1
|
Valle ABCDS, da Silva FFA, Carneiro MÂP, Espuche B, Tavares GD, Bernardes ES, Moya SE, Pittella F. In Vivo HOXB7 Gene Silencing and Cotreatment with Tamoxifen for Luminal A Breast Cancer Therapy. Pharmaceuticals (Basel) 2024; 17:1325. [PMID: 39458966 PMCID: PMC11509954 DOI: 10.3390/ph17101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Acquired resistance and adverse effects are some of the challenges faced by thousands of Luminal A breast cancer patients under tamoxifen (TMX) treatment. Some authors associate the overexpression of HOXB7 with TMX resistance in this molecular subtype, and the knockdown of this gene could be an effective strategy to regain TMX sensitivity. Therefore, we used calcium phosphate hybrid nanoparticles (HNP) for the delivery of short interfering RNA molecule (siRNA) complementary to the HOXB7 gene and evaluated the RNA interference (RNAi) effects associated with TMX treatment in breast cancer in vivo. METHODS HNP were prepared by the self-assembly of a methoxy-poly (ethylene glycol)-block-poly (L-glutamic acid) copolymer (PEG-pGlu) and the coprecipitation of CaPO4 to incorporate siRNA. The in vitro cell viability and migration were evaluated prior to in vivo experiments. Further, animals bearing early-stage and advanced Luminal A breast cancer were treated with HNP-siHOXB7, HNP-siHOXB7 + TMX, and TMX. Antitumoral activity and gene expression were evaluated following histopathological, hematological, and biochemical analysis. RESULTS The HNP were efficient in delivering the siRNA in vitro and in vivo, whilst HOXB7 silencing associated with TMX administration promoted controlled tumor growth, as well as a higher survival rate and reduction in immuno- and hepatotoxicity. CONCLUSIONS Therefore, our findings suggest that HOXB7 can be an interesting molecular target for Luminal A breast cancer, especially associated with hormone therapy, aiming for adverse effect mitigation and higher therapeutic efficacy.
Collapse
Affiliation(s)
- Ana Beatriz Caribé dos Santos Valle
- Laboratório de Desenvolvimento de Sistemas Nanoestruturados, Faculdade de Farmácia, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer, Juiz de Fora 36036-900, Brazil; (A.B.C.d.S.V.); (G.D.T.)
| | - Fábio Fernando Alves da Silva
- Instituto de Pesquisas Energéticas e Nucleares, Centro de Radiofarmácia (IPEN/CECRF), Comissão Nacional de Energia Nuclear, São Paulo 05508-000, Brazil; (F.F.A.d.S.); (M.Â.P.C.); (E.S.B.)
| | - Maria Ângela Pepe Carneiro
- Instituto de Pesquisas Energéticas e Nucleares, Centro de Radiofarmácia (IPEN/CECRF), Comissão Nacional de Energia Nuclear, São Paulo 05508-000, Brazil; (F.F.A.d.S.); (M.Â.P.C.); (E.S.B.)
| | - Bruno Espuche
- Soft Matter Laboratory, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo Miramón 194, 20014 Donostia-San Sebastián, Spain; (B.E.); (S.E.M.)
| | - Guilherme Diniz Tavares
- Laboratório de Desenvolvimento de Sistemas Nanoestruturados, Faculdade de Farmácia, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer, Juiz de Fora 36036-900, Brazil; (A.B.C.d.S.V.); (G.D.T.)
| | - Emerson Soares Bernardes
- Instituto de Pesquisas Energéticas e Nucleares, Centro de Radiofarmácia (IPEN/CECRF), Comissão Nacional de Energia Nuclear, São Paulo 05508-000, Brazil; (F.F.A.d.S.); (M.Â.P.C.); (E.S.B.)
| | - Sergio Enrique Moya
- Soft Matter Laboratory, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo Miramón 194, 20014 Donostia-San Sebastián, Spain; (B.E.); (S.E.M.)
| | - Frederico Pittella
- Laboratório de Desenvolvimento de Sistemas Nanoestruturados, Faculdade de Farmácia, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer, Juiz de Fora 36036-900, Brazil; (A.B.C.d.S.V.); (G.D.T.)
