immunogenic cell death (ICD) is a specific form of regulated cell death induced by a variety of stressors. During ICD, the dying cancer cells release damage-associated molecular patterns (DAMPs), which promote dendritic cell maturation and tumor antigen presentation, subsequently triggering a T-cell-mediated anti-tumor immune response. In recent years, a growing number of studies have demonstrated the potential of natural products to induce ICD and enhance tumor cell immunogenicity. Moreover, there is an increasing interest in identifying new ICD inducers from natural products. PURPOSE This study aimed to emphasize the potential of natural products and their derivatives as ICD inducers to promote research on using natural products in cancer therapy and provide ideas for future novel immunotherapies based on ICD induction. METHOD This review included a thorough search of the PubMed, Web of Science, Scopus, and Google Scholar databases to identify natural products with ICD-inducing capabilities. A comprehensive search for clinical trials on natural ICD inducers was also conducted using ClinicalTrials.gov, as well as the approved patents using the Espacenet and CNKI Patent Database. RESULTS Natural compounds that induce ICD can be categorized into several groups, such as polyphenols, flavonoids, terpenoids, and alkaloids. Natural products can induce the release of DAMPs by triggering endoplasmic reticulum stress, activation of autophagy-related pathways, and reactive oxygen species generation, etc. Ultimately, they activate anti-tumor immune response and improve the efficacy of cancer treatments. CONCLUSION A growing number of ICD inducers from natural products with promising anti-cancer potential have been identified. The detailed information presented in this review will contribute to the further development of natural ICD inducers and cancer treatment strategies based on ICD-induced responses.
Collapse
Affiliation(s)
- Le-Le Zhang
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 442000, China
| | - Du-Juan Zhang
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jia-Xin Shi
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Jia-Mei Yu
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xu-Jia Chen
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xiao Wei
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
12
|
Yang LJ, Han T, Liu RN, Shi SM, Luan SY, Meng SN. Plant-derived natural compounds: A new frontier in inducing immunogenic cell death for cancer treatment. Biomed Pharmacother 2024; 177:117099. [PMID: 38981240 DOI: 10.1016/j.biopha.2024.117099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
Immunogenic cell death (ICD) can activate adaptive immune response in the host with normal immune system. Some synthetic chemotherapeutic drugs and natural compounds have shown promising results in cancer treatment by triggering the release of damage-associated molecules (DAMPs) to trigger ICD. However, most chemotherapeutic drugs exhibit non-selective cytotoxicity and may also induce and promote metastasis, thereby significantly reducing their clinical efficacy. Among the natural compounds that can induce ICD, plant-derived compounds account for the largest proportion, which are of increasing value in the treatment of cancer. Understanding which plant-derived natural compounds can induce ICD and how they induce ICD is crucial for developing strategies to improve chemotherapy outcomes. In this review, we focus on the recent findings regarding plant-derived natural compounds that induce ICD according to the classification of flavonoids, alkaloids, glycosides, terpenoids and discuss the potential mechanisms including endoplasmic reticulum (ER) stress, DNA damage, apoptosis, necroptosis autophagy, ferroptosis. In addition, plant-derived natural compounds that can enhance the ICD induction ability of conventional therapies for cancer treatment is also elaborated. The rational use of plant-derived natural compounds to induce ICD is helpful for the development of new cancer treatment methods.
Collapse
Affiliation(s)
- Li-Juan Yang
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Ting Han
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Ruo-Nan Liu
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Shu-Ming Shi
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Shi-Yun Luan
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Sheng-Nan Meng
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
13
|
Zhao M, Zhu X, Li B, Yan C, Wu C, He L, Cao J, Lu F, Chen H, Li W. Potent cancer therapy by liposome microstructure tailoring with active-to-passive targeting and shell-to-core thermosensitive features. Mater Today Bio 2024; 26:101035. [PMID: 38586871 PMCID: PMC10995888 DOI: 10.1016/j.mtbio.2024.101035] [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: 11/29/2023] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024] Open
Abstract
Liposomes have been widely studied as drug carriers for clinical application, and the key issue is how to achieve effective delivery through targeting strategies. Even though certain cell-level targeting or EPR effect designs have been developed, reaching sufficient drug concentration in intracellular regions remains a challenge due to the singularity of functionality. Herein, benefiting from the unique features of tumor from tissue to cell, a dual-thermosensitive and dual-targeting liposome (DTSL) was creatively fabricated through fine microstructure tailoring, which holds intelligent both tissue-regulated active-to-passive binding and membrane-derived homologous-fusion (HF) properties. At the micro level, DTSL can actively capture tumor cells and accompany the enhanced HF effect stimulated by self-constriction, which achieves a synergistic promotion effect targeting tissues to cells. As a result, this first active-then passive targeting process makes drug delivery more accurate and effective, and after dynamic targeting into cells, the nucleus of DTSL undergoes further thermally responsive contraction, fully releasing internal drugs. In vivo experiments showed that liposomes with dual targeting and dual thermosensitive features almost completely inhibited tumor growth. Summarized, these results provide a reference for a rational design and microstructural tailoring of the liposomal co-delivery system of drugs, suggesting that active-to-passive dual-targeting DTSL can function as a new strategy for cancer treatment.
Collapse
Affiliation(s)
- Mengxin Zhao
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| | - Xiaodong Zhu
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| | - Bailing Li
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chenyang Yan
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| | - Cong Wu
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| | - Lei He
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jingyi Cao
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Fanglin Lu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Han Chen
- Department of General Surgery, 905th Hospital of People's Liberation Army Navy, Naval Medical University, Shanghai, 200433, China
| | - Wei Li
- Department of Nanomedicine & Shanghai Key Lab of Cell Engineering, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
14
|
Elzoghby AO, Samir O, Emam HE, Soliman A, Abdelgalil RM, Elmorshedy YM, Elkhodairy KA, Nasr ML. Engineering nanomedicines for immunogenic eradication of cancer cells: Recent trends and synergistic approaches. Acta Pharm Sin B 2024; 14:2475-2504. [PMID: 38828160 PMCID: PMC11143780 DOI: 10.1016/j.apsb.2024.03.022] [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: 08/01/2023] [Revised: 02/07/2024] [Accepted: 03/09/2024] [Indexed: 06/05/2024] Open
Abstract
Resistance to cancer immunotherapy is mainly attributed to poor tumor immunogenicity as well as the immunosuppressive tumor microenvironment (TME) leading to failure of immune response. Numerous therapeutic strategies including chemotherapy, radiotherapy, photodynamic, photothermal, magnetic, chemodynamic, sonodynamic and oncolytic therapy, have been developed to induce immunogenic cell death (ICD) of cancer cells and thereby elicit immunogenicity and boost the antitumor immune response. However, many challenges hamper the clinical application of ICD inducers resulting in modest immunogenic response. Here, we outline the current state of using nanomedicines for boosting ICD of cancer cells. Moreover, synergistic approaches used in combination with ICD inducing nanomedicines for remodeling the TME via targeting immune checkpoints, phagocytosis, macrophage polarization, tumor hypoxia, autophagy and stromal modulation to enhance immunogenicity of dying cancer cells were analyzed. We further highlight the emerging trends of using nanomaterials for triggering amplified ICD-mediated antitumor immune responses. Endoplasmic reticulum localized ICD, focused ultrasound hyperthermia, cell membrane camouflaged nanomedicines, amplified reactive oxygen species (ROS) generation, metallo-immunotherapy, ion modulators and engineered bacteria are among the most innovative approaches. Various challenges, merits and demerits of ICD inducer nanomedicines were also discussed with shedding light on the future role of this technology in improving the outcomes of cancer immunotherapy.
