1
|
Singh A, Bora S, Kumar P, Kukreti R, Kaushik M. Targeted Nanotherapy by Vinblastine-Loaded Chitosan-Coated PLA Nanoparticles to Improve the Chemotherapy via Reactive Oxygen Species to Hamper Hepatocellular Carcinoma. ACS OMEGA 2025; 10:170-180. [PMID: 39829490 PMCID: PMC11739963 DOI: 10.1021/acsomega.4c02983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 01/22/2025]
Abstract
Liver cancer is a prevalent and significant cause of death in humans. The use of novel biodegradable materials for various biomedical applications is being recently recommended as complementary as well as alternative solution for traditional chemotherapy. This study focuses on the synthesis of biodegradable nanocarriers [chitosan-coated poly(lactic acid) NPs (Cht-PLA NPs)] for the delivery of an anticancer drug vinblastine (Vbx) and to evaluate its therapeutic potential in human hepatocellular carcinoma (HepG2) cells. The Cht-PLA NPs were synthesized using the nanoprecipitation method and characterized by transmission electron microscopy, scanning electron microscopy, Fourier transform infrared spectroscopy, dynamic light scattering, and zeta potential techniques. The results showed that the nanoparticle sizes are in the range of 100-200 nm with positive surface charge. The release profile of the synthesized nanoformulation showed controlled release of the Vbx drug for 72 h. The anticancer efficacy of the synthesized nanoformulation was assessed on the HepG2 cell lines. The in vitro cytotoxicity study revealed that the Vbx-loaded Cht-PLA NPs showed higher toxicity with an increase in concentration as compared to the Vbx alone. Additionally, an in vitro cellular uptake study revealed higher internalization as compared to the drug alone due to the chitosan coating. Further, the ability to stimulate the reactive oxygen species (ROS) generation and variation in mitochondrial membrane potential at the IC50 concentration of Vbx-loaded Cht-PLA NPs was confirmed by using 2,7-dichlorodihydrofluorescein diacetate and rhodamine 123 dyes, respectively, and were analyzed under fluorescence microscopy. Hence, the results showed that Vbx-loaded Cht-PLA NPs possess high anticancer activity due to its higher cellular toxicity, cellular uptake, increased ROS production, and disruption in mitochondrial membrane potential. All these properties of the synthesized nanoformulation suggest it's potential applications in drug delivery systems, targeting liver cancer.
Collapse
Affiliation(s)
- Amit Singh
- Nano-bioconjugate
Chemistry Lab, Cluster Innovation Centre, University of Delhi, Delhi 110007, India
- Department
of Chemistry, University of Delhi, Delhi 110007, India
| | - Shivangi Bora
- Genomics
and Molecular Medicine Unit, Institute of
Genomics and Integrative Biology (IGIB)-Council of Scientific and
Industrial Research (CSIR), Mall Road, Delhi 110007, India
| | - Pankaj Kumar
- Nano-bioconjugate
Chemistry Lab, Cluster Innovation Centre, University of Delhi, Delhi 110007, India
- Department
of Chemistry, University of Delhi, Delhi 110007, India
| | - Ritushree Kukreti
- Genomics
and Molecular Medicine Unit, Institute of
Genomics and Integrative Biology (IGIB)-Council of Scientific and
Industrial Research (CSIR), Mall Road, Delhi 110007, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Mahima Kaushik
- Nano-bioconjugate
Chemistry Lab, Cluster Innovation Centre, University of Delhi, Delhi 110007, India
| |
Collapse
|
2
|
Keshavarz Shahbaz S, Koushki K, Keshavarz Hedayati S, McCloskey AP, Kesharwani P, Naderi Y, Sahebkar A. Polymer nanotherapeutics: A promising approach toward microglial inhibition in neurodegenerative diseases. Med Res Rev 2024; 44:2793-2824. [PMID: 39031446 DOI: 10.1002/med.22064] [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: 04/13/2022] [Revised: 01/30/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
Nanoparticles (NPs) that target multiple transport mechanisms facilitate targeted delivery of active therapeutic agents to the central nervous system (CNS) and improve therapeutic transport and efficacy across the blood-brain barrier (BBB). CNS nanotherapeutics mostly target neurons and endothelial cells, however, microglial immune cells are the first line of defense against neuronal damage and brain infections. Through triggering release of inflammatory cytokines, chemokines and proteases, microglia can however precipitate neurological damage-a significant factor in neurodegenerative diseases. Thus, microglial inhibitory agents are attracting much attention among those researching and developing novel treatments for neurodegenerative disorders. The most established inhibitors of microglia investigated to date are resveratrol, curcumin, quercetin, and minocycline. Thus, there is great interest in developing novel agents that can bypass or easily cross the BBB. One such approach is the use of modified-nanocarriers as, or for, delivery of, therapeutic agents to the brain and wider CNS. For microglial inhibition, polymeric NPs are the preferred vehicles for choice. Here, we summarize the immunologic and neuroinflammatory role of microglia, established microglia inhibitor agents, challenges of CNS drug delivery, and the nanotherapeutics explored for microglia inhibition to date. We also discuss applications of the currently considered "most useful" polymeric NPs for microglial-inhibitor drug delivery in CNS-related diseases.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Khadije Koushki
- Department of Neurosurgery, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | | | - Alice P McCloskey
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Yazdan Naderi
- Department of Pharmacology, Faculty of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Liu D, Wang L, Li H, Li D, Zhou J, Wang J, Zhang Q, Cai D. Co-Delivery of Gemcitabine and Honokiol by Lipid Bilayer-Coated Mesoporous Silica Nanoparticles Enhances Pancreatic Cancer Therapy via Targeting Depletion of Tumor Stroma. Molecules 2024; 29:675. [PMID: 38338418 PMCID: PMC10856273 DOI: 10.3390/molecules29030675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Syndecan-1 (SDC1) modified lipid bilayer (LB)-coated mesoporous silica nanoparticles (MSN) to co-deliver gemcitabine (GEM) and honokiol (HNK) were prepared for the targeting treatment of pancreatic cancer. The encapsulation efficiencies of GEM and HNK in SDC1-LB-MSN-GEM/HNK were determined to be 60.3 ± 3.2% and 73.0 ± 1.1%. The targeting efficiency of SDC1-LB-MSN-GEM/HNK was investigated in BxPC-3 cells in vitro. The fluorescence intensity in the cells treated with SDC1-LB-MSN-Cou6 was 2-fold of LB-MSN-Cou6-treated cells, which was caused by SDC1/IGF1R-mediated endocytosis. As anticipated, its cytotoxicity was significantly increased. Furthermore, the mechanism was verified that SDC1-LB-MSN-HNK induced tumor cell apoptosis through the mitochondrial apoptosis pathway. Finally, the biodistribution, tumor growth inhibition, and preliminary safety studies were performed on BALB/c nude mice bearing BxPC-3 tumor models. The tumor growth inhibition index of SDC1-LB-MSN-GEM/HNK was 56.19%, which was 1.45-fold and 1.33-fold higher than that of the free GEM/HNK and LB-MSN-GEM/HNK treatment groups, respectively. As a result, SDC1-LB-MSN-GEM/HNK combined advantages of both GEM and HNK and simultaneously targeted and eliminated pancreatic cancerous and cancer-associated stromal cells. In summary, the present study demonstrated a new strategy of synergistic GEM and HNK to enhance the therapeutic effect of pancreatic cancer via the targeting depletion of tumor stroma.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qi Zhang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar 161006, China; (D.L.); (L.W.); (H.L.); (D.L.); (J.Z.); (J.W.)
| | - Defu Cai
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar 161006, China; (D.L.); (L.W.); (H.L.); (D.L.); (J.Z.); (J.W.)
| |
Collapse
|
4
|
Li W, Xiong X, Gong Y, Li Z. Preparation and In vitro Evaluation of Folated Pluronic F87/TPGS Co-modified Liposomes for Targeted Delivery of Curcumin. Curr Drug Deliv 2024; 21:592-602. [PMID: 37340749 DOI: 10.2174/1567201820666230619112502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/01/2023] [Accepted: 05/19/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND Using targeted liposomes to encapsulate and deliver drugs has become a hotspot in biomedical research. Folated Pluronic F87/D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) co-modified liposomes (FA-F87/TPGS-Lps) were fabricated for curcumin delivery, and intracellular targeting of liposomal curcumin was investigated. METHODS FA-F87 was synthesized and its structural characterization was conducted through dehydration condensation. Then, cur-FA-F87/TPGS-Lps were prepared via thin film dispersion method combined with DHPM technique, and their physicochemical properties and cytotoxicity were determined. Finally, the intracellular distribution of cur-FA-F87/TPGS-Lps was investigated using MCF-7 cells. RESULTS Incorporation of TPGS in liposomes reduced their particle size, but increased the negative charge of the liposomes as well as their storage stability, and the encapsulation efficiency of curcumin was improved. While, modification of liposomes with FA increased their particle size, and had no impact on the encapsulation efficiency of curcumin in liposomes. Among all the liposomes (cur-F87-Lps, cur-FA-F87-Lps, cur-FA-F87/TPGS-Lps and cur-F87/TPGS-Lps), cur-FA-F87/TPGS-Lps showed highest cytotoxicity to MCF-7 cells. Moreover, cur-FA-F87/TPGS-Lps was found to deliver curcumin into the cytoplasm of MCF-7 cells. CONCLUSION Folate-Pluronic F87/TPGS co-modified liposomes provide a novel strategy for drug loading and targeted delivery.
Collapse
Affiliation(s)
- Wenjuan Li
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, People's Republic of China
| | - Xiangyuan Xiong
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, People's Republic of China
| | - Yanchun Gong
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, People's Republic of China
| | - Ziling Li
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, People's Republic of China
| |
Collapse
|
5
|
Rashid B, Sridewi N, Anwar A, Shahabbudin S, Mon AA. A review on human cancer and potential role of MXenes in cancer therapy. E3S WEB OF CONFERENCES 2024; 488:03021. [DOI: 10.1051/e3sconf/202448803021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Cancer is the second leading cause of death worldwide and is having a serious impact on the global economy. Various treatment modalities are in use to treat cancer but none of the techniques is risk-free. Recently, various nanomaterials such as gold, boron, and other compounds have been investigated for radiotherapy and as anti-cancer drug carriers with promising results. MXenes are 2D novel nanomaterials and their biomedical and anticancer properties are gaining interest due to their high biomedical activity, less bio-toxicity, and photo-responsive nature. However, the biological properties of MXense have not been studied extensively, therefore, limited data is published on its in-vitro and in-vivo anticancer activities, drug loading efficacy, targeted release, and on its photothermal therapy response. In this review, we have discussed the use of nanoparticles and MXenen nanomaterial in cancer therapy. Furthermore, the role of Mxene as a photothermal agent and drug carrier has also been emphasized, along with the present challenges for the use of nanomaterials in the treatment of cancer.