| |
Collapse
|
2
|
Shawky H, Fayed DB, Ibrahim NE. pH-tailored delivery of a multitarget anticancer benzimidazole derivative using a PEGylated β-cyclodextrin-curcumin functionalized nanocomplex. BIOMATERIALS ADVANCES 2024; 163:213964. [PMID: 39053387 DOI: 10.1016/j.bioadv.2024.213964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/29/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
In this study, we aimed to enhance the bioavailability of a benzimidazole derivative with potent anticancer potential through a nano-based approach. Benzimidazole-loaded polyethylene glycol-β-cyclodextrin-functionalized curcumin nanocomplex (BMPE-Cur) was prepared and characterized for its physicochemical properties and drug release profiles under different pH conditions. In addition, the biological activities of the nanocomplex including antioxidant potentials and pro-apoptogenic properties, against HepG2, PC3, and the chemo-resistant MCF-7-ADR cell lines relative to the normal Wi-38 cell line were in vitro assessed and compared with those of the free benzimidazole compound. In addition to FTIR, XRD, and NMR spectral studies, a polymeric nanocomplex with an average particle size of 467.7 nm and high stability was successfully developed, as indicated by the negative zeta potential (-28.24 mV). The nanocomplex also showed prolonged pH-sensitive sustained drug release under conditions that replicated the tumor's extra/intracellular pH. The formulated nanocomplex also demonstrated potent radical scavenging capacity owing to the inclusion of curcumin, a known radical quencher. In addition, compared with the free compound, BMPE-Cur induced DNA fragmentation-driven cell cycle arrest in HepG2, PC3, and MCF-7-ADR cells at the G1/S, G1 & S phases; respectively, with remarkable selectivity. In conclusion, the newly formulated BMPE-Cur nanocomplex represents an attractive multitarget anticancer candidate.
Collapse
Affiliation(s)
- Heba Shawky
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki, 12622 Cairo, Egypt.
| | - Dalia B Fayed
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki, 12622 Cairo, Egypt.
| | - Noha E Ibrahim
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, Dokki, 12622 Cairo, Egypt.
| |
Collapse
|
3
|
Beigrezaei A, Rafipour R. Design of casein-based nanocarriers for targeted delivery of daunorubicin to leukemia cells. Biotechnol Appl Biochem 2024. [PMID: 39324205 DOI: 10.1002/bab.2662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 08/24/2024] [Indexed: 09/27/2024]
Abstract
Daunorubicin (DAU) is a chemotherapy drug approved for the treatment of some cancers. However, the clinical compatibility of DAU is limited due to its lack of specificity and its highly toxic effects, which interfere with normal cells. This toxicity can be reduced with nanocarriers and targeted drug delivery systems. In this study, to develop the drug delivery of DAU, the surface of synthesized nanoparticles was modified by folic acid to target cancer cells optimally. Encapsulation of DAU in protein sodium caseinate (NaCAS) was done by adding calcium ions to bring the casein (CAS) in the solution to a micellar structure to synthesize dense nanoparticles. Fourier-transform infrared spectroscopy transmission, fluorescence spectroscopy, UV-Vis spectroscopy, field emission scanning electron microscopy, and zeta potential studies designed and distinguished the synthesized nanocomplexes. The results showed that CAS nanoparticles successfully encapsulated DAU, and the protein surface was targeted by folic acid. Light scattering analysis determined that the particles with a scattering index number of 306.0 and an average size of 8.117 nm were synthesized. The zeta potential of CAS micelles is more harmful than CAS nanoparticles. This is because calcium ions are added during the formation of CAS nanoparticles during the drug-loading stages. These studies prove that the synthesized "NaCAS-DAU" and "NaCAS-DAU-folic acid" complexes can be favorable carriers in the targeted drug delivery of cancer drugs.
