1
|
Chen H, Qu H, Pan Y, Cheng W, Xue X. Manganese-coordinated nanoparticle with high drug-loading capacity and synergistic photo-/immuno-therapy for cancer treatments. Biomaterials 2025; 312:122745. [PMID: 39098306 DOI: 10.1016/j.biomaterials.2024.122745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/01/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Stimulator of interferon genes (STING) agonists have shown promise in cancer treatment by stimulating the innate immune response, yet their clinical potential has been limited by inefficient cytosolic entry and unsatisfactory pharmacological activities. Moreover, aggressive tumors with "cold" and immunosuppressive microenvironments may not be effectively suppressed solely through innate immunotherapy. Herein, we propose a multifaceted immunostimulating nanoparticle (Mn-MC NP), which integrates manganese II (Mn2+) coordinated photosensitizers (chlorin e6, Ce6) and STING agonists (MSA-2) within a PEGylated nanostructure. In Mn-MC NPs, Ce6 exerts potent phototherapeutic effects, facilitating tumor ablation and inducing immunogenic cell death to elicit robust adaptive antitumor immunity. MSA-2 activates the STING pathway powered by Mn2+, thereby promoting innate antitumor immunity. The Mn-MC NPs feature a high drug-loading capacity (63.42 %) and directly ablate tumor tissue while synergistically boosting both adaptive and innate immune responses. In subsutaneous tumor mouse models, the Mn-MC NPs exhibit remarkable efficacy in not only eradicating primary tumors but also impeding the progression of distal and metastatic tumors through synergistic immunotherapy. Additionally, they contribute to preventing tumor recurrence by fostering long-term immunological memory. Our multifaceted immunostimulating nanoparticle holds significant potential for overcoming limitations associated with insufficient antitumor immunity and ineffective cancer treatment.
Collapse
Affiliation(s)
- Han Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haijing Qu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Yuqing Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Cheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiangdong Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
2
|
Zhang J, Sang X, Yuan Y, Shen J, Fang Y, Qin M, Zheng H, Zhu Z. 4-Deoxy- ε-Pyrromycinone: A Promising Drug/Lead Compound to Treat Tumors. Drug Des Devel Ther 2024; 18:2367-2379. [PMID: 38911033 PMCID: PMC11193465 DOI: 10.2147/dddt.s461594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024] Open
Abstract
Background Anthraquinone drugs are widely used in the treatment of tumors. However, multidrug resistance and severe cardiac toxicity limit its use, which have led to the discovery of new analogues. In this paper, 4-Deoxy-ε-pyrromycinone (4-Deo), belonging to anthraquinone compounds, was first been studied with the anti-tumor effects and the safety in vitro and in vivo as a new anti-tumor drug or lead compound. Methods The quantitative analysis of 4-Deo was established by UV methodology. The anti-cancer effect of 4-Deo in vitro was evaluated by cytotoxicity experiments of H22, HepG2 and Caco2, and the anti-cancer mechanism was explored by cell apoptosis and cycle. The tumor-bearing mouse model was established by subcutaneous inoculation of H22 cells to evaluate the anti-tumor effect of 4-Deo in vivo. The safety of 4-Deo was verified by the in vitro safety experiments of healthy cells and the in vivo safety experiments of H22 tumor-bearing mice. Tumor tissue sections were labeled with CRT, HMGB1, IL-6 and CD115 to explore the preliminary anti-cancer mechanism by immunohistochemistry. Results In vitro experiments demonstrated that 4-Deo could inhibit the growth of H22 by inducing cell necrosis and blocking cells in S phase, and 4-Deo has less damage to healthy cells. In vivo experiments showed that 4-Deo increased the positive area of CRT and HMGB1, which may inhibit tumor growth by triggering immunogenic cell death (ICD). In addition, 4-Deo reduced the positive area of CSF1R, and the anti-tumor effect may be achieved by blocking the transformation of tumor-associated macrophages (TAMs) to M2 phenotype. Conclusion In summary, this paper demonstrated the promise of 4-Deo for cancer treatment in vitro and in vivo. This paper lays the foundation for the study of 4-Deo, which is beneficial for the further development anti-tumor drugs based on the lead compound of 4-Deo.