Collapse
Affiliation(s)
- Ahmed O. Elzoghby
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Omar Samir
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Hagar E. Emam
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Ahmed Soliman
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Riham M. Abdelgalil
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Yomna M. Elmorshedy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Kadria A. Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mahmoud L. Nasr
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| |
Collapse
|
15
|
Qian Z, Zhang Z, Cen L, Ke Y, Shao J, Tian M, Liu B. Mycobacterium smegmatis enhances shikonin-induced immunogenic cell death-an efficient in situ tumor vaccine strategy. J Biomed Res 2024; 38:369-381. [PMID: 38807377 PMCID: PMC11300524 DOI: 10.7555/jbr.38.20240049] [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: 02/25/2024] [Revised: 04/17/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
Tumor vaccines are a promising avenue in cancer immunotherapy. Despite the progress in targeting specific immune epitopes, tumor cells lacking these epitopes can evade the treatment. Here, we aimed to construct an efficient in situ tumor vaccine called Vac-SM, utilizing shikonin (SKN) to induce immunogenic cell death (ICD) and Mycobacterium smegmatis as an immune adjuvant to enhance in situ tumor vaccine efficacy. SKN showed a dose-dependent and time-dependent cytotoxic effect on the tumor cell line and induced ICD in tumor cells as evidenced by the CCK-8 assay and the detection of the expression of relevant indicators, respectively. Compared with the control group, the in situ Vac-SM injection in mouse subcutaneous metastatic tumors significantly inhibited tumor growth and distant tumor metastasis, while also improving survival rates. Mycobacterium smegmatis effectively induced maturation and activation of bone marrow-derived dendritic cells (DCs), and in vivo tumor-draining lymph nodes showed an increased maturation of DCs and a higher proportion of effector memory T-cell subsets with the Vac-SM treatment, based on flow cytometry analysis results. Collectively, the Vac-SM vaccine effectively induces ICD, improves antigen presentation by DCs, activates a specific systemic antitumor T-cell immune response, exhibits a favorable safety profile, and holds the promise for clinical translation for local tumor immunotherapy.
Collapse
Affiliation(s)
- Zhaoye Qian
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210008, China
- Department of Oncology, the Affiliated Huai'an No. 1 People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223000, China
| | - Zhe Zhang
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Lanqi Cen
- Department of Oncology, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, China
| | - Yaohua Ke
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Jie Shao
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Manman Tian
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210008, China
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| |
Collapse
|
16
|
Shang Q, Liu W, Leslie F, Yang J, Guo M, Sun M, Zhang G, Zhang Q, Wang F. Nano-formulated delivery of active ingredients from traditional Chinese herbal medicines for cancer immunotherapy. Acta Pharm Sin B 2024; 14:1525-1541. [PMID: 38572106 PMCID: PMC10985040 DOI: 10.1016/j.apsb.2023.12.008] [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: 09/24/2023] [Revised: 11/15/2023] [Accepted: 12/12/2023] [Indexed: 04/05/2024] Open
Abstract
Cancer immunotherapy has garnered promise in tumor progression, invasion, and metastasis through establishing durable and memorable immunological activity. However, low response rates, adverse side effects, and high costs compromise the additional benefits for patients treated with current chemical and biological agents. Chinese herbal medicines (CHMs) are a potential treasure trove of natural medicines and are gaining momentum in cancer immunomodulation with multi-component, multi-target, and multi-pathway characteristics. The active ingredient extracted from CHMs benefit generalized patients through modulating immune response mechanisms. Additionally, the introduction of nanotechnology has greatly improved the pharmacological qualities of active ingredients through increasing the hydrophilicity, stability, permeability, and targeting characteristics, further enhancing anti-cancer immunity. In this review, we summarize the mechanism of active ingredients for cancer immunomodulation, highlight nano-formulated deliveries of active ingredients for cancer immunotherapy, and provide insights into the future applications in the emerging field of nano-formulated active ingredients of CHMs.
Collapse
Affiliation(s)
- Qi Shang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wandong Liu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou 310053, China
| | - Faith Leslie
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jiapei Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingmei Guo
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingjiao Sun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou 310053, China
- Traditional Chinese Medicine “Preventing Disease” Wisdom Health Project Research Center of Zhejiang, Hangzhou 310053, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Feihu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
17
|
Angulo-Elizari E, Henriquez-Figuereo A, Morán-Serradilla C, Plano D, Sanmartín C. Unlocking the potential of 1,4-naphthoquinones: A comprehensive review of their anticancer properties. Eur J Med Chem 2024; 268:116249. [PMID: 38458106 DOI: 10.1016/j.ejmech.2024.116249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/10/2024]
Abstract
Cancer encompasses a group of pathologies with common characteristics, high incidence, and prevalence in all countries. Although there are treatments available for this disease, they are not always effective or safe, often failing to achieve the desired results. This is why it is necessary to continue the search for new therapies. One of the strategies for obtaining new antitumor drugs is the use of 1,4-naphthoquinone as a scaffold in synthetic or natural products with antitumor activity. This review focuses on compiling studies related to the antitumor activity of 1,4-naphthoquinone and its natural and synthetic derivatives over the last 10 years. The work describes the main natural naphthoquinones with antitumor activity and classifies the synthetic naphthoquinones based on the structural modifications made to the scaffold. Additionally, the formation of metal complexes using naphthoquinones as a ligand is considered. After a thorough review, 197 synthetic compounds with potent biological activity against cancer have been classified according to their chemical structures and their mechanisms of action have been described.
Collapse
Affiliation(s)
- Eduardo Angulo-Elizari
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Andreina Henriquez-Figuereo
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Cristina Morán-Serradilla
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Daniel Plano
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain; Navarra Institute for Health Research (IdisNA), 31008, Pamplona, Spain.
| | - Carmen Sanmartín
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain; Navarra Institute for Health Research (IdisNA), 31008, Pamplona, Spain.
| |
Collapse
|
18
|
Paun RA, Jurchuk S, Tabrizian M. A landscape of recent advances in lipid nanoparticles and their translational potential for the treatment of solid tumors. Bioeng Transl Med 2024; 9:e10601. [PMID: 38435821 PMCID: PMC10905562 DOI: 10.1002/btm2.10601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 03/05/2024] Open
Abstract
Lipid nanoparticles (LNPs) are biocompatible drug delivery systems that have found numerous applications in medicine. Their versatile nature enables the encapsulation and targeting of various types of medically relevant molecular cargo, including oligonucleotides, proteins, and small molecules for the treatment of diseases, such as cancer. Cancers that form solid tumors are particularly relevant for LNP-based therapeutics due to the enhanced permeation and retention effect that allows nanoparticles to accumulate within the tumor tissue. Additionally, LNPs can be formulated for both locoregional and systemic delivery depending on the tumor type and stage. To date, LNPs have been used extensively in the clinic to reduce systemic toxicity and improve outcomes in cancer patients by encapsulating chemotherapeutic drugs. Next-generation lipid nanoparticles are currently being developed to expand their use in gene therapy and immunotherapy, as well as to enable the co-encapsulation of multiple drugs in a single system. Other developments include the design of targeted LNPs to specific cells and tissues, and triggerable release systems to control cargo delivery at the tumor site. This review paper highlights recent developments in LNP drug delivery formulations and focuses on the treatment of solid tumors, while also discussing some of their current translational limitations and potential opportunities in the field.
Collapse
Affiliation(s)
- Radu A. Paun
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Sarah Jurchuk
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Faculty of Dentistry and Oral Health SciencesMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
19
|
Xue P, Li J, Song J, Yu J, Liu H, Jiang Y, Wang Y. Fe 3+ mediated shikonin and PPA coloaded liposomes induce robust immunogenic cell death by integrating ROS enhancement and GSH depletion. Int J Pharm 2024; 649:123657. [PMID: 38040398 DOI: 10.1016/j.ijpharm.2023.123657] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
Reactive oxygen species (ROS) can not only induce cellular oxidative stress, but also trigger antitumor immune response. However, single ROS generated therapy is usually not enough to induce efficient antitumor immune response. Furthermore, the adaptive antioxidant mechanisms coupled with overexpressed ROS can also decrease the antitumor capacity of ROS therapy. To circumvent this problem, we designed a synergistic strategy for inducing robust ROS based ICD effect by constructing a coloaded liposomes (PPA, Pyropheophorbide-alpha and SHK, shikonin) with Fe3+ gradient to simultaneously enhance ROS mediated oxidative stress and glutathione depletion. Interestingly, the coloaded liposome possesses an acid/GSH dual triggered release profile. More importantly, with the help of depleting GSH, LipoPS (coloaded liposome of SHK and PPA) can excite robust ROS and demonstrate synergistic antitumor efficacy with amplified ICD effect. Summarized, the established coloaded liposome LipoPS exhibits good therapeutic security and synergistic antitumor effect with strong antitumor immune activation, providing potential for further development.
Collapse
Affiliation(s)
- Peng Xue
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Jia Song
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China
| | - Yiguo Jiang
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215153, China.