Collapse
Affiliation(s)
- Ali Beigrezaei
- Department of Chemistry, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Ronak Rafipour
- Department of Chemistry, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| |
Collapse
|
4
|
Elshami FI, Elrefaei G, Ibrahim MM, Elmehasseb I, Shaban SY. GSH-responsive and folate receptor-targeted pyridine bisfolate-encapsulated chitosan nanoparticles for enhanced intracellular drug delivery in MCF-7 cells. Carbohydr Res 2024; 543:109207. [PMID: 39018698 DOI: 10.1016/j.carres.2024.109207] [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: 06/13/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Folic acid receptor-targeted drug delivery system is a promising candidate for tumor-targeted delivery because its elevated expression specifically on tumor cells enables the selective delivery of cytotoxic cargo to cancerous tissue, thereby minimizing toxic side effects and increasing the therapeutic index. Pyridine bisfolate-chitosan (PyBFA@CS NPs) and folate-chitosan nanocomposite (FA@CS NPs) were synthesized with suitable particle size (256.0 ± 15.0 and 161.0 ± 5.0 nm), high stability (ζ = -27.0 ± 0.1 and -30.0 ± 0.2 mV), respectively, and satisfactory biocompatibility to target cells expressing folate receptors and try to answer the question: Is the metal center always important for activity? Since almost all pharmaceuticals work by binding to specific proteins or DNA, the in vitro binding of human serum albumin (HSA) to PyBFA@CS NPs and FA@CS NPs has been investigated and compared with PyBFA. Strong affinity to HSA is shown by quenching and binding constants in the range of 105 and 104 M-1, respectively with PyBFA@CS NPs showing the strongest. The compounds-HSA kinetic stability, affinity, and association constants were investigated using a stopped-flow method. The findings showed that all formulations bind by a static quenching mechanism that consists of two reversible steps: rapid second-order binding and a more slowly first-order isomerization reaction. The overall coordination affinity of HSA to PyBFA@CS NPs (6.6 × 106 M-1), PyBFA (4.4 × 106 M-1), and FA@CS NPs (1.3 × 106 M-1) was measured and The relative reactivity is roughly (PyBFA@CS NPs)/(PyBFA)/(FA@CS NPs) = 5/3/1. Additionally, in vitro cytotoxicity revealed that, consistent with the binding constants and coordination affinity, active-targeting formulations greatly inhibited FR-positive MCF-7 cells in compared to FRs-negative A549 cells in the following trend: PyBFA@CS NPs > PyBFA > FA@CS NPs. Furthermore, in vitro drug release of PyBFA@CS NPs was found to be stable in PBS at pH 7.4, however, the in pH 5.4 and in pH 5.4 containing 10 mM glutathione (GSH) (mimicking the tumor microenvironment) reached 43 % and 73 %, respectively indicating that the PyBFA@CS NPs system is sensitive to GSH. Folate-modified nanoparticles, PyBFA@CS NPs, are a promising therapeutic for MCF-7 therapy because they not only showed a greater affinity for HSA, but also showed higher cleavage efficiency toward the minor groove of pBR322 DNA via the hydrolytic way, as well as effective antibacterial activity that avoids the usage of extra antibiotics. .
Collapse
Affiliation(s)
- Fawzya I Elshami
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Gehad Elrefaei
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Mohamed M Ibrahim
- Department of Chemistry, College of Science, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Ibrahim Elmehasseb
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Shaban Y Shaban
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
| |
Collapse
|
5
|
Ronaghi M, Hajibeygi R, Ghodsi R, Eidi A, Bakhtiari R. Preparation of UiO-66 loaded Letrozole nano-drug delivery system: enhanced anticancer and apoptosis activity. AMB Express 2024; 14:38. [PMID: 38622436 PMCID: PMC11018590 DOI: 10.1186/s13568-024-01689-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/12/2024] [Indexed: 04/17/2024] Open
Abstract
The use of drug delivery systems in targeting and achieving the targeting of drugs in treating diseases, especially cancer, has attracted the attention of researchers. Letrozole is one of the drugs for the treatment of breast cancer. In this study, the organic-metallic pharmaceutical porous nanostructure based on zirconium UiO-66 loaded letrozole was synthesized. Its cytotoxicity and effect on apoptosis and migration against breast cancer cell line were investigated. In this experimental study, the UiO-66 nanoparticle-loaded letrozole was synthesized using zirconium chloride (ZrCl4), dimethylformamide (DMF), and HCl. Its characteristics were determined by scanning electron microscopy, and its average size was determined by the DLS method. Also, the rate of letrozole drug release from the nanoparticle was investigated in 24, 48, and 72 h. In addition, its cytotoxicity effects were investigated using the MTT colorimetric method at concentrations of 3.125-100 µg/ml against the breast cancer cell line (MCF-7) in the periods of 48 and 72 h. Also, the expression level of apoptotic genes Bax and Bcl2 was investigated by the Real-Time PCR method. Also, the amount of cell migration was done by the migration assay method. The results showed that UiO-66 bound to letrozole had a spherical morphology and an average size of 9.2 ± 160.1. Also, the letrozole drug was loaded by 62.21 ± 1.80% in UiO-66 nanoparticles and had a slower release pattern than free letrozole in the drug release test, so within 72 h, 99.99% of free letrozole was released in If in UiO-66 containing letrozole, 57.55% of the drug has been released. Also, the cytotoxicity results showed that UiO-66 bound to letrozole has more significant cytotoxic effects than free letrozole (p < 0.05). Also, the results of Bax and Bcl2 gene expression showed that the treatment of MCF-7 cells with UiO-66 nanoparticles attached to letrozole increased the expression of Bax and Bcl2 genes compared to the reference gene Beta-actin in MCF-7 cell line, respectively. (p < 0.05) increased by 3.71 ± 0.42 and (p < 0.01) decreased by 0.636 ± 0.034 (p < 0.05). Cell migration results showed that the concentration of 50 µg/ml of UiO-66 bound to letrozole decreased the migration of MCF-7 cells. Generally, the results of this study showed that UiO-66 loaded letrozole can be used as a suitable drug carrier for cellular purposes, as it has increased the effects of cytotoxicity and the rate of apoptosis in breast cancer cell line (MCF-7), so it can be used with more studies used nanocarriers as a drug delivery system.