Collapse
Affiliation(s)
- Jiping Zhang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Xianan Sang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Yichao Yuan
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Jiawei Shen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Yuanyuan Fang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Minjing Qin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Hangsheng Zheng
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Zhihong Zhu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| |
Collapse
|
3
|
Sun Y, Bai Y, Liu S, Cui S, Xu P. Thermosensitive Micelles Gel to Deliver Quercetin Locally for Enhanced Antibreast Cancer Efficacy: An In Vitro Evaluation. IET Nanobiotechnol 2023; 2023:7971492. [PMID: 38863476 PMCID: PMC11095074 DOI: 10.1049/2023/7971492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/13/2024] Open
Abstract
Although quercetin is low cytotoxicity to normal human cells, quercetin is effective against the growth of some tumors. Given the poor blood stability in vivo, insolubility, low delivery efficiency, and poor medicinal properties of quercetin, we developed a local drug delivery system comprising quercetin core's polymer micelles and F127 hydrogel stroma. In vitro evaluation revealed that quercetin core's polymer micelles have excellent antitumor activity and could inhibit the multiplication of 4T1 breast cancer cells through the apoptosis pathway. Meanwhile, a rheological study revealed that the quercetin core's micelles gel possessed excellent properties of hydrogel formation and injectability of liquid preparation as a local drug delivery system after the quercetin core's polymer micelles were loaded into the F127 hydrogel stroma. Our study findings indicated that the drug stability and stable release capacity of quercetin were vastly improved with the composite formulation of the micelles gel. This not only realized drug injectability but also drug storage in the semisolid form, which is a more comfortable and slower drug-releasing form that will eventually exert a proper therapeutic effect. In conclusion, quercetin micellar hydrogel system has better antitumor activity and excellent hydrogel properties.
Collapse
Affiliation(s)
- Yanxue Sun
- Department of Pharmaceutical Engineering, College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Yun Bai
- Department of Pharmaceutical Engineering, College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Silu Liu
- Department of Pharmaceutical Engineering, College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Shuxia Cui
- Department of Anesthesiology, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Pengcheng Xu
- Department of Pharmaceutical Engineering, College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
4
|
Qu H, Chen H, Cheng W, Wang Y, Xia Y, Zhang L, Ma B, Hu R, Xue X. A Supramolecular Assembly Strategy for Hydrophilic Drug Delivery towards Synergistic Cancer Treatment. Acta Biomater 2023; 164:407-421. [PMID: 37088157 DOI: 10.1016/j.actbio.2023.04.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/24/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
To improve the drug loading, tumor targeting, and delivery simplicity of hydrophilic drugs, we propose a supramolecular assembly strategy that potentially benefits a wide range of hydrophilic drug delivery. Firstly, we choose a hydrophilic drug (tirapazamine) as a model drug to directly co-assemble with chlorin e6 (Ce6) at different molar ratios, and systematically evaluate the resultant Ce6-tirapazamine nanoparticles (CT NPs) in aspects of size distribution, polydispersity, morphology, optical properties and molecular dynamics simulation. Based on the assembling facts between Ce6 and tirapazamine, we summarize a plausible rule of the supramolecular assembly for hydrophilic drugs. To validate our findings, more drugs with increasing hydrophilicity, such as temozolomide, gemcitabine hydrochloride and 5-azacytidine, successfully co-assemble with Ce6 into nanostructures by following similar assembling behaviors, demonstrating that our assembling rule may guide a wide range of hydrophilic drug delivery. Next, the combination of Ce6 and tirapazamine was chosen as the representative to investigate the anti-tumor activities of the supramolecular assemblies. CT NPs showed synergistic anti-tumor efficacy, increased tumor accumulation and significant tumor progression and metastasis inhibition in tumor-bearing mice. We anticipate that the supramolecular assembly mechanism will provide broad guidance for developing easy-to-make but functional nanomedicines. STATEMENT OF SIGNIFICANCE: Although thousands of nanomedicines have been developed, only a few have been approved for clinical use. The manufacturing complexity significantly hinders the "bench-to-bed" translation of nanomedicines. Hence, we need to rethink how to conduct research on translational nanomedicines by avoiding more and more complex chemistry and complicated nanostructures. Here, we summarize a plausible rule according to multiple supramolecular assembly pairs and propose a supramolecular assembly strategy that can improve the drug loading, tumor targeting, and manufacturing simplicity of nanomedicine for hydrophilic drugs. The supramolecular assembly strategy would guide a broader range of drug delivery to provide a new paradigm for developing easy-to-make but multifunctional nanoformulations for synergistic cancer treatment.