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
20
|
Xu W, Liu W, Yang J, Lu J, Zhang H, Ye D. Stimuli-responsive nanodelivery systems for amplifying immunogenic cell death in cancer immunotherapy. Immunol Rev 2024; 321:181-198. [PMID: 37403660 DOI: 10.1111/imr.13237] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/06/2023]
Abstract
Immunogenic cell death (ICD) is a special pattern of tumor cell death, enabling to elicit tumor-specific immune response via the release of damage-associated molecular patterns and tumor-associated antigens in the tumor microenvironment. ICD-induced immunotherapy holds the promise for completely eliminating tumors and long-term protective antitumor immune response. Increasing ICD inducers have been discovered for boosting antitumor immunity via evoking ICD. Nonetheless, the utilization of ICD inducers remains insufficient owing to serious toxic reactions, low localization efficiency within the tumor microenvironmental niche, etc. For overcoming such limitations, stimuli-responsive multifunctional nanoparticles or nanocomposites with ICD inducers have been developed for improving immunotherapeutic efficiency via lowering toxicity, which represent a prospective scheme for fostering the utilization of ICD inducers in immunotherapy. This review outlines the advances in near-infrared (NIR)-, pH-, redox-, pH- and redox-, or NIR- and tumor microenvironment-responsive nanodelivery systems for ICD induction. Furthermore, we discuss their clinical translational potential. The progress of stimuli-responsive nanoparticles in clinical settings depends upon the development of biologically safer drugs tailored to patient needs. Moreover, an in-depth comprehending of ICD biomarkers, immunosuppressive microenvironment, and ICD inducers may accelerate the advance in smarter multifunctional nanodelivery systems to further amplify ICD.
Collapse
Affiliation(s)
- Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Wangrui Liu
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfeng Yang
- Department of Surgery, ShangNan Branch of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiahe Lu
- Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| |
Collapse
|
21
|
Pang G, Wei S, Zhao J, Wang FJ. Improving nanochemoimmunotherapy efficacy by boosting "eat-me" signaling and downregulating "don't-eat-me" signaling with Ganoderma lucidum polysaccharide-based drug delivery. J Mater Chem B 2023; 11:11562-11577. [PMID: 37982298 DOI: 10.1039/d3tb02118a] [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: 11/21/2023]
Abstract
To address the challenges posed by low immunogenicity and immune checkpoints during cancer treatment, we propose an alternative strategy that combines immunogenic cell death (ICD) effects with CD47/SIRPα blockade to reactivate phagocytosis of tumor cells by macrophages with polysaccharide-based drug delivery. In this study, the EGFR inhibitor gefitinib was identified as a novel CD47 modulator, which promoted the translocation of CD47 molecules from the cell membrane to endosomes through the EGFR-Rab5 pathway, leading to reduced cell surface CD47 levels and limiting interaction with SIRPα. Based on this finding, we developed prophagocytic mixed nanodrugs to enhance macrophage phagocytosis by encapsulating ICD inducer doxorubicin and CD47 inhibitor gefitinib with immunostimulatory polysaccharides from Ganoderma lucidum. This approach downregulated cell surface CD47 expression to attenuate "don't-eat-me" signaling, while increasing doxorubicin accumulation in tumors by inhibiting drug-resistance proteins, leading to more exposure of calreticulin and amplifying the "eat-me" signaling. In vivo experiments demonstrated that this approach significantly suppressed intraperitoneal tumor dissemination, reversed doxorubicin-induced weight loss, and effectively induced macrophage polarization, dendritic cell maturation, and CD8+ T cell activation. These findings highlighted the significant potential of our macrophage-centered therapeutic strategy using polysaccharide-based nanocarriers and provided new perspectives for chemoimmunotherapy.
Collapse
Affiliation(s)
- Guibin Pang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China.
| | - Siqi Wei
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China.
| | - Jian Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China.
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Fu-Jun Wang
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang 322100, Zhejiang, P. R. China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, P. R. China.
| |
Collapse
|
22
|
Gao Y, Zhang Y, Xia H, Ren Y, Zhang H, Huang S, Li M, Wang Y, Li H, Liu H. Biomimetic virus-like mesoporous silica nanoparticles improved cellular internalization for co-delivery of antigen and agonist to enhance Tumor immunotherapy. Drug Deliv 2023; 30:2183814. [PMID: 36843529 PMCID: PMC9980018 DOI: 10.1080/10717544.2023.2183814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
Nanocarrier antigen-drug delivery system interacts specifically with immune cells and provides intelligent delivery modes to improve antigen delivery efficiency and facilitate immune progression. However, these nanoparticles often have weak adhesion to cells, followed by insufficient cell absorption, leading to a failed immune response. Inspired by the structure and function of viruses, virus-like mesoporous silica nanoparticles (VMSNs) were prepared by simulating the surface structure, centripetal-radialized spike structure and rough surface topology of the virus and co-acted with the toll-like receptor 7/8 agonist imiquimod (IMQ) and antigens oocyte albumin (OVA). Compared to the conventional spherical mesoporous silica nanoparticles (MSNs), VMSNs which was proven to be biocompatible in both cellular and in vivo level, had higher cell invasion ability and unique endocytosis pathway that was released from lysosomes and promoted antigen cross-expression. Furthermore, VMSNs effectively inhibited B16-OVA tumor growth by activating DCs maturation and increasing the proportion of CD8+ T cells. This work demonstrated that virus-like mesoporous silica nanoparticles co-supply OVA and IMQ, could induce potent tumor immune responses and inhibit tumor growth as a consequence of the surface spike structure induces a robust cellular immune response, and undoubtedly provided a good basis for further optimizing the nanovaccine delivery system.
Collapse
Affiliation(s)
- Yuan Gao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yingxi Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hong Xia
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yuqing Ren
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Haibin Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Siwen Huang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Meiju Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Heran Li
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China,CONTACT Heran Li School of Pharmacy, China Medical University, 77 Puhe Road, Shenyang North New Area, Liaoning, 110122, China; Hongzhuo Liu Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| |
Collapse
|
23
|
Wang P, Lei M, Weng G, Huang R, Lin H, Wei W, Chen Y, He H, Chen P, Zhang D, Chen W, Zhou H, Gao P, Liu S, Wang F. High-dose chemotherapy sensitizes locally advanced esophageal squamous cell carcinoma to PD-1 blockade for a higher pathological complete response rate and survival. Transl Oncol 2023; 36:101736. [PMID: 37478670 PMCID: PMC10375848 DOI: 10.1016/j.tranon.2023.101736] [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: 04/25/2023] [Revised: 06/24/2023] [Accepted: 07/08/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND PD-1 inhibitor and chemotherapy demonstrated durable antitumor activity with a manageable safety profile as the first-line treatment in patients with advanced esophageal squamous cell carcinoma (ESCC). The present study aimed to evaluate the efficacy of PD-1 inhibitors plus different dose intensity neoadjuvant chemotherapy in the treatment of locally advanced ESCC. METHODS Patients with locally advanced but resectable thoracic ESCC, staged as T3 or T4a, N0-3, and M0 or M1 lymph node metastasis (confined to the supraclavicular lymph nodes), were enrolled in this study. The eligible patients received tislelizumab plus different dose intensity chemotherapy for a 21-day cycle with repeated 2-4 cycles before surgery. The primary endpoints are pathological complete response (pCR) and major pathological response (MPR), and the secondary endpoints are objective response rate (ORR), disease control rate (DCR), and disease-free survival (DFS). RESULTS From November 2019 to February 2022, 122 cases received at least two cycles neoadjuvant chemoimmunotherapy and were evaluated by imaging examination. Subsequently, 99 patients underwent surgery and were evaluated by pathological evaluation. According to chemotherapy dose intensity, the patients were divided into three cohorts: cohort 1 (<80% dose intensity), cohort 2 (80-90% dose intensity), cohort 3 (90-100% dose intensity). All surgery patients underwent minimally invasive esophagectomy (MIE). The average pCR was identified in 22.22%; 16% had pCR in cohort 1, 17.65% had pCR in cohort 2, and 30.00% had pCR in cohort 3. MPR was observed in 9 (36.00%) patients in cohort 1, 18 (52.94%) patients in cohort 2, 22 (55.00%) patients in cohort 3. In univariable and multivariable analysis, dose intensity was significantly associated with MPR (p = 0.048) in patients who underwent esophagectomy. For surviving patients, the median follow-up was 13.76 months after esophagectomy. Compared to cohort 1, cohorts 2 and 3 had better DFS (p = 0.056). In addition, the prognosis of patients with MPR was better than that of patients without MPR (p = 0.014). CONCLUSIONS The robust antitumor activity of neoadjuvant chemoimmunotherapy for locally advanced but resectable thoracic ESCC was confirmed. More than 80% of chemotherapy dose intensity combined with immunotherapy resulted in a high pCR rate and prolonged DFS.