Collapse
|
6
|
Chen S, Wang Z, Liu L, Li Y, Ni X, Yuan H, Wang C. Redox homeostasis modulation using theranostic AIE nanoparticles results in positive-feedback drug accumulation and enhanced drug penetration to combat drug-resistant cancer. Mater Today Bio 2022; 16:100396. [PMID: 36060105 PMCID: PMC9434132 DOI: 10.1016/j.mtbio.2022.100396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
Drug-resistant cancers usually have multiple barriers to compromise the effect of therapies, of which multidrug-resistance (MDR) phenotype as the intracellular barrier and dense tumor matrix as the extracellular barrier, significantly contribute to the poor anticancer performance of current drug delivery systems (DDS). Here in this study, we fabricated a novel aggregation-induced emission (AIE)-active polymer capable of self-assembling into ultrasmall nanoparticles (∼20 nm) with D-alpha Tocopheryl Polyethylene Glycol Succinate (TPGS), for dual-encapsulating of doxorubicin (Dox) and sulforaphane (SFN) (AT/Dox/SFN). It revealed that redox homeostasis modulation of MDR cells (MCF-7/Adr) using AT/Dox/SFN can trigger mitochondria damage and ATP deficiency, which reverse the MDR phenotype of MCF-7/Adr cells to afford enhanced cellular uptake of both drug and DDS in a positive-feedback manner. The enhanced cellular drug accumulation further initiates the “neighboring effect” for improved drug penetration. Using this strategy, the growth of in vivo MCF-7/Adr tumors can be effectively inhibited at a low dosage (1/5) of doxorubicin (Dox) as compared to free Dox. In summary, we offer a new approach to overcome both the intracellular and extracellular barriers of drug-resistant cancers and elucidate the potential action mechanisms, which are beneficial for better cancer management. Redox homeostasis modulation in MDR cancer cell results in positive-feedback drug accumulation and enhanced drug penetration. Mitochondria damage and neighboring effect is responsible for MDR reversal and enhanced drug penetration, respectively. AT/Dox/SFN effectively inhibits in vivo MCF-7/Adr tumors at a low dosage (1/5) of doxorubicin (Dox) as compared to free Dox.
Collapse
Affiliation(s)
- Shaoqing Chen
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu 213003, China
| | - Ziyu Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, China
| | - Li Liu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
| | - Yuting Li
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
| | - Xinye Ni
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu 213003, China
- Corresponding author. Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China.
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang, China
- Corresponding author.
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
- Corresponding author.
| |
Collapse
|
7
|
Macedo LDO, Morales IA, Barbosa EJ, Stephano MA, de Araujo GL, Bou-Chacra NA. Thermal study, process optimization, and water solubility improvement of a freeze-dried artemether nanosuspension for malaria treatment. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
8
|
Effects of Sorafenib and Quercetin Alone or in Combination in Treating Hepatocellular Carcinoma: In Vitro and In Vivo Approaches. Molecules 2022; 27:molecules27228082. [PMID: 36432184 PMCID: PMC9697794 DOI: 10.3390/molecules27228082] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Sorafenib is the first drug approved to treat advanced hepatocellular carcinoma (HCC) and continues as the gold-standard therapy against HCC. However, acquired drug resistance represents a main concern about sorafenib therapy. The flavanol quercetin found in plants has shown great anti-cancer and anti-inflammatory properties. In this work, quercetin was used as a therapeutic agent alone or in combination with a sorafenib chemotherapy drug to improve the routine HCC treatment with sorafenib. The in vitro and in vivo results presented here confirm that quercetin alone or in combination with sorafenib significantly inhibited HCC growth, induced cell cycle arrest and induced apoptosis and necrosis. Further molecular data shown in this report demonstrate that quercetin alone or combined with sorafenib downregulated key inflammatory, proliferative and angiogenesis-related genes (TNF-α, VEGF, P53 and NF-κB). Combined quercetin/sorafenib treatment markedly improved the morphology of the induced liver damage and showed significant antioxidant and anti-tumor effects. The advantage of combined treatment efficacy reported here can be attributed to quercetin's prominent effects in modulating cell cycle arrest, apoptosis, oxidative stress and inflammation.
Collapse
|
9
|
Li X, Hu L, Tan C, Wang X, Ran Q, Chen L, Li Z. Platelet-promoting drug delivery efficiency for inhibition of tumor growth, metastasis, and recurrence. Front Oncol 2022; 12:983874. [PMID: 36276066 PMCID: PMC9582853 DOI: 10.3389/fonc.2022.983874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Nanomedicines are considered one of the promising strategies for anticancer therapy; however, the low targeting efficiency of nanomedicines in vivo is a great obstacle to their clinical applications. Camouflaging nanomedicines with either platelet membrane (PM) or platelet would significantly prolong the retention time of nanomedicines in the bloodstream, enhance the targeting ability of nanomedicines to tumor cells, and reduce the off-target effect of nanomedicines in major organs during the anticancer treatment. In the current review, the advantages of using PM or platelet as smart carriers for delivering nanomedicines to inhibit tumor growth, metastasis, and recurrence were summarized. The opportunities and challenges of this camouflaging strategy for anticancer treatment were also discussed.
Collapse
Affiliation(s)
- Xiaoliang Li
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Lanyue Hu
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Chengning Tan
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Xiaojie Wang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Qian Ran
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Li Chen
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- *Correspondence: Li Chen, ; Zhongjun Li,
| | - Zhongjun Li
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma, Burn and Combined Injuries, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- *Correspondence: Li Chen, ; Zhongjun Li,
| |
Collapse
|
10
|
Controlled release and targeted drug delivery with poly(lactic-co-glycolic acid) nanoparticles: reviewing two decades of research. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00584-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
11
|
Arcusa R, Villaño D, Marhuenda J, Cano M, Cerdà B, Zafrilla P. Potential Role of Ginger (Zingiber officinale Roscoe) in the Prevention of Neurodegenerative Diseases. Front Nutr 2022; 9:809621. [PMID: 35369082 PMCID: PMC8971783 DOI: 10.3389/fnut.2022.809621] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/15/2022] [Indexed: 12/30/2022] Open
Abstract
Ginger is composed of multiple bioactive compounds, including 6-gingerol, 6-shogaol, 10-gingerol, gingerdiones, gingerdiols, paradols, 6-dehydrogingerols, 5-acetoxy-6-gingerol, 3,5-diacetoxy-6-gingerdiol, and 12-gingerol, that contribute to its recognized biological activities. Among them, the major active compounds are 6-shogaol and 6-gingerol. Scientific evidence supports the beneficial properties of ginger, including antioxidant and anti-inflammatory capacities and in contrast, a specific and less studied bioactivity is the possible neuroprotective effect. The increase in life expectancy has raised the incidence of neurodegenerative diseases (NDs), which present common neuropathological features as increased oxidative stress, neuroinflammation and protein misfolding. The structure-activity relationships of ginger phytochemicals show that ginger can be a candidate to treat NDs by targeting different ligand sites. Its bioactive compounds may improve neurological symptoms and pathological conditions by modulating cell death or cell survival signaling molecules. The cognitive enhancing effects of ginger might be partly explained via alteration of both the monoamine and the cholinergic systems in various brain areas. Moreover, ginger decreases the production of inflammatory related factors. The aim of the present review is to summarize the effects of ginger in the prevention of major neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and multiple sclerosis.
Collapse
|
12
|
Zuo H, Qiang J, Wang Y, Wang R, Wang G, Chai L, Ren G, Zhao Y, Zhang G, Zhang S. Design of red blood cell membrane-cloaked dihydroartemisinin nanoparticles with enhanced antimalarial efficacy. Int J Pharm 2022; 618:121665. [PMID: 35288223 DOI: 10.1016/j.ijpharm.2022.121665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/06/2022] [Accepted: 03/10/2022] [Indexed: 01/06/2023]
Abstract
Targeting delivery and prolonging action duration of artemisinin drugs are effective strategies for improving antimalarial treatment outcomes. Here, dihydroartemisinin (DHA) loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles (PDNs) were prepared and further cloaked with red blood cell (RBC) membranes via electrostatic interactions to yield RBC membrane-cloaked PDNs (RPDNs). The prepared RPDNs displayed a notable "core-shell" structure, with a negative surface charge of -29.2 ± 4.19 mV, a relatively uniform size distribution (86.4 ± 2.54 nm, polydispersity index of 0.179 ± 0.011), an average encapsulation efficiency (70.1 ± 0.79%), and a 24-h sustained-release behavior in vitro. Compared with PDNs, RPDNs showed markedly decreased phagocytic activity by RAW 264.7 cells and had prolonged blood circulation duration. The Pearson correlation coefficient of RPDNs distribution in infected red blood cells (iRBCs) was 0.7173, suggesting that RPDNs could effectively target Plasmodium-iRBCs. In PyBy265-infected mice, RPDNs showed a higher inhibition ratio (88.39 ± 2.69%) than PDNs (83.13 ± 2.12%) or DHA (58.74 ± 3.78%), at the same dose of 8.8 μmol/kg. The ED90 of RPDNs (8.13 ± 0.18 μmol/kg) was substantially lower than that of PDNs (14.48 ± 0.23 μmol/kg) and DHA (17.67 ± 3.38 μmol/kg). Furthermore, no apparent abnormalities were detected in routine blood examination, liver function indexes, and pathological analysis of tissue sections of PyBy265-infected mice following RPDNs treatment. In conclusion, the prepared RPDNs exhibited enhanced antimalarial efficacy, prolonged circulation, targeted delivery to Plasmodium-iRBCs, and satisfactory biocompatibility.
Collapse
Affiliation(s)
- Hengtong Zuo
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Jihong Qiang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Yidan Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Rongrong Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Geng Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Liqing Chai
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Guolian Ren
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Yongdan Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Guoshun Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Shuqiu Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
13
|
Le Z, He Z, Liu H, Liu L, Liu Z, Chen Y. Antioxidant Enzymes Sequestered within Lipid-Polymer Hybrid Nanoparticles for the Local Treatment of Inflammatory Bowel Disease. ACS APPLIED MATERIALS & INTERFACES 2021; 13:55966-55977. [PMID: 34792322 DOI: 10.1021/acsami.1c19457] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The local treatment of inflammatory bowel disease (IBD) by enzyme therapeutics is challenging owing to hostile environments in the gastrointestinal tract, leading to the hydrolysis and enzymatic degradation of labile proteins. In this study, safe and efficient local drug delivery systems were developed by antioxidant superoxide dismutase (SOD) sequestered within lipid-polymer hybrid nanoparticles through sequential self-assembly processes. Interestingly, we found that the sequestered SOD exhibited long-term enzymatic stability and comparable biological activity to the enzymes in the native form, probably owing to particle encapsulation providing a physical barrier to prevent the enzymolysis of proteins. We demonstrated that nanoparticle-based local drug delivery systems showed excellent mucus-penetrating ability and inflammation-targeting properties, owing to the particle surface with a poly(ethylene glycol) (PEG) coating and folate functionalization, thus improving mucosal retention time and drug delivery efficiency within the colorectal region. Furthermore, SOD-containing lipid-polymer hybrid nanoparticles could effectively mitigate inflammatory responses by regulating the secretion of inflammation-associated cytokines, thus increasing therapeutic outcomes in colitis mice through intrarectal administration. The findings indicated that antioxidant enzymes sequestered within lipid-polymer hybrid nanoparticles might be potential enzyme therapeutics for the local treatment of some inflammatory diseases in the near future.
Collapse
Affiliation(s)
- Zhicheng Le
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Functional Biomaterials Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510006, China
| | - Zepeng He
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Functional Biomaterials Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510006, China
| | - Hong Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Functional Biomaterials Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510006, China
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Functional Biomaterials Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhijia Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Functional Biomaterials Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510006, China
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Functional Biomaterials Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
14
|
Raspantini GL, Luiz MT, Abriata JP, Eloy JDO, Vaidergorn MM, Emery FDS, Marchetti JM. PCL-TPGS polymeric nanoparticles for docetaxel delivery to prostate cancer: Development, physicochemical and biological characterization. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Wang Y, Cong H, Wang S, Yu B, Shen Y. Development and application of ultrasound contrast agents in biomedicine. J Mater Chem B 2021; 9:7633-7661. [PMID: 34586124 DOI: 10.1039/d1tb00850a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
With the rapid development of molecular imaging, ultrasound (US) medicine has evolved from traditional imaging diagnosis to integrated diagnosis and treatment at the molecular level. Ultrasound contrast agents (UCAs) play a crucial role in the integration of US diagnosis and treatment. As the micro-bubbles (MBs) in UCAs can enhance the cavitation effect and promote the biological effect of US, UCAs have also been studied in the fields of US thrombolysis, mediated gene transfer, drug delivery, and high intensity focused US. The application range of UCAs is expanding, and the value of their applications is improving. This paper reviews the development and application of UCAs in biomedicine in recent years, and the existing problems and prospects are pointed out.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China.