Collapse
Affiliation(s)
- Haijing Qu
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Han Chen
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Cheng
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yanjun Wang
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China; Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Centre for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, China
| | - Yangyang Xia
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China; Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Centre for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, China
| | - Linghao Zhang
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Buyong Ma
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rong Hu
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Centre for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, China.
| | - Xiangdong Xue
- School of Pharmacy, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
5
|
Yuan L, Su Y, Yu B, Shen Y, Cong H. D-A-D organic small molecules with AIE effect for fluorescence imaging guided photothermal therapy. Biomater Sci 2023; 11:985-997. [PMID: 36541206 DOI: 10.1039/d2bm01912d] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Near infrared (NIR) fluorescent organic molecules as fluorescent probes accurately guide photothermal therapy as a potential antitumor method. However, the aggregation and quenching of organic fluorescent molecules and poor tissue permeability greatly limit their therapeutic effect and clinical transformation. In this paper, with a D-A-D structure as the molecular skeleton, cyclopentadithiophene (CPDT) as the donor (D), diketopyrrolopyrrole (DPP) as the acceptor (A), and long-chain isooctane as the shielding unit, organic fluorescent small molecules with a strong absorption band and bright NIR-II emission were synthesized. Then, tetraphenylethylene (TPE) molecules with typical AIE structure characteristics were introduced on both sides of the organic fluorescent small molecules, and an organic small molecular fluorophore (TDA) with AIE characteristics and the photothermal effect was designed. Through a series of experimental characterization techniques, it is proved that TDA NPs have good biocompatibility and tissue permeability, and can accurately locate the tumor location through NIR-II fluorescence imaging to achieve local photothermal treatment of tumors.
Collapse
Affiliation(s)
- Lin Yuan
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao 266071, China.
| | - Yingbin Su
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao 266071, China.
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao 266071, China. .,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao 266071, China. .,Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao 266071, China. .,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.,School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| |
Collapse
|
6
|
Wang F, Duan H, Xu W, Sheng G, Sun Z, Chu H. Light-activated nanomaterials for tumor immunotherapy. Front Chem 2022; 10:1031811. [PMID: 36277335 PMCID: PMC9585221 DOI: 10.3389/fchem.2022.1031811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/20/2022] [Indexed: 11/20/2022] Open
Abstract
Tumor immunotherapy mainly relies on activating the immune system to achieve antitumor treatment. However, the present tumor immunotherapy used in the clinic showed low treatment efficacy with high systematic toxicity. To overcome the shortcomings of traditional drugs for immunotherapy, a series of antitumor immunotherapies based on nanomaterials have been developed to enhance the body’s antitumor immune response and reduce systematic toxicity. Due to the noninvasiveness, remote controllability, and high temporal and spatial resolution of light, photocontrolled nanomaterials irradiated by excitation light have been widely used in drug delivery and photocontrolled switching. This review aims to highlight recent advances in antitumor immunotherapy based on photocontrolled nanomaterials. We emphasized the advantages of nanocomposites for antitumor immunotherapy and highlighted the latest progress of antitumor immunotherapy based on photoactivated nanomaterials. Finally, the challenges and future prospects of light-activated nanomaterials in antitumor immunity are discussed.