Collapse
Affiliation(s)
- Peiyuan Wang
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, China; Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, China
| | - Mengxia Lei
- Department of Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Guibin Weng
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Rongfang Huang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hui Lin
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Wenwei Wei
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yujie Chen
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hao He
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Peng Chen
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Derong Zhang
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Weijie Chen
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hang Zhou
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Pengqiang Gao
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Shuoyan Liu
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| | - Feng Wang
- Department of Thoracic Oncology Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| |
Collapse
|
24
|
Amiri M, Molavi O, Sabetkam S, Jafari S, Montazersaheb S. Stimulators of immunogenic cell death for cancer therapy: focusing on natural compounds. Cancer Cell Int 2023; 23:200. [PMID: 37705051 PMCID: PMC10500939 DOI: 10.1186/s12935-023-03058-7] [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: 04/05/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023] Open
Abstract
A growing body of evidence indicates that the anticancer effect of the immune system can be activated by the immunogenic modulation of dying cancer cells. Cancer cell death, as a result of the activation of an immunomodulatory response, is called immunogenic cell death (ICD). This regulated cell death occurs because of increased immunogenicity of cancer cells undergoing ICD. ICD plays a crucial role in stimulating immune system activity in cancer therapy. ICD can therefore be an innovative route to improve anticancer immune responses associated with releasing damage-associated molecular patterns (DAMPs). Several conventional and chemotherapeutics, as well as preclinically investigated compounds from natural sources, possess immunostimulatory properties by ICD induction. Natural compounds have gained much interest in cancer therapy owing to their low toxicity, low cost, and inhibiting cancer cells by interfering with different mechanisms, which are critical in cancer progression. Therefore, identifying natural compounds with ICD-inducing potency presents agents with promising potential in cancer immunotherapy. Naturally derived compounds are believed to act as immunoadjuvants because they elicit cancer stress responses and DAMPs. Acute exposure to DAMP molecules can activate antigen-presenting cells (APCs), such as dendritic cells (DCs), which leads to downstream events by cytotoxic T lymphocytes (CTLs) and natural killer cells (NKs). Natural compounds as inducers of ICD may be an interesting approach to ICD induction; however, parameters that determine whether a compound can be used as an ICD inducer should be elucidated. Here, we aimed to discuss the impact of multiple ICD inducers, mainly focusing on natural agents, including plant-derived, marine molecules, and bacterial-based compounds, on the release of DAMP molecules and the activation of the corresponding signaling cascades triggering immune responses. In addition, the potential of synthetic agents for triggering ICD is also discussed.
Collapse
Affiliation(s)
- Mina Amiri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahnaz Sabetkam
- Department of Anatomy, Faculty of Medicine, university of Kyrenia, Kyrenia, Northern Cyprus
- Department of Anatomy and histopathology, Faculty of medicine, Tabriz medical sciences, Islamic Azad University, Tabriz, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
25
|
Chen L, Zhao D, Ren X, Ren J, Meng X, Fu C, Li X. Shikonin-Loaded Hollow Fe-MOF Nanoparticles for Enhanced Microwave Thermal Therapy. ACS Biomater Sci Eng 2023; 9:5405-5417. [PMID: 37638660 PMCID: PMC10498989 DOI: 10.1021/acsbiomaterials.3c00644] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/15/2023] [Indexed: 08/29/2023]
Abstract
Microwave (MW) thermal therapy has been widely used for the treatment of cancer in clinics, but it still shows limited efficacy and a high recurrence rate owing to non-selective heat delivery and thermo-resistance. Regulating glycolysis shows great promise to improve MW thermal therapy since glycolysis plays an important role in thermo-resistance, progression, metabolism, and recurrence. Herein, we developed a delivery nanosystem of shikonin (SK)-loaded and hyaluronic acid (HA)-modified hollow Fe-MOF (HFM), HFM@SK@HA, as an efficient glycolysis-meditated agent to improve the efficacy of MW thermal therapy. The HFM@SK@HA nanosystem shows a high SK loading capacity of 31.7 wt %. The loaded SK can be effectively released from the HFM@SK@HA under the stimulation of an acidic tumor microenvironment and MW irradiation, overcoming the intrinsically low solubility and severe toxicity of SK. We also find that the HFM@SK@HA can not only greatly improve the heating effect of MW in the tumor site but also mediate MW-enhancing dynamic therapy efficiency by catalyzing the endogenous H2O2 to generate reactive oxygen species (ROS). As such, the MW irradiation treatment in the presence of HFM@SK@HA in vitro enables a highly improved anti-tumor efficacy due to the combined effect of released SK and generated ROS on inhibiting glycolysis in cancer cells. Our in vivo experiments show that the tumor inhibition rate is up to 94.75% ± 3.63% with no obvious recurrence during the 2 weeks after treatment. This work provides a new strategy for improving the efficacy of MW thermal therapy.
Collapse
Affiliation(s)
- Lufeng Chen
- Department
of Radiation Oncology, First Clinical Medical
School and First Hospital of Shanxi Medical University, No.85 Jiefang Road, Taiyuan City 030001, PR China
| | - Dongming Zhao
- Department
of Radiation Oncology, First Clinical Medical
School and First Hospital of Shanxi Medical University, No.85 Jiefang Road, Taiyuan City 030001, PR China
- Department
of Pathology, Basic Medical School, Shanxi
Medical University, No.56 Xinjian Road, Taiyuan City 030001, PR China
| | - Xiangling Ren
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Jun Ren
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xianwei Meng
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Changhui Fu
- Laboratory
of Controllable Preparation and Application of Nanomaterials, Technical
Institute of Physics and Chemistry, Chinese
Academy of Sciences, No.29 East Road Zhongguancun, Beijing 100190, PR China
- CAS
Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xianfeng Li
- Department
of Radiation Oncology, First Clinical Medical
School and First Hospital of Shanxi Medical University, No.85 Jiefang Road, Taiyuan City 030001, PR China
- Department
of Pathology, Basic Medical School, Shanxi
Medical University, No.56 Xinjian Road, Taiyuan City 030001, PR China
| |
Collapse
|
26
|
Tang L, Yin Y, Cao Y, Fu C, Liu H, Feng J, Wang W, Liang XJ. Extracellular Vesicles-Derived Hybrid Nanoplatforms for Amplified CD47 Blockade-Based Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303835. [PMID: 37384818 DOI: 10.1002/adma.202303835] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Immunomodulation of tumor-associated macrophages (TAMs) into tumor-inhibiting M1-like phenotype is a promising but challenging strategy. Cleverly, tumor cells overexpress CD47, a "don't eat me" signal that ligates with the signal regulatory protein alpha (SIRPα) on macrophages to escape phagocytosis. Thus, effective re-education of TAMs into the "eat me" type and blocking the CD47-SIRPα signaling play pivotal roles in tumor immunotherapy. Herein, it is reported that hybrid nanovesicles (hEL-RS17) derived from extracellular vesicles of M1 macrophages and decorated with RS17 peptide, an antitumor peptide that specifically binds to CD47 on tumor cells and blocks CD47-SIRPα signaling, can actively target tumor cells and remodel TAM phenotypes. Consequently, more M1-like TAMs infiltrate into tumor tissue to phagocytize more tumor cells due to CD47 blockade. By further co-encapsulating chemotherapeutic agent shikonin, photosensitizer IR820, and immunomodulator polymetformin in hEL-RS17, an enhanced antitumor effect is obtained due to the combinational treatment modality and close synergy among each component. Upon laser irradiation, the designed SPI@hEL-RS17 nanoparticles exert potent antitumor efficacy against both 4T1 breast tumor and B16F10 melanoma models, which not only suppresses primary tumor growth but also inhibits lung metastasis and prevents tumor recurrence, exhibiting great potential in boosting CD47 blockade-based antitumor immunotherapy.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yuqi Cao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Cong Fu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Hening Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Jingwen Feng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
27
|
Zhu X, Li S. Nanomaterials in tumor immunotherapy: new strategies and challenges. Mol Cancer 2023; 22:94. [PMID: 37312116 PMCID: PMC10262535 DOI: 10.1186/s12943-023-01797-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
Tumor immunotherapy exerts its anti-tumor effects by stimulating and enhancing immune responses of the body. It has become another important modality of anti-tumor therapy with significant clinical efficacy and advantages compared to chemotherapy, radiotherapy and targeted therapy. Although various kinds of tumor immunotherapeutic drugs have emerged, the challenges faced in the delivery of these drugs, such as poor tumor permeability and low tumor cell uptake rate, had prevented their widespread application. Recently, nanomaterials had emerged as a means for treatment of different diseases due to their targeting properties, biocompatibility and functionalities. Moreover, nanomaterials possess various characteristics that overcome the defects of traditional tumor immunotherapy, such as large drug loading capacity, precise tumor targeting and easy modification, thus leading to their wide application in tumor immunotherapy. There are two main classes of novel nanoparticles mentioned in this review: organic (polymeric nanomaterials, liposomes and lipid nanoparticles) and inorganic (non-metallic nanomaterials and metallic nanomaterials). Besides, the fabrication method for nanoparticles, Nanoemulsions, was also introduced. In summary, this review article mainly discussed the research progress of tumor immunotherapy based on nanomaterials in the past few years and offers a theoretical basis for exploring novel tumor immunotherapy strategies in the future.