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China. .,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Song Wang
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China.
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, 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 Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China. .,Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
16
|
Famta P, Shah S, Chatterjee E, Singh H, Dey B, Guru SK, Singh SB, Srivastava S. Exploring new Horizons in overcoming P-glycoprotein-mediated multidrug-resistant breast cancer via nanoscale drug delivery platforms. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100054. [PMID: 34909680 PMCID: PMC8663938 DOI: 10.1016/j.crphar.2021.100054] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/19/2022] Open
Abstract
The high probability (13%) of women developing breast cancer in their lifetimes in America is exacerbated by the emergence of multidrug resistance after exposure to first-line chemotherapeutic agents. Permeation glycoprotein (P-gp)-mediated drug efflux is widely recognized as the major driver of this resistance. Initial in vitro and in vivo investigations of the co-delivery of chemotherapeutic agents and P-gp inhibitors have yielded satisfactory results; however, these results have not translated to clinical settings. The systemic delivery of multiple agents causes adverse effects and drug-drug interactions, and diminishes patient compliance. Nanocarrier-based site-specific delivery has recently gained substantial attention among researchers for its promise in circumventing the pitfalls associated with conventional therapy. In this review article, we focus on nanocarrier-based co-delivery approaches encompassing a wide range of P-gp inhibitors along with chemotherapeutic agents. We discuss the contributions of active targeting and stimuli responsive systems in imparting site-specific cytotoxicity and reducing both the dose and adverse effects.
Collapse
Affiliation(s)
- Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Essha Chatterjee
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Hoshiyar Singh
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Biswajit Dey
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Santosh Kumar Guru
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Shashi Bala Singh
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
17
|
Xiao J, Yan M, Zhou K, Chen H, Xu Z, Gan Y, Hong B, Tian G, Qian J, Zhang G, Wu Z. A nanoselenium-coating biomimetic cytomembrane nanoplatform for mitochondrial targeted chemotherapy- and chemodynamic therapy through manganese and doxorubicin codelivery. J Nanobiotechnology 2021; 19:227. [PMID: 34330298 PMCID: PMC8325191 DOI: 10.1186/s12951-021-00971-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/23/2021] [Indexed: 01/15/2023] Open
Abstract
The cell membrane is widely considered as a promising delivery nanocarrier due to its excellent properties. In this study, self-assembled Pseudomonas geniculate cell membranes were prepared with high yield as drug nanocarriers, and named BMMPs. BMMPs showed excellent biosafety, and could be more efficiently internalized by cancer cells than traditional red cell membrane nanocarriers, indicating that BMMPs could deliver more drug into cancer cells. Subsequently, the BMMPs were coated with nanoselenium (Se), and subsequently loaded with Mn2+ ions and doxorubicin (DOX) to fabricate a functional nanoplatform (BMMP-Mn2+/Se/DOX). Notably, in this nanoplatform, Se nanoparticles activated superoxide dismutase-1 (SOD-1) expression and subsequently up-regulated downstream H2O2 levels. Next, the released Mn2+ ions catalyzed H2O2 to highly toxic hydroxyl radicals (·OH), inducing mitochondrial damage. In addition, the BMMP-Mn2+/Se nanoplatform inhibited glutathione peroxidase 4 (GPX4) expression and further accelerated intracellular reactive oxygen species (ROS) generation. Notably, the BMMP-Mn2+/Se/DOX nanoplatform exhibited increased effectiveness in inducing cancer cell death through mitochondrial and nuclear targeting dual-mode therapeutic pathways and showed negligible toxicity to normal organs. Therefore, this nanoplatform may represent a promising drug delivery system for achieving a safe, effective, and accurate cancer therapeutic plan.
Collapse
Affiliation(s)
- Jianmin Xiao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Miao Yan
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Ke Zhou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Hui Chen
- Department of Dental Implant Center, Key Laboratory of Oral Diseases Research of Anhui Province, Stomatologic Hospital & College, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Zhaowei Xu
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Yuehao Gan
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Biao Hong
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.,University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Geng Tian
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Junchao Qian
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| | - Guilong Zhang
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, People's Republic of China.
| | - Zhengyan Wu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| |
Collapse
|
18
|
Liu T, Jing F, Huang P, Geng Z, Xu J, Li J, Chen D, Zhu Y, Wang Z, Huang W, Chen C. Thymopentin alleviates premature ovarian failure in mice by activating YY2/Lin28A and inhibiting the expression of let-7 family microRNAs. Cell Prolif 2021; 54:e13089. [PMID: 34180104 PMCID: PMC8349654 DOI: 10.1111/cpr.13089] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Thymopentin (5TP) significantly improved typical murine premature ovarian failure (POF) symptoms induced by a high‐fat and high‐sugar (HFHS) diet. However, its effect and mechanism remain unclear. Materials and methods RNA‐Seq was used to detect the differentially expressed genes among each group. HFHS‐induced POF mouse model was generated and injected with siRNA using Poly (lactic‐co‐glycolic acid) (PLGA) as a carrier. Results RNA‐Seq suggested that 5TP promoted the expression of Yin Yang 2 (YY2) in mouse ovarian granulosa cell (mOGC) of HFHS‐POF mice. Luciferase reporter assay indicated that 5TP promoted the binding of YY2 to the specific sequence C(C/T)AT(G/C)(G/T) on the Lin28A promoter and promoted Lin28A transcription and expression. We continuously injected PLGA‐cross‐linked siRNA nanoparticles targeting YY2 into HFHS‐POF mice (siYY2@PLGA), which significantly reduced the therapeutic effect of 5TP. siYY2@PLGA injection also significantly attenuated the upregulation of Lin28a expression in mOGCs induced by 5TP and enhanced the expression of let‐7 family microRNAs, thereby inhibiting the proliferation and division of mOGCs. qPCR results showed that there was a significant difference in the expression levels of exosome‐derived Yy2 mRNAs between POF patients and normal women, and that there was a specific correlation between the expression level of exosome‐derived Yy2 and the peripheral concentrations of the blood hormones pregnenolone, progesterone and oestradiol. Conclusions Thymopentin promotes the transcriptional activation of Lin28A via stimulating transcription factor YY2 expression, inhibits the activity of let‐7 family microRNAs and alleviates the ageing of ovarian granulosa cells, ultimately achieving a therapeutic effect on POF in mice.
Collapse
Affiliation(s)
- Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fangyuan Jing
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Zixiang Geng
- Department of Acupuncture, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianghong Xu
- Department of gynaecology, Jingan Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Jiahui Li
- Department of gynaecology, Jingan Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Danping Chen
- Department of gynaecology, Jingan Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Ying Zhu
- Department of gynaecology, Jingan Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Zhenxin Wang
- Department of Laboratory Medicine of Zhongshan Hospital and institute of Biomedical Science, Fudan University, Shanghai, China
| | - Willian Huang
- Hainan Zhonghe Pharmaceutical Co., Ltd, Haikou, China
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
19
|
Li H, Li M, Fu J, Ao H, Wang W, Wang X. Enhancement of oral bioavailability of quercetin by metabolic inhibitory nanosuspensions compared to conventional nanosuspensions. Drug Deliv 2021; 28:1226-1236. [PMID: 34142631 PMCID: PMC8218931 DOI: 10.1080/10717544.2021.1927244] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Quercetin-loaded nanosuspensions (Que-NSps) added metabolic inhibitors were evaluated as drug delivery system to promote the oral bioavailability of quercetin. Que-NSps were prepared respectively using d-alpha tocopherol acid polyethylene glycol succinate (TPGS) or Soybean Lecithin (SPC) as stabilizer. On the basis, Piperine (Pip) or sodium oleate (SO) was, respectively, encapsulated in Que-NSps as phase II metabolic inhibitors. The resulting Que-NSps all displayed a mean particle size of about 200 nm and drug loading content was in the range of 22.3–27.8%. The release of quercetin from Que-NSps was slow and sustained. After oral administration of 50 mg/kg different Que-NSps, the levels of free quercetin in plasma were significantly promoted, the concentration of quercetin metabolites (isorhamnetin and quercetin 3-O-β-d-Glucuronide) were decreased. The absolute bioavailability was, respectively 15.55%, 6.93%, 12.38%, and 23.58% for TPGS-Que-NSps, TPGS-SO-Que-NSps, SPC-Que-NSps, and SPC-Pip-Que-NSps, and 3.61% for quercetin water suspension. SPC-Pip-Que-NSps turned out to an ideal nanocarrier combined nano drug delivery system together with metabolic inhibitor to promote oral absorption of quercetin.
Collapse
Affiliation(s)
- Haowen Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing, PR China
| | - Manzhen Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing, PR China
| | - Jingxin Fu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing, PR China
| | - Hui Ao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing, PR China
| | - Weihua Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing, PR China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Haidian District, Beijing, PR China
| |
Collapse
|
20
|
Cheng F, Pan Q, Gao W, Pu Y, Luo K, He B. Reversing Chemotherapy Resistance by a Synergy between Lysosomal pH-Activated Mitochondrial Drug Delivery and Erlotinib-Mediated Drug Efflux Inhibition. ACS APPLIED MATERIALS & INTERFACES 2021; 13:29257-29268. [PMID: 34130450 DOI: 10.1021/acsami.1c03196] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mitochondrial drug delivery has attracted increasing attention in various mitochondrial dysfunction-associated disorders such as cancer owing to the important role of energy production. Herein, we report a lysosomal pH-activated mitochondrial-targeting polymer nanoparticle to overcome drug resistance by a synergy between mitochondrial delivery of doxorubicin (DOX, an anticancer drug) and erlotinib-mediated inhibition of drug efflux. The obtained nanoparticles, DE-NPs could maintain negative charge and have long blood circulation while undergoing charge reversal at lysosomal pH after internalization by cancer cells. Thereafter, the acidity-activated polycationic and hydrophobic polypeptide domains boost lysosomal escape and mitochondrial-targeting drug delivery, leading to mitochondrial dysfunction, ATP suppression, and cell apoptosis. Moreover, the suppressed ATP supply and erlotinib enabled dual inhibition of drug efflux by DOX-resistant MCF-7/ADR cells, leading to significantly augmented intracellular DOX accumulation and a synergistic anticancer effect with a 17-fold decrease of IC50 relative to DOX. In vivo antitumor study demonstrates that DE-NPs efficiently suppressed the tumor burden in MCF-7/ADR tumor-bearing mice and led to negligible toxicity. This work establishes that a combination of mitochondrial drug delivery and drug efflux inhibition could be a promising strategy for combating multidrug resistance.
Collapse
Affiliation(s)
- Furong Cheng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Center for Translational Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
- Department of Pharmaceutics, College of Pharmacy, Virginia Commonwealth University, Richmond 23219, Virginia, United States
| | - Qingqing Pan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Kui Luo
- Huaxi MR Research Center, Department of Radiology, West China Hospital, Functional and Molecular Imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
21
|
Li K, Pang L, Pan X, Fan S, Wang X, Wang Q, Dai P, Gao W, Gao J. GE11 Modified PLGA/TPGS Nanoparticles Targeting Delivery of Salinomycin to Breast Cancer Cells. Technol Cancer Res Treat 2021; 20:15330338211004954. [PMID: 34056977 PMCID: PMC8182624 DOI: 10.1177/15330338211004954] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Salinomycin (Sal) is a potent inhibitor with effective anti-breast cancer properties in clinical therapy. The occurrence of various side effect of Sal greatly limits its application. The epidermal growth factor receptor (EGFR) family is a family of receptors highly expressed in most breast cancer cells. GE11 is a dodecapeptide which shows excellent EGFR affinity. A series of nanoparticles derivatives with GE11 peptide conjugated PLGA/TPGS were synthesized. Nanoprecipitation method was used to prepare the Sal loaded nanoparticles at the optimized concentration. The characterization, targeting efficacy, and antitumor activity were detected both in vitro and in vivo. Encapsulation of Sal in GE11 modified PLGA/TPGS nanoparticles shows an improved therapy efficacy and lower systemic side effect. This represents the delivery system a promising strategy to enhance the therapeutic effect against EGFR highly expressed breast cancer.