Collapse
Affiliation(s)
- Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Huijuan Duan
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Weizhe Xu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Gang Sheng
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- *Correspondence: Hongqian Chu,
| |
Collapse
|
7
|
Calixarenes as Host Molecules for Drug Carriers in the Cosmetic and Medical Field. Macromol Res 2022. [DOI: 10.1007/s13233-022-0094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
8
|
Feng Q, Wang M, Muhtar E, Wang Y, Zhu H. Nanoparticles of a New Small-Molecule P-Selectin Inhibitor Attenuate Thrombosis, Inflammation, and Tumor Growth in Two Animal Models. Int J Nanomedicine 2021; 16:5777-5795. [PMID: 34471352 PMCID: PMC8403725 DOI: 10.2147/ijn.s316863] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/18/2021] [Indexed: 12/26/2022] Open
Abstract
Purpose To assess whether the newly designed small-molecule oral P-selectin inhibitor 3S-1,2,3,4-tetrahydro-β-carboline-3-methyl aspartyl ester (THCMA) as a nanomedicine enhances antithrombosis, anti-inflammation, and antitumor activity more than the clinical trial drug PSI-697. Methods THCMA was designed as an amphiphile containing pharmacophores of PSI-697. Its nanofeatures were explored with TEM, SEM, Tyndall effect, ζ-potential, FT-ICR-MS, and NOESY 2D 1H NMR. The P-selectin inhibitory effect of THCMA was demonstrated with molecular docking, ultraviolet (UV) spectra, and competitive ELISA. In vivo and in vitro assays — anti-arterial thrombosis, anti–venous thrombosis, anti-inflammation, antitumor growth, anti–platelet aggregation, rat-tail bleeding time, anticoagulation index, soluble P-selectin (sP-selectin) expression, and serum TNFα expression — were performed to explore bioactivity and potential mechanisms. Water solubility of THCMA was measured using UV-absorption spectra. Results THCMA self-assembled into nanorings of approximately 100 nm in diameter. Its water solubility was about 1,030-fold that of PSI-697. THCMA exhibited more potent P-selectin inhibitory effect than PSI-697. The oral efficacy of THCMA was 100-fold that of PSI-697 in inhibiting arterial and venous thrombosis and tenfold in inhibiting inflammation. THCMA inhibited thrombosis at a dose that produces no coagulation disorders and no bleeding risk. THCMA exhibited enhanced antitumor activity over PSI-697 without systemic chemotherapy toxicity. THCMA significantly inhibited platelet aggregation in vitro and downregulated the expression levels of serum sP-selectin and TNFα in vivo. Conclusion A new small-molecule P-selectin inhibitor, THCMA, has been successfully designed as a nanomedicine with largely enhanced oral efficacy compared to the clinical trial drug PSI-697, and thus might be developed for the oral treatment of arterial thrombosis, venous thrombosis, inflammation, and cancer-associated thrombosis.
Collapse
Affiliation(s)
- Qiqi Feng
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Mengyang Wang
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Eldar Muhtar
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Yaonan Wang
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Haimei Zhu
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| |
Collapse
|
9
|
Xue X, Ricci M, Qu H, Lindstrom A, Zhang D, Wu H, Lin TY, Li Y. Iron-crosslinked Rososome with robust stability and high drug loading for synergistic cancer therapy. J Control Release 2021; 329:794-804. [PMID: 33039481 PMCID: PMC7904601 DOI: 10.1016/j.jconrel.2020.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/21/2020] [Accepted: 10/05/2020] [Indexed: 12/24/2022]
Abstract
Development of liposomal nanomedicine with robust stability, high drug loading and synergistic efficacy is a promising strategy for effective cancer therapy. Here, we present an iron-crosslinked rosmarinic liposome (Rososome) which can load high contents of drugs (including 25.8% rosmarinic acid and 9.04% doxorubicin), keep stable in a high concentration of anionic detergent and exhibit synergistic anti-cancer efficacy. The Rososomes were constructed by rosmarinic acid-lipid conjugates which not only work synergistically with doxorubicin by producing reactive oxygen species but also provide catechol moieties for the iron cross-linkages. The cross-linkages can lock the payloads tightly, endowing the crosslinked Rososome with better stability and pharmacokinetics than its non-crosslinked counterpart. On the syngeneic mouse model of breast cancer, the iron-crosslinked Rososomes exhibit better anticancer efficacy than free rosmarinic acid, doxorubicin, non-crosslinked Rososome and commercial liposomal formulation of doxorubicin (DOXIL). This study introduces a novel strategy for the development of liposomes with robust stability, high drug loading and synergistic anti-cancer efficacy.
Collapse
Affiliation(s)
- Xiangdong Xue
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Marina Ricci
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA; Department of Clinical and Biological Sciences, University of Torino, Corso Raffaello 30, Turin 10125, Italy
| | - Haijing Qu
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Aaron Lindstrom
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Dalin Zhang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Hao Wu
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Tzu-Yin Lin
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
10
|
Zhang S, Zhang Y. Promoting Dual-Targeting Anticancer Effect by Regulating the Dynamic Intracellular Self-Assembly. ACS APPLIED MATERIALS & INTERFACES 2020; 12:41105-41112. [PMID: 32819089 DOI: 10.1021/acsami.0c12271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Despite the promise of nanomedicine in the fight against complex diseases, the enthusiasm for its pharmaceutical development is backed by the elevated costs associated with the R&D process. Therefore, as a compromise solution, nanotechnology was mainly applied as a drug delivery system to improve bioavailability and controllability of pharmaceutical drugs. Attempting to break the restrictions without elevating potential costs, we multiply the functions of excipients in the nanodelivery system by endowing subcellular-targeting ability. To prove the concept, fluorescent endoplasmic reticulum-targeted short peptides were covalently connected to chemotherapy medication chlorambucil achieving enhanced drug-loading efficiency. Via visualized intracellular dynamic enzyme-catalyzed hydrolysis, the ER-targeting excipient and nucleus-targeting chlorambucil are released simultaneously, achieving a synergistic anticancer effect and elucidating the influence of intracellular self-assembly transition on enzymatic reactions.