Collapse
Affiliation(s)
- Xudong Zhu
- Department of General Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China.
| |
Collapse
|
28
|
Xie Y, Ren Z, Chen H, Tang H, Zhu M, Lv Z, Bao H, Zhang Y, Liu R, Shen Y, Zheng Y, Miao D, Guo X, Chen H, Wang S, Pei J. A novel estrogen-targeted PEGylated liposome co-delivery oxaliplatin and paclitaxel for the treatment of ovarian cancer. Biomed Pharmacother 2023; 160:114304. [PMID: 36724638 DOI: 10.1016/j.biopha.2023.114304] [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: 11/30/2022] [Revised: 01/14/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Ovarian cancer is the second cause of death among gynecological malignancies. In this study, we designed a novel estrogen-targeted PEGylated liposome loaded with oxaliplatin and paclitaxel (ES-SSL-OXA/PTX) which could target estrogen receptor (ER) highly expressed on the surface of SKOV-3 cells to enhance therapeutic efficacy and reduce the side effects for SKOV-3 tumor therapy. ES-SSL-OXA/PTX was prepared by thin film hydration method and exhibited a uniform spherical morphology. Encapsulation efficiency (EE) were determined by HPLC method with the results of 44.10% for OXA and 65.85% for PTX. The mean particle size and polydispersity index (PDI) were 168.46 nm and 0.145, respectively. In vivo and in vitro targeting study confirmed that ES-SSL-OXA/PTX has optimum specific targeting ability. Meanwhile, In vitro and in vivo antitumor results of ES-SSL-OXA/PTX exhibited a superior antiproliferative effect on SKOV-3 cells and a stronger anti-tumor efficacy with the tumor inhibition rate of 85.24%. The pharmacokinetics results of ES-SSL-OXA/PTX showed a prolonged half-life time and a slowed clearance rate. The preliminary safety study of acute toxicity and long-term toxicity demonstrated ES-SSL-OXA/PTX exhibited a reduced toxicity profile. Based on the above results, ES-SSL-OXA/PTX could be a promising novel formulation for the treatment of ovarian cancer in future clinic.
Collapse
Affiliation(s)
- Yizhuo Xie
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Zhihui Ren
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Hongyu Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Huan Tang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Ming Zhu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Zhe Lv
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Han Bao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yan Zhang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Rui Liu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yujia Shen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yucui Zheng
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Dongfanghui Miao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xin Guo
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Hongli Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Shanshan Wang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jin Pei
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
29
|
Li W, Jiang Y, Lu J. Nanotechnology-enabled immunogenic cell death for improved cancer immunotherapy. Int J Pharm 2023; 634:122655. [PMID: 36720448 PMCID: PMC9975075 DOI: 10.1016/j.ijpharm.2023.122655] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023]
Abstract
Tumor immunotherapy has revolutionized the field of oncology treatments in recent years. As one of the promising strategies of cancer immunotherapy, tumor immunogenic cell death (ICD) has shown significant potential for tumor therapy. Nanoparticles are widely used for drug delivery due to their versatile characteristics, such as stability, slow blood elimination, and tumor-targeting ability. To increase the specificity of ICD inducers and improve the efficiency of ICD induction, functionally specific nanoparticles, such as liposomes, nanostructured lipid carriers, micelles, nanodiscs, biomembrane-coated nanoparticles and inorganic nanoparticles have been widely reported as the vehicles to deliver ICD inducers in vivo. In this review, we summarized the strategies of different nanoparticles for ICD-induced cancer immunotherapy, and systematically discussed their advantages and disadvantages as well as provided feasible strategies for solving these problems. We believe that this review will offer some insights into the design of effective nanoparticulate systems for the therapeutic delivery of ICD inducers, thus, promoting the development of ICD-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- 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
| | - 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; NCI-designated University of Arizona Comprehensive 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
|
30
|
Nanomaterials: Breaking through the bottleneck of tumor immunotherapy. Int J Biol Macromol 2023; 230:123159. [PMID: 36610572 DOI: 10.1016/j.ijbiomac.2023.123159] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/23/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
Immunotherapy exerts its excellent anti-tumor effects by stimulating and enhancing the immune response of the body, and has become another important class of anti-tumor therapy besides chemotherapy, targeted therapy and radiotherapy. Various types of immunotherapeutic drugs have gained their clinical values, but the in vivo delivery of drugs still faces many challenges, such as poor tumor permeability and low tumor cell uptake rate. In recent years, owing to highly targeting properties, better biocompatibility, and easy functionalization, nanomaterials have been widely applicated in tumor treatment, especially in tumor immunotherapy. Furthermore, nanomaterials have large drug loading capacity, strong tumor targeting and easy modification, which can effectively overcome the drawbacks of traditional immunotherapy. This paper reviews the progress of nanomaterial-based tumor immunotherapy in recent years and provides a theoretical basis for exploring new nanomaterial-based tumor immunotherapy strategies.
Collapse
|
31
|
Alhamhoom Y, Kakinani G, Rahamathulla M, Ali M. Osmani R, Hani U, Yoonus Thajudeen K, Kiran Raj G, Gowda DV. Recent advances in the liposomal nanovesicles based immunotherapy in the treatment of cancer: A review. Saudi Pharm J 2023; 31:279-294. [PMID: 36942270 PMCID: PMC10023551 DOI: 10.1016/j.jsps.2022.12.008] [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: 11/28/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy, along with chemotherapy, targeted delivery, radiation and surgery has become one of the most common cancer treatments. The aim of cancer immunology is to use the bodys immune system to combat tumors and develop a robust antitumor immune response. In the last few years, immune checkpoint inhibitors and chimeric antigen receptor-modified T cells have made substantial advancements in cancer immunotherapy. By boosting cell type-specific delivery and immunological responses, nanocarriers like liposomes have the ability to enhance greater immune responses. The efficacy of anti-tumor therapeutics is being significantly improved as liposomes can assist in resolving a number of issues that can arise from a variety of cancer immunotherapies. Since, liposomes can be loaded with both hydrophilic and hydrophobic drugs and protect the immunotherapeutic agents loaded inside the core, they offer significant advantages over other nano delivery systems. The use of liposomes for accurate and timely delivery of immunotherapies to particular targeted neoplasms, with little or no injury to healthy cells, maximizes immunotherapy efficacy. Liposomes are also suitable vehicles for delivering medications simultaneously with other therapies such as chemotherapy, radiation, and phototherapy. Liposomal nanoparticles will be introduced and used as an objective immunotherapy delivery system for great precision, making them a viable cancer treatment approach.With an emphasis on dendritic cells, T cells, tumor and natural killer cells, and macrophages; outline of many forms of immune-therapies in oncology and cutting-edge advances in liposomal nanovesicles for cancer immunotherapy are covered in this review.
Collapse
Affiliation(s)
- Yahya Alhamhoom
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Greeshma Kakinani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Mohamed Rahamathulla
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Riyaz Ali M. Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Kamal Yoonus Thajudeen
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - G. Kiran Raj
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Devegowda V. Gowda
- Department of Pharmaceutics, Cauvery College of Pharmacy, Mysuru 570 028, Karnataka, India
| |
Collapse
|
32
|
Yan C, Li Q, Sun Q, Yang L, Liu X, Zhao Y, Shi M, Li X, Luo K. Promising Nanomedicines of Shikonin for Cancer Therapy. Int J Nanomedicine 2023; 18:1195-1218. [PMID: 36926681 PMCID: PMC10013574 DOI: 10.2147/ijn.s401570] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/15/2023] [Indexed: 03/18/2023] Open
Abstract
Malignant tumor, the leading cause of death worldwide, poses a serious threat to human health. For decades, natural product has been proven to be an essential source for novel anticancer drug discovery. Shikonin (SHK), a natural molecule separated from the root of Lithospermum erythrorhizon, shows great potential in anticancer therapy. However, its further clinical application is significantly restricted by poor bioavailability, adverse effects, and non-selective toxicity. With the development of nanotechnology, nano drug delivery systems have emerged as promising strategies to improve bioavailability and enhance the therapeutic efficacy of drugs. To overcome the shortcoming of SHK, various nano drug delivery systems such as liposomes, polymeric micelles, nanoparticles, nanogels, and nanoemulsions, were developed to achieve efficient delivery for enhanced antitumor effects. Herein, this review summarizes the anticancer pharmacological activities and pharmacokinetics of SHK. Additionally, the latest progress of SHK nanomedicines in cancer therapy is outlined, focusing on long circulation, tumor targeting ability, tumor microenvironment responsive drug release, and nanosystem-mediated combination therapy. Finally, the challenges and prospects of SHK nanomedicines in the future clinical application are spotlighted.