Collapse
Affiliation(s)
- Kaichun Li
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Liying Pang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xiaorong Pan
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Shaonan Fan
- Department of Radiation Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Xinxin Wang
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Qiaoyun Wang
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Ping Dai
- Department of Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Wei Gao
- Department of Radiation Oncology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Jie Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
22
|
Chen SH, Liu TI, Chuang CL, Chen HH, Chiang WH, Chiu HC. Alendronate/folic acid-decorated polymeric nanoparticles for hierarchically targetable chemotherapy against bone metastatic breast cancer. J Mater Chem B 2021; 8:3789-3800. [PMID: 32150202 DOI: 10.1039/d0tb00046a] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To considerably enhance treatment efficacy for bone metastatic breast cancer via dual bone/tumor-targeted chemotherapy, a nanoparticle-based delivery system comprising poly(lactic-co-glycolic acid) (PLGA) as the hydrophobic core coated with alendronate-modified d-α-tocopheryl polyethylene glycol succinate (ALN-TPGS) and folic acid-conjugated TPGS (FA-TPGS) was developed as a vehicle for paclitaxel (PTX) in this work. The ALN/FA-decorated nanoparticles not only showed superior ALN-mediated binding affinity for hydroxyapatite abundant in bone tissue but also promoted uptake of payloads by folate receptor-overexpressing cancer cells to significantly augment PTX cytotoxicity. Notably, through dual-targetable delivery to the bone matrix and folate receptor-overexpressing 4T1 tumors, the PTX-loaded nanoparticles substantially accumulated in bone metastases in vivo and inhibited 4T1 tumor growth and lung metastasis, leading to significant improvement of the survival rate of treated mice. Upon treatment with the ALN/FA-decorated PTX-loaded nanoparticles, the bone destruction and bone loss of the tumor-bearing mice were appreciably retarded, and the adverse effects on normal tissues were alleviated. These results demonstrate that the ALN/FA-decorated PTX-loaded delivery system developed in this study shows great promise for the effective treatment of bone metastatic breast cancer.
Collapse
Affiliation(s)
- Shih-Hong Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan. and Department of Anesthesiology, Taipei Tzu Chi Hospital, New Taipei City 231, Taiwan and Department of Anesthesiology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Te-I Liu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Cheng-Lin Chuang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Hsin-Hung Chen
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan.
| | - Wen-Hsuan Chiang
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan.
| | - Hsin-Cheng Chiu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
23
|
Wei X, Song M, Li W, Huang J, Yang G, Wang Y. Multifunctional nanoplatforms co-delivering combinatorial dual-drug for eliminating cancer multidrug resistance. Am J Cancer Res 2021; 11:6334-6354. [PMID: 33995661 PMCID: PMC8120214 DOI: 10.7150/thno.59342] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/26/2021] [Indexed: 02/05/2023] Open
Abstract
Clinically, the primary cause of chemotherapy failure belongs to the occurrence of cancer multidrug resistance (MDR), which directly leads to the recurrence and metastasis of cancer along with high mortality. More and more attention has been paid to multifunctional nanoplatform-based dual-therapeutic combination to eliminate resistant cancers. In addition to helping both cargoes improve hydrophobicity and pharmacokinetic properties, increase bioavailability, release on demand and enhance therapeutic efficacy with low toxic effects, these smart co-delivery nanocarriers can even overcome drug resistance. Here, this review will not only present different types of co-delivery nanocarriers, but also summarize targeted and stimuli-responsive combination nanomedicines. Furthermore, we will focus on the recent progress in the co-delivery of dual-drug using such intelligent nanocarriers for surmounting cancer MDR. Whereas it remains to be seriously considered that there are some knotty issues in the fight against MDR of cancers via using co-delivery nanoplatforms, including limited intratumoral retention, the possible changes of combinatorial ratio under complex biological environments, drug release sequence from the nanocarriers, and subsequent free-drug resistance after detachment from the nanocarriers. It is hoped that, with the advantage of continuously developing nanomaterials, two personalized therapeutic agents in combination can be better exploited to achieve the goal of cooperatively combating cancer MDR, thus advancing the time to clinical transformation.
Collapse
|
24
|
Wei G, Wang Y, Yang G, Wang Y, Ju R. Recent progress in nanomedicine for enhanced cancer chemotherapy. Theranostics 2021; 11:6370-6392. [PMID: 33995663 PMCID: PMC8120226 DOI: 10.7150/thno.57828] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/31/2021] [Indexed: 12/24/2022] Open
Abstract
As one of the most important cancer treatment strategies, conventional chemotherapy has substantial side effects and leads easily to cancer treatment failure. Therefore, exploring and developing more efficient methods to enhance cancer chemotherapy is an urgently important problem that must be solved. With the development of nanotechnology, nanomedicine has showed a good application prospect in improving cancer chemotherapy. In this review, we aim to present a discussion on the significant research progress in nanomedicine for enhanced cancer chemotherapy. First, increased enrichment of drugs in tumor tissues relying on different targeting ligands and promoting tissue penetration are summarized. Second, specific subcellular organelle-targeted chemotherapy is discussed. Next, different combinational strategies to reverse multidrug resistance (MDR) and improve the effective intracellular concentration of therapeutics are discussed. Furthermore, the advantages of combination therapy for cancer treatment are emphasized. Finally, we discuss the major problems facing therapeutic nanomedicine for cancer chemotherapy, and propose possible future directions in this field.
Collapse
Affiliation(s)
- Guoqing Wei
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Yu Wang
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Guang Yang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Yi Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Rong Ju
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| |
Collapse
|
25
|
Zhang J, Deng M, Shi X, Zhang C, Qu X, Hu X, Wang W, Kong D, Huang P. Cascaded amplification of intracellular oxidative stress and reversion of multidrug resistance by nitric oxide prodrug based-supramolecular hydrogel for synergistic cancer chemotherapy. Bioact Mater 2021; 6:3300-3313. [PMID: 33778206 PMCID: PMC7970318 DOI: 10.1016/j.bioactmat.2021.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/24/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
Supramolecular hydrogel was facilely developed by self-assembly of NO prodrug conjugated hydrogelator sequence. The locoregionally sustained NO release from the hydrogel could be triggered by intracellular over-expressed GSH/GST. NO could effectively reverse the P-gp mediated MDR effect and facilitate the intracellular accumulation of DOX. This type of stimuli-sensitive NO delivery platform holds great potential for combating drug-resistance cancer.
Collapse
Affiliation(s)
- Jimin Zhang
- Hebei Key Laboratory of Functional Polymers, National-Local Joint Engineering Laboratory for Energy Conservation of Chemical Process Integration and Resources Utilization, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Meigui Deng
- Hebei Key Laboratory of Functional Polymers, National-Local Joint Engineering Laboratory for Energy Conservation of Chemical Process Integration and Resources Utilization, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Xiaoguang Shi
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Xiongwei Qu
- Hebei Key Laboratory of Functional Polymers, National-Local Joint Engineering Laboratory for Energy Conservation of Chemical Process Integration and Resources Utilization, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Xiuli Hu
- Hebei Key Laboratory of Functional Polymers, National-Local Joint Engineering Laboratory for Energy Conservation of Chemical Process Integration and Resources Utilization, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
26
|
Vitamin E succinate with multiple functions: A versatile agent in nanomedicine-based cancer therapy and its delivery strategies. Int J Pharm 2021; 600:120457. [PMID: 33676991 DOI: 10.1016/j.ijpharm.2021.120457] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 11/20/2022]
Abstract
Vitamin E succinate (VES), a succinic acid ester of vitamin E, is one of the most effective anticancer compounds of the vitamin E family. VES can inhibit tumor growth by multiple pathways mainly involve tumor proliferation inhibition, apoptosis induction, and metastasis prevention. More importantly, the mitochondrial targeting and damaging property of VES endows it with great potential in exhibiting synergetic effect with conventional chemotherapeutic drugs and overcoming multidrug resistance (MDR). Given the lipophilicity of VES that hinders its bioavailability and therapeutic activity, nanotechnology with multiple advantages has been widely explored to deliver VES and opened up new avenues for its in vivo application. This review aims to introduce the anticancer mechanisms of VES and summarize its delivery strategies using nano-drug delivery systems. Specifically, VES-based combination therapy for synergetic anticancer effect, MDR-reversal, and oral chemotherapy improvement are highlighted. Finally, the challenges and perspectives are discussed.
Collapse
|
27
|
Wang C, Wei X, Shao G. Functional Doxorubicin-Loaded Omega-3 Unsaturated Fatty Acids Nanoparticles in Reversing Hepatocellular Carcinoma Multidrug Resistance. Med Sci Monit 2021; 27:e927727. [PMID: 33524008 PMCID: PMC7863563 DOI: 10.12659/msm.927727] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background This study investigated a nanoparticle drug delivery system to reverse multidrug resistance (MDR) and assessed its anticancer efficacy in hepatocellular carcinoma (HCC). Material/Methods Docosahexaenoic acid (DHA) was used as the functional excipient and doxorubicin (DOX) as the chemotherapeutic drug to synthesize DOX nanoparticles (DOX-nano). The human HCC cell line HepG2 was used for experiments. HepG2/DOX, HepG2+DOX, HepG2+DOX-nano, HepG2/DOX+DOX, and HepG2/DOX+DOX-nano groups cells were treated with DOX or DOX-nano (5 μg/mL). Nude mice bearing a HepG2/DOX xenograft were divided into model, DOX, vector-nano, and DOX-nano groups and injected with saline, DOX reagent, vector-nano, and DOX-nano (2 mg/kg), respectively. Next, cytotoxicity, cellular uptake, cell apoptosis and migration, fluorescence imaging, TUNEL assay, and tumor inhibition effects were assessed in vitro and in vivo. Furthermore, expression of MDR-related proteins was also detected using western blotting. Results Fluorescence imaging showed that the DOX uptake in the DOX-nano-treated group was the strongest in the HCC cells or tumors. Cell apoptosis was significantly increased in DOX-nano-treated HepG2/DOX cells and tumors, and cell migration was significantly inhibited in the DOX-nano-treated HepG2/DOX cells compared with the other groups. The tumor inhibitory rate in DOX-nano-injected tumors was also significantly higher than in other groups. The expression of breast cancer resistance protein, B-cell lymphoma 2, lung resistance protein, multidrug resistance protein, and protein kinase C alpha was significantly decreased in DOX-nano-treated HepG2/DOX cells and xenograft tumors. Significantly better antitumor and MDR-reversing effects were also observed in the HepG2+DOX group compared with the HepG2/DOX group. Conclusions This study revealed the potential efficacy of a DOX-nano drug delivery system for the treatment of HCC, using HepG2/DOX cells and nude mice bearing HepG2/DOX xenografts.