Collapse
Affiliation(s)
- Shijin Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna Son, Okinawa 904-0495, Japan
| | - Ye Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna Son, Okinawa 904-0495, Japan
| |
Collapse
|
11
|
Kim K, Khang D. Past, Present, and Future of Anticancer Nanomedicine. Int J Nanomedicine 2020; 15:5719-5743. [PMID: 32821098 PMCID: PMC7418170 DOI: 10.2147/ijn.s254774] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
This review aims to summarize the methods that have been used till today, highlight methods that are currently being developed, and predict the future roadmap for anticancer therapy. In the beginning of this review, established approaches for anticancer therapy, such as conventional chemotherapy, hormonal therapy, monoclonal antibodies, and tyrosine kinase inhibitors are summarized. To counteract the side effects of conventional chemotherapy and to increase limited anticancer efficacy, nanodrug- and stem cell-based therapies have been introduced. However, current level of understanding and strategies of nanodrug and stem cell-based therapies have limitations that make them inadequate for clinical application. Subsequently, this manuscript reviews methods with fewer side effects compared to those of the methods mentioned above which are currently being investigated and are already being applied in the clinic. The newer strategies that are already being clinically applied include cancer immunotherapy, especially T cell-mediated therapy and immune checkpoint inhibitors, and strategies that are gaining attention include the manipulation of the tumor microenvironment or the activation of dendritic cells. Tumor-associated macrophage repolarization is another potential strategy for cancer immunotherapy, a method which activates macrophages to immunologically attack malignant cells. At the end of this review, we discuss combination therapies, which are the future of cancer treatment. Nanoparticle-based anticancer immunotherapies seem to be effective, in that they effectively use nanodrugs to elicit a greater immune response. The combination of these therapies with others, such as photothermal or tumor vaccine therapy, can result in a greater anticancer effect. Thus, the future of anticancer therapy aims to increase the effectiveness of therapy using various therapies in a synergistic combination rather than individually.
Collapse
Affiliation(s)
- Kyungeun Kim
- College of Medicine, Gachon University, Incheon 21999, South Korea
| | - Dongwoo Khang
- College of Medicine, Gachon University, Incheon 21999, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea.,Gachon Advanced Institute for Health Science & Technology (GAIHST), Gachon University, Incheon 21999, South Korea.,Department of Physiology, School of Medicine, Gachon University, Incheon 21999, South Korea
| |
Collapse
|
12
|
Gupta S, Singh I, Sharma AK, Kumar P. Ultrashort Peptide Self-Assembly: Front-Runners to Transport Drug and Gene Cargos. Front Bioeng Biotechnol 2020; 8:504. [PMID: 32548101 PMCID: PMC7273840 DOI: 10.3389/fbioe.2020.00504] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
The translational therapies to promote interaction between cell and signal come with stringent eligibility criteria. The chemically defined, hierarchically organized, and simpler yet blessed with robust intermolecular association, the peptides, are privileged to make the cut-off for sensing the cell-signal for biologics delivery and tissue engineering. The signature service and insoluble network formation of the peptide self-assemblies as hydrogels have drawn a spell of research activity among the scientists all around the globe in the past decades. The therapeutic peptide market players are anticipating promising growth opportunities due to the ample technological advancements in this field. The presence of the other organic moieties, enzyme substrates and well-established protecting groups like Fmoc and Boc etc., bring the best of both worlds. Since the large sequences of peptides severely limit the purification and their isolation, this article reviews the account of last 5 years' efforts on novel approaches for formulation and development of single molecule amino acids, ultra-short peptide self-assemblies (di- and tri- peptides only) and their derivatives as drug/gene carriers and tissue-engineering systems.
Collapse
Affiliation(s)
- Seema Gupta
- Chemistry Department, Acharya Narendra Dev College, University of Delhi, New Delhi, India
| | - Indu Singh
- Chemistry Department, Acharya Narendra Dev College, University of Delhi, New Delhi, India
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Ashwani K. Sharma
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Pradeep Kumar
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| |
Collapse
|