Collapse
Affiliation(s)
- Chunmei Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Qiuxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Qiang Sun
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Lu Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Xing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Yuxin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Mingyi Shi
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Kaipei Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| |
Collapse
|
33
|
Xu X, Liu Y, Shao S, Li J, Xu Z, Yin Y, Zhao L, Wang Y, Liu D. Synthesis of Paclitaxel Derivatives for Remote Loading into Liposomes and Improved Therapeutic Effect. Molecules 2022; 27:molecules27227967. [PMID: 36432067 PMCID: PMC9694711 DOI: 10.3390/molecules27227967] [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: 10/22/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
Abstract
A series of novel paclitaxel derivatives modified by boronic acid according to the characteristics of the interaction between RB(OH)2 and different strapping agents of intraliposomal aqueous phase were designed and synthesized, which were then used to develop remote poorly water-soluble drugs loading into liposomes. Meanwhile, we screened nineteen paclitaxel boronic acid derivatives for their cytotoxic activities against three cancer cell lines (A549, HCT-116 and 4T1) and one normal cell line (LO2), and performed liposome formulation screening of active compounds. Among all the compounds, the liposome of 4d, with excellent drug-encapsulated efficiency (>95% for drug-to-lipid ratio of 0.1 w/w), was the most stable. Furthermore, the liposomes of compound 4d (8 mg/kg, 4 times) and higher dose of compound 4d (24 mg/kg, 4 times) showed better therapeutic effect than paclitaxel (8 mg/kg, 4 times) in the 4T1 tumor model in vivo, and the rates of tumor inhibition were 74.3%, 81.9% and 58.5%, respectively. This study provided a reasonable design strategy for the insoluble drugs to improve their drug loading into liposomes and anti-tumor effect in vivo.
Collapse
Affiliation(s)
- Xiangwei Xu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Shanshan Shao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhaochu Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yueling Yin
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
- Correspondence: (L.Z.); (Y.W.); (D.L.); Tel.: +86-024-4352-0218 (D.L.)
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Correspondence: (L.Z.); (Y.W.); (D.L.); Tel.: +86-024-4352-0218 (D.L.)
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
- Correspondence: (L.Z.); (Y.W.); (D.L.); Tel.: +86-024-4352-0218 (D.L.)
| |
Collapse
|
34
|
Koletti AE, Kontogiannopoulos KN, Gardikis K, Letsiou S, Papageorgiou VP, Assimopoulou AN. Nanostructured lipid carriers of alkannins and shikonins: Experimental design, characterization and bioactivity studies. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Zhang Y, Li J, Pu K. Recent advances in dual- and multi-responsive nanomedicines for precision cancer therapy. Biomaterials 2022; 291:121906. [DOI: 10.1016/j.biomaterials.2022.121906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022]
|
36
|
Targeting Glucose Metabolism Enzymes in Cancer Treatment: Current and Emerging Strategies. Cancers (Basel) 2022; 14:cancers14194568. [PMID: 36230492 PMCID: PMC9559313 DOI: 10.3390/cancers14194568] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Reprogramming of glucose metabolism is a hallmark of cancer and can be targeted by therapeutic agents. Some metabolism regulators, such as ivosidenib and enasidenib, have been approved for cancer treatment. Currently, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Furthermore, some natural products have shown efficacy in killing tumor cells by regulating glucose metabolism, offering novel therapeutic opportunities in cancer. However, most of them have failed to be translated into clinical applications due to low selectivity, high toxicity, and side effects. Recent studies suggest that combining glucose metabolism modulators with chemotherapeutic drugs, immunotherapeutic drugs, and other conventional anticancer drugs may be a future direction for cancer treatment. Abstract Reprogramming of glucose metabolism provides sufficient energy and raw materials for the proliferation, metastasis, and immune escape of cancer cells, which is enabled by glucose metabolism-related enzymes that are abundantly expressed in a broad range of cancers. Therefore, targeting glucose metabolism enzymes has emerged as a promising strategy for anticancer drug development. Although several glucose metabolism modulators have been approved for cancer treatment in recent years, some limitations exist, such as a short half-life, poor solubility, and numerous adverse effects. With the rapid development of medicinal chemicals, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Additionally, several studies have found that some natural products can suppress cancer progression by regulating glucose metabolism enzymes. In this review, we summarize the mechanisms underlying the reprogramming of glucose metabolism and present enzymes that could serve as therapeutic targets. In addition, we systematically review the existing drugs targeting glucose metabolism enzymes, including small-molecule modulators and natural products. Finally, the opportunities and challenges for glucose metabolism enzyme-targeted anticancer drugs are also discussed. In conclusion, combining glucose metabolism modulators with conventional anticancer drugs may be a promising cancer treatment strategy.
Collapse
|
37
|
Li J, Cai W, Yu J, Zhou S, Li X, He Z, Ouyang D, Liu H, Wang Y. Autophagy inhibition recovers deficient ICD-based cancer immunotherapy. Biomaterials 2022; 287:121651. [PMID: 35777331 DOI: 10.1016/j.biomaterials.2022.121651] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/16/2022] [Accepted: 06/23/2022] [Indexed: 12/07/2022]
Abstract
ICD effect is usually accompanied with robust autophagy that can depredate immune-associated antigens in tumors, thereby weakening the immune response against tumor growth. To circumvent this dilemma, we combined an ICD inducer (Shikonin, SHK) with an autophagy inhibitor (hydroxychloroquine, HCQ) for colon cancer immunotherapy. Notably, HCQ boosted SHK-induced antigen exposure in colon cancer in vitro and in vivo. However, autophagy inhibition caused loss of ATP, which compromised antitumor immune response. Therefore, a compensatory strategy was employed by introducing ATP as a remote loading gradient of the liposome to encapsulate HCQ (LipHCQa). LipHCQa achieved an excellent antitumor efficiency without dampening the immune response. Furthermore, a systematic determination of the optimal dosage of combined LipSHK and LipHCQa suggested that autophagy inhibiting at an appropriate dosage level was beneficial for maximizing ICD-based antitumor immunity. This study proved that autophagy inhibitors can recover the deficient ICD-based antitumor immune response and present potential clinical applications for cancer immunotherapy.
Collapse
Affiliation(s)
- Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang, 110016, China
| | - Wenxu Cai
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang, 110016, China
| | - Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang, 110016, China
| | - Shuang Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang, 110016, China
| | - Xianlu Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang, 110016, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang, 110016, China
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang, 110016, China.
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, Shenyang, 110016, China.
| |
Collapse
|
38
|
Chopra H, Bibi S, Goyal R, Gautam RK, Trivedi R, Upadhyay TK, Mujahid MH, Shah MA, Haris M, Khot KB, Gopan G, Singh I, Kim JK, Jose J, Abdel-Daim MM, Alhumaydhi FA, Emran TB, Kim B. Chemopreventive Potential of Dietary Nanonutraceuticals for Prostate Cancer: An Extensive Review. Front Oncol 2022; 12:925379. [PMID: 35903701 PMCID: PMC9315356 DOI: 10.3389/fonc.2022.925379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/25/2022] [Indexed: 12/24/2022] Open
Abstract
There are more than two hundred fifty different types of cancers, that are diagnosed around the world. Prostate cancer is one of the suspicious type of cancer spreading very fast around the world, it is reported that in 2018, 29430 patients died of prostate cancer in the United State of America (USA), and hence it is expected that one out of nine men diagnosed with this severe disease during their lives. Medical science has identified cancer at several stages and indicated genes mutations involved in the cancer cell progressions. Genetic implications have been studied extensively in cancer cell growth. So most efficacious drug for prostate cancer is highly required just like other severe diseases for men. So nutraceutical companies are playing major role to manage cancer disease by the recommendation of best natural products around the world, most of these natural products are isolated from plant and mushrooms because they contain several chemoprotective agents, which could reduce the chances of development of cancer and protect the cells for further progression. Some nutraceutical supplements might activate the cytotoxic chemotherapeutic effects by the mechanism of cell cycle arrest, cell differentiation procedures and changes in the redox states, but in other, it also elevate the levels of effectiveness of chemotherapeutic mechanism and in results, cancer cell becomes less reactive to chemotherapy. In this review, we have highlighted the prostate cancer and importance of nutraceuticals for the control and management of prostate cancer, and the significance of nutraceuticals to cancer patients during chemotherapy.