Collapse
Affiliation(s)
- Chunlei Wang
- Pharmaceutical Preparation Section, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China (mainland).,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China (mainland)
| | - Xiaoyan Wei
- Pharmaceutical Preparation Section, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China (mainland).,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China (mainland)
| | - Guoliang Shao
- Pharmaceutical Preparation Section, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China (mainland).,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
28
|
Hejmady S, Pradhan R, Alexander A, Agrawal M, Singhvi G, Gorain B, Tiwari S, Kesharwani P, Dubey SK. Recent advances in targeted nanomedicine as promising antitumor therapeutics. Drug Discov Today 2020; 25:2227-2244. [DOI: 10.1016/j.drudis.2020.09.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/29/2020] [Accepted: 09/26/2020] [Indexed: 12/18/2022]
|
29
|
Liu T, Lin J, Chen C, Nie X, Dou F, Chen J, Wang Z, Gong Z. MicroRNA-146b-5p overexpression attenuates premature ovarian failure in mice by inhibiting the Dab2ip/Ask1/p38-Mapk pathway and γH2A.X phosphorylation. Cell Prolif 2020; 54:e12954. [PMID: 33166004 PMCID: PMC7791167 DOI: 10.1111/cpr.12954] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/10/2020] [Accepted: 10/21/2020] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE To examine the role of high-fat and high-sugar (HFHS) diet-induced oxidative stress, which is a risk factor for various diseases, in premature ovarian failure (POF). MATERIALS AND METHODS Ovarian granulosa cells (OGCs) were isolated from mice and cultured in medium supplemented with HFHS and poly (lactic-co-glycolic acid) (PLGA)-cross-linked miR-146b-5p nanoparticles (miR-146@PLGA). RNA and protein expression levels were examined using quantitative real-time polymerase chain reaction and Western blotting, respectively. HFHS diet-induced POF model mice were administered miR-146@PLGA. RESULTS The ovarian tissue of mice fed a HFHS diet exhibited the typical pathological characteristics of POF. HFHS supplementation induced oxidative stress injury in the mouse OGCs, activation of the Dab2ip/Ask1/p38-Mapk signalling pathway and phosphorylation of γH2A.X in vitro and in vivo. The results of the luciferase reporter assay revealed that miR-146 specifically downregulated p38-Mapk14 expression. Meanwhile, co-immunoprecipitation and Western blot analyses revealed that HFHS supplementation upregulated nuclear p38-Mapk14 expression and consequently enhanced γH2A.X (Ser139) phosphorylation. The HFHS diet-induced POF mouse model treated with miR-146@PLGA exhibited downregulated p38-Mapk14 expression in the OGCs, mitigated OGC ageing and alleviated the symptoms of POF. CONCLUSIONS This study demonstrated that HFHS supplementation activates the Dab2ip/Ask1/p38-Mapk signalling pathway and promotes γH2A.X phosphorylation by inhibiting the expression of endogenous miR-146b-5p, which results in OGC ageing and POF development.
Collapse
Affiliation(s)
- Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Jiajia Lin
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoli Nie
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fangfang Dou
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiulin Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenxin Wang
- Department of Laboratory Medicine of Zhongshan Hospital and Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Zhangbin Gong
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
30
|
Parvez S, Yadagiri G, Singh A, Karole A, Singh OP, Sundar S, Mudavath SL. Improvising anti-leishmanial activity of amphotericin B and paromomycin using co-delivery in d-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) tailored nano-lipid carrier system. Chem Phys Lipids 2020; 231:104946. [PMID: 32621810 DOI: 10.1016/j.chemphyslip.2020.104946] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 01/13/2023]
Abstract
In the current study, we have focused on the design, development and in-vitro evaluation of d-α-tocopheryl polyethylene glycol 1000 succinate modified amphotericin B (AmB) and paromomycin (PM) loaded solid lipid nanoparticles (TPGS-SLNPs) by emulsion-solvent evaporation method. The optimized TPGS-SLNPs had a mean particle size of 199.4 ± 18.9 nm with a polydispersity index of 0.22 ± 0.14 and entrapment efficiency for AmB and PM was found to be 94 ± 1.5 % and 89 ± 0.50 % respectively. The prepared lipid nanoparticles were characterized by Powdered X-ray diffraction study, Fourier transform infrared spectroscopy, Nuclear magnetic resonance spectroscopy to confirm the absence of any interaction between lipids and drugs. The developed formulation showed a sustained drug release over a period of 48 h and were stable at different temperatures. Finally, TPGS-SLNPs (1 μg/mL) was found to significantly (P < 0.001) mitigate the intra-cellular amastigote growth compared to free AmB. The results obtained suggest TPGS-SLNPs could be an efficient carrier for delivering poorly water-soluble drugs and efficiently enhance its therapeutic potential.
Collapse
Affiliation(s)
- Shabi Parvez
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science & Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Ganesh Yadagiri
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science & Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Aakriti Singh
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science & Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Archana Karole
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science & Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Om Prakash Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Shyam Sundar
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Shyam Lal Mudavath
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science & Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India.
| |
Collapse
|
31
|
Santana R, Zuluaga R, Gañán P, Arrasate S, Onieva E, González-Díaz H. Predicting coated-nanoparticle drug release systems with perturbation-theory machine learning (PTML) models. NANOSCALE 2020; 12:13471-13483. [PMID: 32613998 DOI: 10.1039/d0nr01849j] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nanoparticles (NPs) decorated with coating agents (polymers, gels, proteins, etc.) form Nanoparticle Drug Delivery Systems (DDNS), which are of high interest in nanotechnology and biomaterials science. There have been increasing reports of experimental data sets of biological activity, toxicity, and delivery properties of DDNS. However, these data sets are still dispersed and not as large as the datasets of DDNS components (NP and drugs). This has prompted researchers to train Machine Learning (ML) algorithms that are able to design new DDNS based on the properties of their components. However, most ML models reported up to date predictions of the specific activities of NP or drugs over a determined target or cell line. In this paper, we combine Perturbation Theory and Machine Learning (PTML algorithm) to train a model that is able to predict the best components (NP, coating agent, and drug) for DDNS design. In so doing, we downloaded a dataset of >30 000 preclinical assays of drugs from ChEMBL. We also downloaded an NP data set formed by preclinical assays of coated Metal Oxide Nanoparticles (MONPs) from public sources. Both the drugs and NP datasets of preclinical assays cover multiple conditions of assays that can be listed as two arrays, namely, cjdrug and cjNP. The cjdrug array includes >504 biological activity parameters (c0drug), >340 target proteins (c1drug), >650 types of cells (c2drug), >120 assay organisms (c3drug), and >60 assay strains (c4drug). On the other hand, the cjNP array includes 3 biological activity parameters (c0NP), 40 types of proteins (c1NP), 10 shapes of nanoparticles (c2NP), 6 assay media (c3NP), and 12 coating agents (c4NP). After downloading, we pre-processed both the data sets by separate calculation PT operators that are able to account for changes (perturbations) in the drug, coating agents, and NP chemical structure and/or physicochemical properties as well as for the assay conditions. Next, we carry out an information fusion process to form a final dataset of above 500 000 DDNS (drug + MONP pairs). We also trained other linear and non-linear PTML models using R studio scripts for comparative purposes. To the best of our knowledge, this is the first multi-label PTML model that is useful for the selection of drugs, coating agents, and metal or metal-oxide nanoparticles to be assembled in order to design new DDNS with optimal activity/toxicity profiles.
Collapse
Affiliation(s)
- Ricardo Santana
- University of Deusto, Avda. Universidades, 24, 48007 Bilbao, Spain. and Grupo de Investigación Sobre Nuevos Materiales, Facultad de Ingeniería Química, Universidad Pontificia Bolivariana, Circular 1° N° 70-01, Medellín, Colombia
| | - Robin Zuluaga
- Facultad de Ingeniería Agroindustrial, Universidad Pontificia Bolivariana, Circular 1° N° 70-01, Medellín, Colombia
| | - Piedad Gañán
- Grupo de Investigación Sobre Nuevos Materiales, Facultad de Ingeniería Química, Universidad Pontificia Bolivariana, Circular 1° N° 70-01, Medellín, Colombia
| | - Sonia Arrasate
- Department of Organic Chemistry II, University of Basque Country UPV/EHU, 48940, Leioa, Spain.
| | - Enrique Onieva
- University of Deusto, Avda. Universidades, 24, 48007 Bilbao, Spain.
| | - Humbert González-Díaz
- Department of Organic Chemistry II, University of Basque Country UPV/EHU, 48940, Leioa, Spain. and IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain and Biofisika Institue CSIC-UPVEHU, University of Basque Country UPV/EHU, 48940, Leioa, Spain
| |
Collapse
|
32
|
Alurkar S, Goswami C, Bokil K, Raut N, Babu G. A Multicenter Retrospective Study to Evaluate Safety and Efficacy of Tocopheryl Polyethylene Glycol Succinate Docetaxel in Various Cancers. Indian J Med Paediatr Oncol 2020. [DOI: 10.4103/ijmpo.ijmpo_214_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
AbstractIn order to overcome the polysorbate induced hypersensitivity reactions with chemotherapy drugs, novel drug-delivery mechanisms have been developed in the last decade. D-alpha-tocopheryl polyethylene glycol succinate (TPGS) is formed by esterification of alpha-tocopheryl succinate and polyethylene glycol 1000.This was a real-world retrospective analysis designed to evaluate safety and efficacy of TPGS-docetaxel in various cancers. Patients hospitalized between June 2018 and May 2019 were included in the data set. While the efficacy was assessed by the Response Evaluation Criteria in Solid Tumors criteria, safety was assessed by the National Cancer Institute Common Terminology Criteria-adverse event (AE) criteria.A total of 61 patients who received at least one dose of TPGS-docetaxel were incorporated into the analysis set. The dose of TPGS docetaxel ranged from 20 mg/m2 to 120 mg/m2, commonly prescribed dose being 75 mg/m2. While 25 (40.98%) patients had a partial response, 17 (27.86%) patients had stable disease. Five (8.19%) patients progressed and 4 (6.55%) patients died during the chemotherapy, which was adjudicated to be unrelated to the drug as opined by the treating clinician. AE were reported in 42 patients in the safety data set. There were no AEs pertaining to hypersensitivity reported during the study. One AE of Grade 3 hand foot syndrome was encountered.The preliminary evidence suggests that the novel TPGS-based docetaxel formulation is efficacious in various cancers, and importantly, it has an enhanced safety profile, as it is devoid of polysorbate 80 induced hypersensitivity reactions.
Collapse
Affiliation(s)
| | | | | | | | - Govind Babu
- HCG Bangalore Institute of Oncology, Bengaluru, Karnataka, India
| |
Collapse
|
33
|
Effects of D-α-tocopherol polyethylene glycol succinate-emulsified poly(lactic-co-glycolic acid) nanoparticles on the absorption, pharmacokinetics, and pharmacodynamics of salinomycin sodium. Anticancer Drugs 2020; 30:72-80. [PMID: 30239423 DOI: 10.1097/cad.0000000000000695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although salinomycin sodium (SS) has shown in-vitro potential to inhibit cancer stem cell growth and development, its low water solubility makes it a poor candidate as an oral chemotherapeutic agent. To improve the bioavailability of SS, SS was encapsulated here using D-α-tocopherol polyethylene glycol succinate (TPGS)-emulsified poly(lactic-co-glycolic acid) (PLGA) nanoparticles and compared with its parent SS in terms of absorption, pharmacokinetics, and efficacy in suppressing nasopharyngeal carcinomas stem cells. The pharmacokinetics of SS and salinomycin sodium-loaded D-α-tocopherol polyethylene glycol succinate-emulsified poly(lactic-co-glycolic acid) nanoparticles (SLN) prepared by nanoprecipitation were analyzed in-vivo by timed-interval blood sampling and oral administration of SS and SLN to rats. Sensitive liquid chromatography-mass spectrometry (LC-MS) was developed to quantify plasma drug concentrations. SS and SLN transport in Caco-2 cells was also investigated. The therapeutic efficacy of SS and SLN against cancer stem cells was determined by orally administering the drugs to mice bearing CNE1 and CNE2 nasopharyngeal carcinoma xenografts and then evaluating CD133 cell proportions and tumorsphere formation. The in-vivo trial with rats showed that the Cmax, AUC(0-t), and Tmax for orally administered SLN were all significantly higher than those for SS (P<0.05). These findings were corroborated by a Caco-2 cell Transwell assay showing that relative SLN absorption was greater than that of SS on the basis of their apparent permeability coefficients (Papp). Significantly, therapeutic SLN efficacy against nasopharyngeal carcinoma stem cells was superior to that of SS. TPGS-emulsified PLGA nanoparticles effectively increase SS solubility and bioavailability. SLN is, therefore, promising as an oral chemotherapeutic agent against cancer stem cells.
Collapse
|
34
|
Jiang Y, Zhou Y, Zhang CY, Fang T. Co-Delivery of Paclitaxel and Doxorubicin by pH-Responsive Prodrug Micelles for Cancer Therapy. Int J Nanomedicine 2020; 15:3319-3331. [PMID: 32494132 PMCID: PMC7227817 DOI: 10.2147/ijn.s249144] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/18/2020] [Indexed: 12/18/2022] Open
Abstract
Background It is of great significance to develop intelligent co-delivery systems for cancer chemotherapy with improved therapeutic efficacy and few side-effects. Materials and Methods Here, we reported a co-delivery system based on pH-sensitive polyprodrug micelles for simultaneous delivery of doxorubicin (DOX) and paclitaxel (PTX) as a combination chemotherapy with pH-triggered drug release profiles. The physicochemical properties, drug release profiles and mechanism, and cytotoxicity of PTX/DOX-PMs have been thoroughly investigated. Results and Discussion The pH-sensitive polyprodrug was used as nanocarrier, and PTX was encapsulated into the micelles with high drug-loading content (25.6%). The critical micelle concentration (CMC) was about 3.16 mg/L, indicating the system could form the micelles at low concentration. The particle size of PTX/DOX-PMs was 110.5 nm, and increased to approximately 140 nm after incubation for 5 days which showed that the PTX/DOX-PMs had high serum stability. With decrease in pH value, the particle size first increased, and thenwas no longer detectable. Similar change trend was observed for CMC values. The zetapotential increased sharply with decrease in pH. These results demonstrated the pHsensitivity of PTX/DOX-PMs. In vitro drug release experiments and study on release mechanism showed that the drug release rate and accumulative release for PTX and DOX were dependent on the pH, showing the pH-triggered drug release profiles. Cytotoxicity assay displayed that the block copolymer showed negligible cytotoxicity, while the PTX/DOX-PMs possessed high cytotoxic effect against several tumor cell lines compared with free drugs and control. Conclusion All the results demonstrated that the co-delivery system based on pH-sensitive polyprodrug could be a potent nanomedicine for combination cancer chemotherapy. In addition, construction based on polyprodrug and chemical drug could be a useful method to prepare multifunctional nanomedicine.
Collapse
Affiliation(s)
- Yanhua Jiang
- Department of Anesthesiology, First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Yongjian Zhou
- Department of Anesthesiology, First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Can Yang Zhang
- Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Te Fang
- Department of Anesthesiology, First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| |
Collapse
|
35
|
Ren G, Chen P, Tang J, Guo W, Wang R, Li N, Li Y, Zhang G, Wang R, Zhang S. In vivo and in vitro evaluation of dihydroartemisinin prodrug nanocomplexes as a nano-drug delivery system: characterization, pharmacokinetics and pharmacodynamics. RSC Adv 2020; 10:17270-17279. [PMID: 35521441 PMCID: PMC9053626 DOI: 10.1039/d0ra02150d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 03/27/2020] [Indexed: 12/30/2022] Open
Abstract
To develop new, more effective and lower toxicity antitumor dihydroartemisinin (DHA) nanocomplexes, a DHA prodrug synthesized in this study was used to prepare DHA prodrug self-assembled nanocomplexes (DHANPs) by molecular self-assembly technology. The optimization, pharmacokinetics and in vitro and in vivo antitumor efficiency of DHANPs were assessed. The results showed that the entrapment efficiency, drug loading, particle size and zeta potential of the optimized formulation were 92.37 ± 3.68%, 76.98 ± 3.07%, 145.9 ± 2.11 nm and -16.0 ± 0.52 mV, respectively. DHANPs had a uniform size distribution and good stability during storage. The release of DHA prodrugs from DHANPs was slow in a PBS solution (pH 7.4). The pharmacokinetic study indicated that DHANPs could significantly improve the blood concentration of DHA. DHANPs exhibited lower cytotoxicity to 4T1 cells. More importantly, DHANPs could increase the quality life of mice in comparison with that of the DHA solution in 4T1 tumor-bearing mice. In short, the optimized DHA prodrug nanocomplexes show good long-term stability during the experimental time, extend the life-cycle of DHA in rats and can act as a prospective nano-drug delivery system for future artemisinin-based anti-tumor drugs.
Collapse
Affiliation(s)
- Guolian Ren
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
| | - Pei Chen
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
- School of Pharmacy, The Second Military Medical University Shanghai China
| | - Jiaqi Tang
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
| | - Wenju Guo
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
| | - Rongrong Wang
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
| | - Ning Li
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
| | - Yujie Li
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
| | - Guoshun Zhang
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
| | - Ruili Wang
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
| | - Shuqiu Zhang
- School of Pharmacy, Shanxi Medical University Taiyuan 030001 Shanxi China
| |
Collapse
|
36
|
Yang X, Shi X, Zhang Y, Xu J, Ji J, Ye L, Yi F, Zhai G. Photo-triggered self-destructive ROS-responsive nanoparticles of high paclitaxel/chlorin e6 co-loading capacity for synergetic chemo-photodynamic therapy. J Control Release 2020; 323:333-349. [PMID: 32325174 DOI: 10.1016/j.jconrel.2020.04.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 02/07/2023]
Abstract
To improve the anti-cancer therapeutic effect of nanosystems for chemo-photodynamic therapy, there remain several hurdles to be addressed, e.g., limited co-loading efficiency, insufficient stimulus-responsiveness and lack of synergetic effect. This work reported novel reactive‑oxygen-species (ROS)-responsive chlorin e6 (Ce6) and paclitaxel (PTX) co-encapsulated chondroitin sulfate-g-poly (propylene sulfide) nanoparticles (CP/ChS-g-PPS NPs), wherein the drug loading efficiencies of Ce6 and PTX were as high as 14.93% and 24.31%, respectively. To enlarge the ROS signal at tumor sites thus enhancing the ROS-responsiveness of ChS-g-PPS NPs, near-infrared (NIR) light was utilized to induce Ce6 to produce more ROS to destruct the NPs. Our data showed that the photo-triggered self-destructive property of NPs helped drugs to spread deeper in tumors upon laser irradiation, making the NPs promising to thoroughly remove tumor cells. CP/ChS-g-PPS NPs exhibited a synergetic chemo-photodynamic therapy effect in vitro, which was suggested by the combination indexes of PTX and Ce6 lower than 1 when 20-80% inhibition rates of MCF-7 cells were achieved. As for the in vivo antitumor activity, the tumor inhibition rates of CP/ChS-g-PPS NPs (with laser irradiation) were as high as 92.76% and 88.57% in 4T1 bearing BALB/c mice and MCF-7 bearing BALB/c nude mice, respectively, which were significantly higher than those of other treatment groups. This work provided a simple yet effective strategy to develop photo-triggered ROS-responsive NPs for synergetic chemo-photodynamic therapy with quick ROS-responsive self-destruction, spatiotemporally controllability, reduced off-target toxicity, and desirable therapeutic effect.
Collapse
Affiliation(s)
- Xiaoye Yang
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yanan Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jiangkang Xu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
37
|
Akhter MH, Kumar S, Nomani S. Sonication tailored enhance cytotoxicity of naringenin nanoparticle in pancreatic cancer: design, optimization, and in vitro studies. Drug Dev Ind Pharm 2020; 46:659-672. [PMID: 32208984 DOI: 10.1080/03639045.2020.1747485] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective: In vitro, optimization, characterization, and cytotoxic studies of NAR nanoparticles (NPs) to against pancreatic cancer.Method: The sonication tailored Naringenin (NARG)-loaded poly (lactide-co-glycolic acid) (PLGA) NPs was fabricated for potential cytotoxic effect against pancreatic cancer. NARG NPs were prepared by emulsion-diffusion evaporation technique applying BoxBehnken experimental design based on three-level and three-factors. The effect of independent variables surfactant concentration (X1), polymer concentration (X2), and sonication time (X3) were studied on responses particle size (Y1), and drug release % (Y2). NPs characterized for particles size and size distribution, polydispersity index (PDI), zeta potential, transmission electron microscope (TEM), scanning electron microscope (SEM), Fourier transform infrared spectroscopy (FT-IR), Differential scanning calorimeter (DSC), and X-ray diffraction (XRD) studies. Further, the studies was fitted to various drug release kinetic model and cytotoxicity evaluated in vitro.Results: The nanosized particles were spherical, uniform with an average size of 150.45 ± 12.45 nm, PDI value 0.132 ± 0.026, zeta potential -20.5 ± 2.5 mV, and cumulative percentage release 85.67 ± 6.23%. In vitro release of NARG from nanoparticle evaluated initially burst followed by sustained release behavior. The Higuchi was best fitted model to drug release from NARG NPs. The cytotoxicity study of NARG NPs apparently showed higher cytotoxic effect over free NARG (p < 0.05). The stability study of optimized formulation revealed no significant physico-chemical changes during 3 months.Conclusions: Thus, NARG-loaded NPs gave ameliorated anticancer effect over plain NARG.
Collapse
Affiliation(s)
| | - Sandeep Kumar
- Alwar Pharmacy College Rajasthan University of Health Sciences (RUHS), M.I.A. Alwar-Rajasthan, Alwar, India.,Karnataka Antibiotics and Pharmaceutical Limited, Bengaluru, India
| | | |
Collapse
|
38
|
Wang H, Williams GR, Xie X, Wu M, Wu J, Zhu LM. Stealth Polydopamine-Based Nanoparticles with Red Blood Cell Membrane for the Chemo-Photothermal Therapy of Cancer. ACS APPLIED BIO MATERIALS 2020; 3:2350-2359. [DOI: 10.1021/acsabm.0c00094] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Haijun Wang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Gareth R. Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K
| | - Xiaotian Xie
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Meng Wu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Junzi Wu
- College of Basic Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Li-Min Zhu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| |
Collapse
|
39
|
Song Y, Zhou B, Du X, Wang Y, Zhang J, Ai Y, Xia Z, Zhao G. Folic acid (FA)-conjugated mesoporous silica nanoparticles combined with MRP-1 siRNA improves the suppressive effects of myricetin on non-small cell lung cancer (NSCLC). Biomed Pharmacother 2020; 125:109561. [PMID: 32106385 DOI: 10.1016/j.biopha.2019.109561] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/08/2019] [Accepted: 10/17/2019] [Indexed: 01/05/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a common diagnosed cancer disease worldwide and its management remains a challenge. Synergistic cancer therapeutic strategy is interesting for multiple advantages, such as excellent targeting accuracy, low side effects, and promoted therapeutic efficiency. In the present study, myricetin (Myr)-loaded mesoporous silica nanoparticles (MSN) combined with multidrug resistance protein (MRP-1) siRNA was prepared. The surface of the synthesized nanoparticles was modified with folic acid (FA) to promote the therapeutic efficiency of Myr for the treatment of NSCLC. The collected particles were nano-sized and showed a sustained release of Myr in the physiological conditions. FA-conjugated nanoformulations displayed a significant uptake in lung cancer cells compared with that of the non-targeted nanoparticles. The in vitro drug release results suggested a sustained release in FA-conjugated MSN with Myr and MRP-1 nanoparticles compared to the free Myr and MSN combined with MRP-1/Myr. Treatments with FA-conjugated MSN combined with Myr and MRP-1 markedly reduced the cell viability of lung cancer cell lines, including A549 and NCI-H1299, which was accompanied with the decreased number of colony formation. In addition, FA-conjugated MSN loaded with Myr and MRP-1 significantly induced apoptosis in lung cancer cells, along with up-regulated expression levels of cleaved Caspase-3 and PARP. In vivo fluorescence results demonstrated that FA-conjugated MSN with Myr and MRP-1 nanoparticles could specifically accumulate at tumor sites. Compared with free Myr and MSN combined with MRP-1/Myr nanoparticles, FA-conjugated MSN loaded with Myr and MRP-1 nanoparticles could more effectively suppress tumor growth with little side effects. Overall, FA-conjugated nanoparticulate system could provide a novel and effective platform for the treatment of NSCLC.