Collapse
Affiliation(s)
- Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-milat University, Islamabad, Pakistan
- Yunnan Herbal Laboratory, College of Ecology and Environmental Sciences, Yunnan University, Kunming, China
| | - Rajat Goyal
- Maharishi Markandeshwar (MM) School of Pharmacy, Maharishi Markandeshwar University, Sadopur-Ambala, India
- Maharishi Markandeshwar (MM) College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | - Rupesh K. Gautam
- Maharishi Markandeshwar (MM) School of Pharmacy, Maharishi Markandeshwar University, Sadopur-Ambala, India
| | - Rashmi Trivedi
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, India
| | - Mohd Hasan Mujahid
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, India
| | | | - Muhammad Haris
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Kartik Bhairu Khot
- Department of Pharmaceutics, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore, India
| | - Gopika Gopan
- Department of Pharmaceutics, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore, India
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Jin Kyu Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jobin Jose
- Department of Pharmaceutics, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore, India
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
39
|
Zheng Y, Xie L, Tie X, Cao L, Li Q, Quan Y, Tang L, Li Y. Remote drug loading into liposomes via click reaction. MATERIALS HORIZONS 2022; 9:1969-1977. [PMID: 35583553 DOI: 10.1039/d2mh00380e] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The development of liposome-based drugs was severely limited due to inefficient loading strategies. Herein, we developed a click reaction-mediated loading procedure by designing an enzyme-sensitive maleimide (MAL) tag for ferrying chemotherapeutics into preformed liposomes containing glutathione (GSH). Based on this strategy, various hydrophobic drugs could be encapsulated into liposomes within 5-30 min with encapsulation efficiency >95% and loading capacity of 10-30% (w/w). The entrapped cargo could be slowly released from the liposomes, followed by rapid enzyme-mediated conversion into active drugs to exert antitumor activity under physiological conditions. The resulting drug-loaded liposomes significantly prolonged the blood circulation of cargos and displayed more potent in vivo antitumor efficacy than free drugs at the equitoxic dose. More importantly, this method is a remote drug loading strategy in nature, which is suitable for industrial production. This is the first demonstration of active loading of MAL-tagged chemotherapeutics in liposomes for improved antitumor efficacies, which has the potential to serve as a universal drug loading strategy for the development of liposomal formulations of chemotherapeutics.
Collapse
Affiliation(s)
- Yaxin Zheng
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Lei Xie
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Xiaoru Tie
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Lei Cao
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Qingyuan Li
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Yue Quan
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Lingfeng Tang
- School of Pharmacy, Key Laboratory of Sichuan Province for Specific Structure of Small Molecule Drugs, Chengdu Medical College, Chengdu, China
| | - Yang Li
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, P. R. China.
| |
Collapse
|
40
|
Peng Y, He P, Gao X, Liu G, Cheng H. A Superstable Homogeneous Lipiodol-Nanoformulation to Overcome the Dilemma of Interventional Embolization Chemotherapy. Front Bioeng Biotechnol 2022; 10:952194. [PMID: 35800328 PMCID: PMC9253561 DOI: 10.3389/fbioe.2022.952194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/03/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
| | | | | | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Hongwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
41
|
Fu X, Shi Y, Zang H, Wang Q, Wang Y, Wu H, Qiu S, Shen H, Mo F, Zhang Y, Lin G. Combination of oxaliplatin and POM-1 by nanoliposomes to reprogram the tumor immune microenvironment. J Control Release 2022; 347:1-13. [PMID: 35508221 DOI: 10.1016/j.jconrel.2022.04.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022]
Abstract
Some chemotherapy can damage tumor cells, releasing damage-related molecular patterns including ATP to improve immunological recognition against the tumor by immunogenic cell death (ICD). However, the immune-stimulating ATP may be rapidly degraded into immunosuppressive adenosine by highly expressed CD39 and CD73 in the tumor microenvironment, which leads to immune escape. Based on the above paradox, a liposome nanoplatform combined with ICD inducer (oxaliplatin) and CD39 inhibitor (POM-1) is designed for immunochemotherapy. The liposomes efficiently load the phospholipid-like oxaliplatin prodrug, and the cationic charged surface could adsorb POM-1. Rationally designed DSPE-PEGn-pep, on the one hand, could cover and hide POM-1 to avoid systematic toxicity and, on the other, achieve a response and charge reversal to favor POM-1 shedding and tumor deep penetration. This combination maximizes the ICD effect, and takes two-pronged advantage of stimulating the immune response and relieving immune suppression. The designed POL can effectively inhibit the growth of in situ, lung metastasis and postoperative recurrence melanoma model and form long-term immune memory. With the powerful clinical transformation potential of nanoliposome platforms, this new synergistic strategy is expected to enhance anticancer effects safely and effectively.
Collapse
Affiliation(s)
- Xianglei Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yanbin Shi
- School of Mechanical and Automotive Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Hengchang Zang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qingjie Wang
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hang Wu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shengnan Qiu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hua Shen
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Fanyang Mo
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yankun Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Guimei Lin
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
42
|
Ma J, Zhang C, Shi G, Yue D, Shu Y, Hu S, Qi Z, Chen Y, Zhang B, Zhang Y, Huang A, Su C, Zhang Y, Deng H, Cheng P. High-dose VitC plus oncolytic adenoviruses enhance immunogenic tumor cell death and reprogram tumor immune microenvironment. Mol Ther 2022; 30:644-661. [PMID: 34547462 PMCID: PMC8821933 DOI: 10.1016/j.ymthe.2021.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/25/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023] Open
Abstract
Preclinical and clinical studies have validated the antitumor effects of several oncolytic viruses (OVs). However, the efficacy of OVs is limited when they are administered as monotherapies. Combination therapy is a promising direction for oncolytic virotherapy in the future. A high dose of vitamin C (VitC) exerts anticancer effects by triggering the accretion of substantial amounts of reactive oxygen species (ROS). OVs can induce immunogenic tumor cell death and elicit an antitumor immune response. ROS play an important role in immunogenic cell death (ICD). This study aimed to explore whether high-dose VitC in combination with oncolytic adenoviruses (oAds) exhibited a synergistic antitumor effect. High-dose VitC synergized with oAds against tumor by enhancing immunogenic tumor cell death. Combination therapy with high-dose VitC and oAds significantly increased the number of T cells in the tumor microenvironment (TME) and promoted the activation of T cells. Furthermore, the antitumor effect of the combination therapy was CD8+ T cell dependent. In addition, combination therapy with high-dose VitC and oAds reprogramed the immunosuppressive TME. Our study provides a new strategy for combination therapy of OVs.
Collapse
Affiliation(s)
- Jinhu Ma
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Chunxue Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Dan Yue
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Yongheng Shu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Shichuan Hu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Zhongbing Qi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Yanwei Chen
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Bin Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Yong Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Anliang Huang
- Department of Pathology, Chengdu Fifth People’s Hospital, Chengdu, PR China
| | - Chao Su
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Yan Zhang
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China
| | - Ping Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China,Corresponding author: Prof. Ping Cheng, State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 17 People’s South Road, Chengdu 610041, PR China.
| |
Collapse
|
43
|
Ratiometric co-delivery of doxorubicin and paclitaxel prodrug by remote-loading liposomes for the treatment of triple-negative breast cancer. Drug Deliv Transl Res 2022; 12:2537-2549. [DOI: 10.1007/s13346-021-01105-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 11/26/2022]
|
44
|
The nano delivery systems and applications of mRNA. Eur J Med Chem 2022; 227:113910. [PMID: 34689071 PMCID: PMC8497955 DOI: 10.1016/j.ejmech.2021.113910] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/06/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023]
Abstract
The current COVID-19 epidemic has greatly accelerated the application of mRNA technology to our real world, and during this battle mRNA has proven it's unique advantages compared to traditional biopharmaceutical and vaccine technology. In order to overcome mRNA instability in human physiological environments, mRNA chemical modifications and nano delivery systems are two key factors for their in vivo applications. In this review, we would like to summarize the challenges for clinical translation of mRNA-based therapeutics, with an emphasis on recent advances in innovative materials and delivery strategies. The nano delivery systems include lipid delivery systems (lipid nanoparticles and liposomes), polymer complexes, micelles, cationic peptides and so on. The similarities and differences of lipid nanoparticles and liposomes are also discussed. In addition, this review also present the applications of mRNA to other areas than COVID-19 vaccine, such as infectious diseases, tumors, and cardiovascular disease, for which a variety of candidate vaccines or drugs have entered clinical trials. Furthermore, mRNA was found that it might be used to treat some genetic disease, overcome the immaturity of the immune system due to the small fetal size in utero, treat some neurological diseases that are difficult to be treated surgically, even be used in advancing the translation of iPSC technology et al. In short, mRNA has a wide range of applications, and its era has just begun.