Collapse
Affiliation(s)
- Yinxue Song
- Department of Emergency, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Bin Zhou
- Department of Thoracic Surgery, Shanxian Central Hospital, Shanxian, Shandong, 274300, China
| | - Xiangyang Du
- Department of Respiration, Shandong Provincial Third Hospital, Jinan, Shandong, 250031, China.
| | - Yong Wang
- R&D Center of Zhengzhou Bio-Medicinal Institute, Zhengzhou, 450052, China.
| | - Jie Zhang
- R&D Center of Zhengzhou Bio-Medicinal Institute, Zhengzhou, 450052, China
| | - Yanqiu Ai
- R&D Center of Zhengzhou Bio-Medicinal Institute, Zhengzhou, 450052, China
| | - Zongjiang Xia
- Department of New Drugs Development, Shanghai Genecure Pharmaceutical Institute, Shanghai, 200040, China
| | - Gaofeng Zhao
- Department of New Drugs Development, Shanghai Genecure Pharmaceutical Institute, Shanghai, 200040, China
| |
Collapse
|
40
|
Dissolving Microneedles Loading TPGS Biphasic Functionalized PLGA Nanoparticles for Efficient Chemo‐Photothermal Combined Therapy of Melanoma. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
41
|
Chen Y, Mo L, Wang X, Chen B, Hua Y, Gong L, Yang F, Li Y, Chen F, Zhu G, Ni W, Zhang C, Cheng Y, Luo Y, Shi J, Qiu M, Wu S, Tan Z, Wang K. TPGS-1000 exhibits potent anticancer activity for hepatocellular carcinoma in vitro and in vivo. Aging (Albany NY) 2020; 12:1624-1642. [PMID: 31986488 PMCID: PMC7053644 DOI: 10.18632/aging.102704] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 01/02/2020] [Indexed: 12/15/2022]
Abstract
D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS1000) is the most active water-soluble derivative of vitamin E and has been widely used as a carrier of solvents, plasticizers, emulsifiers, absorbent agents and refractory drug delivery systems. However, its anti-hepatocellular carcinoma (HCC) properties have not been explored. HCC cells were treated with different concentrations of TPGS1000. Cell survival was tested by CCK8 assay, and cell migration was tested by wound healing and Transwell assay. EdU staining verified cell proliferation, and signalling pathway was assayed by Western blot analysis. The BALB/c-nu mouse xenograft model was established to test HCC cell growth in vivo. In vitro TPGS1000 significantly inhibited the viability and mobility of HCC cells (HepG2, Hep3B and Huh7) in a dose-dependent manner. Cell cycle analysis indicated that TPGS1000 treatment arrested the HCC cell cycle in the G0/G1 phase, and induction of cell apoptosis was confirmed by TUNEL and Annexin V-7-AAD staining. Further pharmacological analysis indicated that collapse of the transmembrane potential of mitochondria, increased ROS generation, PARP-induced cell apoptosis and FoxM1-p21-mediated cell cycle arresting, were involved in the anti-HCC activity of TPGS1000. Moreover, treatment in vivo with TPGS1000 effectively impaired the growth of HCC xenografts in nude mice.
Collapse
Affiliation(s)
- Yidan Chen
- Cancer Research Institute, Hangzhou Cancer Hospital, Zhejiang, China
| | - Liqin Mo
- Cancer Research Institute, Hangzhou Cancer Hospital, Zhejiang, China
| | - Xuan Wang
- Life Sciences Research Institute, College of Life and Environmental Sciences, Hangzhou Normal University, Zhejiang, China
| | - Bi Chen
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Yunfen Hua
- College of Pharmaceutical Science, Zhejiang University of Technology, Zhejiang, China
| | - Linyan Gong
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Fei Yang
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Yongqiang Li
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Fangfang Chen
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Guiting Zhu
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Wei Ni
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Cheng Zhang
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Yuming Cheng
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Yan Luo
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Junping Shi
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Mengsheng Qiu
- Life Sciences Research Institute, College of Life and Environmental Sciences, Hangzhou Normal University, Zhejiang, China
| | - Shixiu Wu
- Cancer Research Institute, Hangzhou Cancer Hospital, Zhejiang, China
| | - Zhou Tan
- Life Sciences Research Institute, College of Life and Environmental Sciences, Hangzhou Normal University, Zhejiang, China
| | - Kaifeng Wang
- Cancer Research Institute, Hangzhou Cancer Hospital, Zhejiang, China.,State Key Laboratory for Oncogenes and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai Cancer Institute, Shanghai, China
| |
Collapse
|
42
|
Caban-Toktas S, Sahin A, Lule S, Esendagli G, Vural I, Karlı Oguz K, Soylemezoglu F, Mut M, Dalkara T, Khan M, Capan Y. Combination of Paclitaxel and R-flurbiprofen loaded PLGA nanoparticles suppresses glioblastoma growth on systemic administration. Int J Pharm 2020; 578:119076. [PMID: 31988035 DOI: 10.1016/j.ijpharm.2020.119076] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/22/2022]
Abstract
Malignant gliomas are highly lethal. Delivering chemotherapeutic drugs to the brain in sufficient concentration is the major limitation in their treatment due to the blood-brain barrier (BBB). Drug delivery systems may overcome this limitation and can improve the transportation through the BBB. Paclitaxel is an antimicrotubule agent with effective anticancer activity but limited BBB permeability. R-Flurbiprofen is a nonsteroidal antienflammatory drug and has potential anticancer activity. Accordingly, we designed an approach combining R-flurbiprofen and paclitaxel and positively-charged chitosan-modified poly-lactide-co-glycolic acid (PLGA) nanoparticles (NPs) and to transport them to glioma tissue. NPs were characterized and, cytotoxicity and cellular uptake studies were carried out in vitro. The in vivo efficacy of the combination and formulations were evaluated using a rat RG2 glioma tumor model. Polyethylene glycol (PEG) modified and chitosan-coated PLGA NPs demonstrated efficient cytotoxic activity and were internalized by the tumor cells in RG2 cell culture. In vivo studies showed that the chitosan-coated and PEGylated NPs loaded with paclitaxel and R-flurbiprofen exhibited significantly higher therapeutic activity against glioma. In conclusion, PLGA NPs can efficiently carry their payloads to glioma tissue and the combined use of anticancer and anti-inflammatory drugs may exert additional anti-tumor activity.
Collapse
Affiliation(s)
- Secil Caban-Toktas
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Adem Sahin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sevda Lule
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Gunes Esendagli
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey.
| | - Imran Vural
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| | - Kader Karlı Oguz
- Department of Radiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey; National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
| | - Figen Soylemezoglu
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| | - Melike Mut
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| | - Turgay Dalkara
- Department of Neurology, Faculty of Medicine and Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
| | - Mansoor Khan
- Texas A&M Health Science Center, Irma Lerma Rangel College of Pharmacy, Texas, USA.
| | - Yilmaz Capan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
43
|
Zhang Q, Tian X, Cao X. Transferrin-functionalised microemulsion co-delivery of β-elemene and celastrol for enhanced anti-lung cancer treatment and reduced systemic toxicity. Drug Deliv Transl Res 2020; 9:667-678. [PMID: 30798476 DOI: 10.1007/s13346-019-00623-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this study, we developed an intravenously injectable, transferrin-functionalised microemulsion that simultaneously carries β-elemene and celastrol (called Tf-EC-MEs) for enhanced anti-lung cancer treatment and reduced systemic toxicity. These dual-drug-loaded Tf-EC-MEs not only displayed synergistic antiproliferative effects on cultured cells in vitro, but also showed enhanced efficacy in vivo via active tumour targeting. In preparatory experiments, we found that β-elemene was capable of being used as oil phase, which enhanced drug-loading efficiency and allowed the mass ratio of β-elemene and celastrol to be optimised. In cellular studies, Tf-EC-MEs exhibited significantly improved A549 cellular uptake compared with β-elemene+celastrol (conventional combination treatment) and EC-MEs (non-active targeted treatment), demonstrating remarkable synergistic antiproliferative effects and higher rates of cell apoptosis. In A549-bearing xenograft mouse tumour models, Tf-EC-MEs exhibited enhanced antitumour activity compared to all other treatments. More importantly, Tf-EC-MEs did not cause the obvious systemic toxicity commonly found with mono-celastrol treatment. Collectively, these findings suggest that Tf-EC-MEs are a promising strategy for the combination drug treatment of lung cancer.
Collapse
Affiliation(s)
- Qian Zhang
- The Affiliated Sir Run Run Hospital, Nanjing Medical University, Nanjing, 210009, People's Republic of China
- The Affiliated Nanjing Hospital, Nanjing Medical University, Changle road 68, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Xin Tian
- Fourth Military Medical University, Xi'an, 710003, People's Republic of China
- Traditional Chinese Medicine Hospital of Shaanxi Province, Xi'an, 710003, People's Republic of China
| | - Xiufeng Cao
- The Affiliated Nanjing Hospital, Nanjing Medical University, Changle road 68, Nanjing, 210009, Jiangsu, People's Republic of China.
| |
Collapse
|
44
|
Mahmood MA, Madni A, Rehman M, Rahim MA, Jabar A. Ionically Cross-Linked Chitosan Nanoparticles for Sustained Delivery of Docetaxel: Fabrication, Post-Formulation and Acute Oral Toxicity Evaluation. Int J Nanomedicine 2019; 14:10035-10046. [PMID: 31908458 PMCID: PMC6929931 DOI: 10.2147/ijn.s232350] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/02/2019] [Indexed: 01/19/2023] Open
Abstract
Introduction Polymeric nanoparticles are potential carriers for the efficient delivery of hydrophilic and hydrophobic drugs due to their multifaceted applications. Docetaxel is relatively less hydrophobic and twice as potent as paclitaxel. Like other taxane chemotherapeutic agents, docetaxel is not well tolerated and shows toxicity in the patients. Nanoencapsulation of potent chemotherapeutic agents has been shown to improve tolerability and therapeutic outcome. Therefore, the present study was designed to fabricate chitosan and sodium tripolyphosphate (STPP) based on ionically cross-linked nanoparticles for sustained release of docetaxel. Methods Nanoparticles were prepared by the ionic-gelation method by dropwise addition of the STPP solution into the chitosan solution in different ratios. CNPs were characterized for post-formulation parameters like size, zeta potential, scanning electron microscope (SEM), FTIR, DSC/TGA, pXRD, and in-vitro drug release, as well as for acute oral toxicity studies in Wistar rats. Results and discussion The optimized docetaxel loaded polymeric nanoparticles were in the size range (172.6nm–479.65 nm), and zeta potential (30.45–35.95 mV) required to achieve enhanced permeation and retention effect. In addition, scanning electron microscopy revealed rough and porous surface, whereas, FTIR revealed the compatible polymeric nanoparticles. Likewise, the thermal stability was ensured through DSC and TG analysis, and powder X-ray diffraction analysis exhibited solid-state stability of the docetaxel loaded nanoparticles. The in-vitro drug release evaluation in phosphate buffer saline (pH 7.4) showed sustained release pattern, i.e. 51.57–69.93% within 24 hrs. The data were fitted to different release kinetic models which showed Fickian diffusion as a predominant release mechanism (R2= 0.9734–0.9786, n= 0.264–0.340). Acceptable tolerability was exhibited by acute oral toxicity in rabbits and no abnormality was noted in growth, behavior, blood biochemistry or histology and function of vital organs. Conclusion Ionically cross-linked chitosan nanoparticles are non-toxic and biocompatible drug delivery systems for sustained release of chemotherapeutic agents, such as docetaxel.