Collapse
|
45
|
Qian Y, Mao J, Leng X, Zhu L, Xue R, Jin Z, Jiang H, Liu H, Zhang F, Bi X, Chen Z, Wang J. Co-delivery of proanthocyanidin and mitoxantrone induce synergistic immunogenic cell death to potentiate cancer immunotherapy. Biomater Sci 2022; 10:4549-4560. [DOI: 10.1039/d2bm00611a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Immunological checkpoint inhibitors provide a revolutionary method for cancer treatment. However, due to low tumor mutations and insufficient infiltration of immune cells in the tumor microenvironment, 85% of colorectal cancer...
Collapse
|
46
|
Kaur K, Singh A, Sharma H, Punj S, Bedi N. Formulation Strategies and Therapeutic Applications of Shikonin and Related Derivatives. RECENT ADVANCES IN DRUG DELIVERY AND FORMULATION 2022; 16:55-67. [PMID: 35236278 DOI: 10.2174/2667387816666220302112201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
Shikonin and its derivatives are excellent representatives of biologically active naphthoquinones. A wide range of investigations carried out in the last few decades validated their pharmacological efficacy. Besides having magnificent therapeutic potential, shikonin and its derivatives suffer from various pharmacokinetic, toxicity, and stability issues like poor bioavailability, nephrotoxicity, photodegradation, etc. Recently, various research groups have developed an extensive range of formulations to tackle these issues to ease their path to clinical practice. The latest formulation approaches have been focused on exploiting the unique features of novel functional excipients, which in turn escalate the therapeutic effect of shikonin. Moreover, the codelivery approach in various drug delivery systems has been taken into consideration in a recent while to reduce toxicity associated with shikonin and its derivatives. This review sheds light on the essential reports and patents published related to the array of formulations containing shikonin and its derivatives.
Collapse
Affiliation(s)
- Kirandeep Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Atamjit Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Hamayal Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Sanha Punj
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Neena Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| |
Collapse
|
47
|
Feng W, Shi W, Liu S, Liu H, Liu Y, Ge P, Zhang H. Fe(III)-Shikonin Supramolecular Nanomedicine for Combined Therapy of Tumor via Ferroptosis and Necroptosis. Adv Healthc Mater 2022; 11:e2101926. [PMID: 34738742 DOI: 10.1002/adhm.202101926] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/26/2021] [Indexed: 01/15/2023]
Abstract
Most of the antitumor chemotherapeutic drugs execute the therapeutic performance upon eliciting tumor cell apoptosis, which may cause chemoresistance of tumors. Design of novel drugs to eradicate apoptosis-resistant tumors via non-apoptotic cell death pathways is promising for improving the long-term chemotherapeutic efficacy. Herein, a Fe(III)-Shikonin metal-polyphenol-coordinated supramolecular nanomedicine for combined therapy of tumor via ferroptosis and necroptosis is designed. The construction of the nanomedicine based on the coordinated self-assembly between Fe3+ and Shikonin not only overcomes the shortcomings of Shikonin including its low bioavailability and high toxicity toward normal tissues, but also integrates the theranostics functions of Fe ions. Under the exposure of the high concentration of glutathione (GSH) in tumor cells, the as-prepared nanomedicine will disassemble into Fe2+ and Shikonin, followed by stimulating the tumor cell death through ferroptosis and necroptosis. In addition, benefiting from the stealth effect of polyethylene glycol (PEG) and the targeting ability of cyclo(Arg-Gly-Asp-d-Phe-Lys) (cRGD) to αv β3 -integrin, NH2 -PEG-cRGD-modified nanomedicine exhibits a GSH-responsive therapy toward 4T1 tumor in vivo and self-enhanced longitudinal relaxation (T1 )-weighted imaging property. Since the self-assembly of natural Shikonin and human body-necessary Fe element is facile and feasible, the work may provide a promising supramolecular nanomedicine for next-generation chemotherapeutic applications.
Collapse
Affiliation(s)
- Wenjie Feng
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University Changchun 130012 P. R. China
| | - Wanrui Shi
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University Changchun 130012 P. R. China
| | - Shuwei Liu
- Joint Laboratory of Opto‐Functional Theranostics in Medicine and Chemistry The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Huiwen Liu
- Joint Laboratory of Opto‐Functional Theranostics in Medicine and Chemistry The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Yi Liu
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University Changchun 130012 P. R. China
- Joint Laboratory of Opto‐Functional Theranostics in Medicine and Chemistry The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Pengfei Ge
- Department of Neurosurgery The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University Changchun 130012 P. R. China
- Joint Laboratory of Opto‐Functional Theranostics in Medicine and Chemistry The First Hospital of Jilin University Changchun 130021 P. R. China
| |
Collapse
|
48
|
Li J, Li S, Li Y, Yuan G, Shen Y, Peng Y, Kong L, Yang C, Zhang Z, Li Z. A magnetic resonance nanoprobe with STING activation character collaborates with platinum-based drug for enhanced tumor immunochemotherapy. J Nanobiotechnology 2021; 19:415. [PMID: 34895243 PMCID: PMC8666035 DOI: 10.1186/s12951-021-01158-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/21/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Immunochemotherapy is a potent anti-tumor strategy, however, how to select therapeutic drugs to enhance the combined therapeutic effect still needs to be explored. METHODS AND RESULTS: Herein, a magnetic resonance nanoprobe (MnP@Lip) with STING (Stimulator of INterferon Genes) activation character was synthesized and co-administered with platinum-based chemotherapeutics for enhanced immunochemotherapy. MnP@Lip nanoparticles was prepared by simple fabrication process with good reproducibility, pH-sensitive drug release behavior and biocompatibility. In vitro experiments elucidated that Mn2+ can promote the polarization of M0 and/or M2 macrophages to M1 phenotype, and promote the maturation of BMDC cells. Upon Mn2+ treatment, the STING pathway was activated in tumor cells, mouse lung epithelial cells, and immune cells. More importantly, anti-tumor experiments in vivo proved that MnP@Lip combined with platinum-based chemotherapeutics increased T cells infiltration in the tumor microenvironment, and inhibited tumor growth in the orthotopic therapeutic and postoperative tumor models. CONCLUSIONS This kind of therapeutic strategy that combined MnP@Lip nanoparticles with platinum-based chemotherapeutics may provide a novel insight for immunochemotherapy.
Collapse
Affiliation(s)
- Jiali Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, People's Republic of China
| | - Shichao Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, People's Republic of China
| | - Yang Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Guanjie Yuan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, People's Republic of China
| | - Yaqi Shen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, People's Republic of China
| | - Yang Peng
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, People's Republic of China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.,Hubei Engineering Research Center for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, People's Republic of China.
| |
Collapse
|
49
|
Wan Y, Wang X, Zhang P, Zhang M, Kou M, Shi C, Peng X, Wang X. Control of Foodborne Staphylococcus aureus by Shikonin, a Natural Extract. Foods 2021; 10:foods10122954. [PMID: 34945505 PMCID: PMC8700560 DOI: 10.3390/foods10122954] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 01/08/2023] Open
Abstract
Foodborne Staphylococcus aureus (S. aureus) has attracted widespread attention due to its foodborne infection and food poisoning in human. Shikonin exhibits antibacterial activity against a variety of microorganisms, but there are few studies on its antibacterial activity against S. aureus. This study aims to explore the antibacterial activity and mechanism of shikonin against foodborne S. aureus. The results show that the minimum inhibitory concentrations (MICs) and the minimum bactericidal concentrations (MBCs) of shikonin were equal for all tested strains ranging from 35 μg/mL to 70 μg/mL. Shikonin inhibited the growth of S. aureus by reducing intracellular ATP concentrations, hyperpolarizing cell membrane, destroying the integrity of cell membrane, and changing cell morphology. At the non-inhibitory concentrations (NICs), shikonin significantly inhibited biofilm formation of S. aureus, which was attributed to inhibiting the expression of cidA and sarA genes. Moreover, shikonin also markedly inhibited the transcription and expression of virulence genes (sea and hla) in S. aureus. In addition, shikonin has exhibited antibacterial ability against both planktonic and biofilm forms of S. aureus. Importantly, in vivo results show that shikonin has excellent biocompatibility. Moreover, both the heat stability of shikonin and the antimicrobial activity of shikonin against S. aureus were excellent in food. Our findings suggest that shikonin are promising for use as a natural food additive, and it also has great potential in effectively controlling the contamination of S. aureus in food and reducing the number of illnesses associated with S. aureus.
Collapse
|