Collapse
Affiliation(s)
- Muhammad Ahmad Mahmood
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.,Drug Testing Laboratory, Bahawalpur 63100, Pakistan
| | - Asadullah Madni
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Mubashar Rehman
- Faculty of Pharmacy, University of Central Punjab, Lahore 54000, Pakistan
| | - Muhammad Abdur Rahim
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Abdul Jabar
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| |
Collapse
|
45
|
Gao J, Liu J, Xie F, Lu Y, Yin C, Shen X. Co-Delivery of Docetaxel and Salinomycin to Target Both Breast Cancer Cells and Stem Cells by PLGA/TPGS Nanoparticles. Int J Nanomedicine 2019; 14:9199-9216. [PMID: 32063706 PMCID: PMC6884979 DOI: 10.2147/ijn.s230376] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/13/2019] [Indexed: 01/27/2023] Open
Abstract
PURPOSE Conventional chemotherapy is hampered by the presence of breast cancer stem cells (BCSCs). It is crucial to eradicating both the bulky breast cancer cells and BCSCs, using a combination of conventional chemotherapy and anti-CSCs drugs. However, the synergistic ratio of drug combinations cannot be easily maintained in vivo. In our previous studies, we demonstrated that the simultaneous delivery of two drugs via nanoliposomes could maintain the synergistic drug ratio for 12 h in vivo. However, nanoliposomes have the disadvantage of quick drug release, which makes it difficult to maintain the synergistic drug ratio for a long time. Herein, we developed a co-delivery system for docetaxel (DTX)-a first-line chemotherapy drug for breast cancer-and salinomycin (SAL)-an anti-BCSCs drug-in rigid nanoparticles constituted of polylactide-co-glycolide/D-alpha-tocopherol polyethylene glycol 1000 succinate (PLGA/TPGS). METHODS Nanoparticles loaded with SAL and DTX at the optimized ratio (NSD) were prepared by the nanoprecipitation method. The characterization, cellular uptake, and cytotoxicity of nanoparticles were investigated in vitro, and the pharmacokinetics, tissue distribution, antitumor and anti-CSCs activity of nanoparticles were evaluated in vivo. RESULTS We demonstrated that a SAL/DTX molar ratio of 1:1 was synergistic in MCF-7 cells and MCF-7-MS. Moreover, the enhanced internalization of nanoparticles was observed in MCF-7 cells and MCF-7-MS. Furthermore, the cytotoxicity of NSD against both MCF-7 cells and MCF-7-MS was stronger than the cytotoxicity of any single treatment in vitro. Significantly, NSD could prolong the circulation time and maintain the synergistic ratio of SAL to DTX in vivo for 24 h, thus exhibiting superior tumor targeting and anti-tumor activity compared to other treatments. CONCLUSION Co-encapsulation of SAL and DTX in PLGA/TPGS nanoparticles could maintain the synergistic ratio of drugs in vivo in a better manner; thus, providing a promising strategy for synergistic inhibition of breast cancer.
Collapse
Affiliation(s)
- Jie Gao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Scientific Research Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Junjie Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai, People’s Republic of China
| | - Fangyuan Xie
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, People’s Republic of China
| | - Ying Lu
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai, People’s Republic of China
| | - Chuan Yin
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Xian Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| |
Collapse
|
46
|
Iron Transport Tocopheryl Polyethylene Glycol Succinate in Animal Health and Diseases. Molecules 2019; 24:molecules24234289. [PMID: 31775281 PMCID: PMC6930530 DOI: 10.3390/molecules24234289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022] Open
Abstract
Gut health is the starting place for maintaining the overall health of an animal. Strategies to maintain gut health are, thus, an important part in achieving the goal of improving animal health. A new strategy to do this involves two molecules: the iron transport protein ovotransferrin (IT) and α-tocopheryl polyethylene glycol succinate (TPGS), which result in the novel formulation of ITPGS. These molecules help reduce gut pathogens, while enhancing the absorption and bioavailability of therapeutic drugs, phytomedicines, and nanomedicines. This, in turn, helps to maintain normal health in animals. Maintaining the gastrointestinal tract (GIT) in its normal condition is key for successful absorption and efficacy of any nutrient. A compromised GIT, due to an imbalance (dysbiosis) in the GIT microbiome, can lead to an impaired GI barrier system with impaired absorption and overall health of the animal. The molecules in ITPGS may address the issue of poor absorption by keeping the GI system healthy by maintaining the normal microbiome and improving the absorption of nutrients through multiple mechanisms involving antioxidative, anti-inflammatory, immunomodulatory, and antimicrobial activities. The ITPGS technology can allow the dose of active pharmaceutical or herbal medicine to be significantly reduced in order to attain equal or better efficacy. With complimentary actions between IT and TPGS, ITPGS presents a novel approach to increase the bioavailability of drugs, phytoconstituents, nutrients, and nanomedicines by enhanced transport to the tissues at the site of action, while reducing gut pathogen load. The ITPGS approach appears to be a novel strategy for maintaining the health of animals by manipulation of microbiota.
Collapse
|
47
|
Santana R, Zuluaga R, Gañán P, Arrasate S, Onieva E, González-Díaz H. Designing nanoparticle release systems for drug-vitamin cancer co-therapy with multiplicative perturbation-theory machine learning (PTML) models. NANOSCALE 2019; 11:21811-21823. [PMID: 31691701 DOI: 10.1039/c9nr05070a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Nano-systems for cancer co-therapy including vitamins or vitamin derivatives have showed adequate results to continue with further research studies to better understand them. However, the number of different combinations of drugs, vitamins, nanoparticle types, coating agents, synthesis conditions, and system types (nanocapsules, micelles, etc.) to be tested is very large generating a high cost in experimentations. In this context, there are reports of large datasets of preclinical assays of compounds (like in the ChEMBL database) and increasing but yet limited reports of experimental measurements of nano-systems per se. On the other hand, Machine Learning is gaining momentum in Nanotechnology and Pharmaceutical Sciences as a tool for rational design of new drugs and drug-release nano-systems. In this work, we propose to combine Perturbation Theory principles and Machine Learning to develop a PTML model for rational selection of the components of cancer co-therapy drug-vitamin release nano-systems (DVRNs). In doing so, we apply information fusion techniques with 2 data sets: (1) a large ChEMBL dataset of >36 000 preclinical assays of vitamin derivatives and a new dataset of >1000 outcomes of DVRNs, collected herein from the literature for the first time. The ChEMBL dataset used covers a considerable number of assay conditions (cjvit) each one with multiple levels. These conditions included >504 biological activity parameters (c0vit), >340 types of proteins (c1vit), >650 types of cells (c2vit), >120 assay organisms (c3vit), >60 assay strains (c4vit). Regarding the DVRNs, there are 25 different types of nano-systems (njn), with up to 16 conditions (cjn) including also different levels such as 8 biological activity parameters (c0n), 9 raw nanomaterials (c4n), 15 assay cells (c11n), etc. In the first stage, we used Moving Average operators to quantify the perturbations (deviations) in all input variables with respect to the conditions. After that, we used multiplicative PT operators to carry out data fusion, and dimension reduction, and Linear Discriminant Analysis (LDA) to seek the PTML model. The best PTML model found showed values of specificity, sensitivity, and accuracy in the range of 83-88% in training and external validation series for >130 000 cases (DVRNs vs. ChEMBL data pairs) formed after data fusion. To the best of our knowledge, this is the first general purpose model for the rational design of DVRNs for cancer co-therapy.
Collapse
|
48
|
Zhen L, Wei Q, Wang Q, Zhang H, Adu-Frimpong M, Kesse Firempong C, Xu X, Yu J. Preparation and in vitro/in vivo evaluation of 6-Gingerol TPGS/PEG-PCL polymeric micelles. Pharm Dev Technol 2019; 25:1-8. [DOI: 10.1080/10837450.2018.1558239] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Lijun Zhen
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Qiuyu Wei
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Huiyun Zhang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Michael Adu-Frimpong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Caleb Kesse Firempong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People’s Republic of China
- Department of Biochemistry and Biotechnology, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People’s Republic of China
| |
Collapse
|
49
|
Faustova M, Nikolskaya E, Sokol M, Zabolotsky A, Mollaev M, Zhunina O, Fomicheva M, Lobanov A, Severin E, Yabbarov N. High-effective reactive oxygen species inducer based on Mn-tetraphenylporphyrin loaded PLGA nanoparticles in binary catalyst therapy. Free Radic Biol Med 2019; 143:522-533. [PMID: 31520768 DOI: 10.1016/j.freeradbiomed.2019.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 01/09/2023]
Abstract
The mechanisms of binary catalyst therapy (BCT) and photodynamic therapy (PDT) are based on the formation of reactive oxygen species (ROS). This ROS formation results from specific chemical reactions. In BCT, light exposure does not necessarily initiate ROS formation and BCT application is not limited to regions of tissues that are accessible to illumination like photodynamic therapy (PDT). The principle of BCT is electron transition, resulting in the interaction of a transition metal complex (catalyst) and substrate molecule. MnIII- tetraphenylporphyrin chloride (MnClTPP) in combination with an ascorbic acid (AA) has been proposed as an appropriate candidate for cancer treatment regarding the active agents in BCT. The goal of this study was to determine whether MnClTPP in combination with AA would be a promising agent for BCT. The problem of used MnClTPP's, low solubility in water, was solved by MnClTPP loading into PLGA matrix. H2O2 produced during AA decomposition oxidized MnClTPP to high-reactive oxo-MnV species. MnClTPP in presence AA leads to the production of excessive ROS levels in vitro. ROS are mainly substrates of catalase and superoxide dismutase (H2O2 and O2●-). SOD1 and catalase were identified as the key players of the MnClTPP ROS-induced cell defense system. The cytotoxicity of MnClTPP-loaded nanoparticles (NPs) was greatly increased in the presence of specific catalase inhibitor (3-amino-1,2,4-triazole (3AT)) and superoxide dismutase 1 (SOD1) inhibitor (diethyldithiocarbamate (DDC)). Cell death resulted from the combined activation of caspase-dependent (caspase 3/9 system) and independent pathways, namely the AIF translocation to nuclei. Preliminary acute toxicity and in vivo anticancer studies have been revealed the safe and potent anticancer effect of PLGA-entrapped MnClTPP in combination with AA. The findings indicate that MnClTPP-loaded PLGA NPs are promising agents for BCT.
Collapse
Affiliation(s)
- Maria Faustova
- MIREA, Russian Technological University, Lomonosov Institute of Fine Chemical Technologies, 119454, Moscow, Russia
| | | | - Maria Sokol
- JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149, Moscow, Russia
| | | | - Murad Mollaev
- MIREA, Russian Technological University, Lomonosov Institute of Fine Chemical Technologies, 119454, Moscow, Russia
| | - Olga Zhunina
- Semenov Institute of Chemical Physics, 119991, Moscow, Russia
| | - Margarita Fomicheva
- JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149, Moscow, Russia
| | - Anton Lobanov
- Semenov Institute of Chemical Physics, 119991, Moscow, Russia
| | - Evgeniy Severin
- JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149, Moscow, Russia
| | - Nikita Yabbarov
- Semenov Institute of Chemical Physics, 119991, Moscow, Russia.
| |
Collapse
|
50
|
Ruan L, Wang M, Zhou M, Lu H, Zhang J, Gao J, Chen J, Hu Y. Doxorubicin–Metal Coordinated Micellar Nanoparticles for Intracellular Codelivery and Chemo/Chemodynamic Therapy in Vitro. ACS APPLIED BIO MATERIALS 2019; 2:4703-4707. [DOI: 10.1021/acsabm.9b00879] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Lifo Ruan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mengxue Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huiru Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jiayu Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|