Collapse
Affiliation(s)
- Maryam Ronaghi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ramtin Hajibeygi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Reza Ghodsi
- Department of Chemical and Petrochemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Akram Eidi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ronak Bakhtiari
- Department of Pathobiology, Division of Microbiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Faddah H, Nsairat H, Shalan NM, El-Tanani M, Alqudah DA, Alshaer W. Preparation, Optimization and In vitro Evaluation of Doxorubicin-loaded into Hyaluronic Acid Coated Niosomes Against Breast Cancer. Chem Biodivers 2024; 21:e202301470. [PMID: 38161147 DOI: 10.1002/cbdv.202301470] [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: 09/20/2023] [Revised: 12/28/2023] [Accepted: 12/31/2023] [Indexed: 01/03/2024]
Abstract
Doxorubicin (DOX) is widely used against solid tumors. Niosomes are self-assembled nanocarriers of non-ionic surfactants. DOX loaded into cationic niosomes (DOX-Nio) was prepared via thin film hydration method. DOX-Nio was then decorated with a hyaluronic acid (DOX-HA-Nio) via electrostatic interaction. DOX-Nio and DOX-HA-Nio displayed a particle size of 120.0±1.02 and 182.9±2.3 nm, and charge of + 35.5±0.15 and -15.6±0.25 mV, respectively, with PDI < 0.3. DOX-HA-Nio showed a good stability regarding size and charge over 4 weeks at 4 °C and maintain their integrity after lyophilization. HPLC results showed a 94.1±4.2 % encapsulation efficiency of DOX with good entrapment and slow, prolonged DOX release even after 48 hrs. Cell viability assay showed an IC50 of 14.26 nM for the DOX-HA-Nio against MCF-7 cell line with micromolar IC50 results against CD-44 negative cell lines (NIH/3T3). DOX-HA-Nio was proven to be an effective, targeted nanocarrier for DOX against MCF-7 cell line.
Collapse
Affiliation(s)
- Haya Faddah
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Naeem M Shalan
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Dana A Alqudah
- Cell Therapy Center, the University of Jordan, Amman, 11942, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, the University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
7
|
Kumar A, Gupta GD, Kumar M. Magnetomorph: The Future of Targeted Drug Delivery. Curr Drug Targets 2024; 25:449-453. [PMID: 38639288 DOI: 10.2174/0113894501309729240414081308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024]
Affiliation(s)
- Ashutosh Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
8
|
Hien TT, Ambite I, Wan MLY, Cavalera M, Esmaeili P, Chaudhuri A, Sabari S, Babjuk M, Svanborg C. Long-term prevention of bladder cancer progression by alpha1-oleate alone or in combination with chemotherapy. Int J Cancer 2023; 153:584-599. [PMID: 36891980 DOI: 10.1002/ijc.34500] [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: 10/31/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023]
Abstract
Bladder cancer is common and one of the most costly cancer forms, due to a lack of curative therapies. Recently, clinical safety and efficacy of the alpha1-oleate complex was demonstrated in a placebo-controlled study of nonmuscle invasive bladder cancer. Our study investigated if long-term therapeutic efficacy is improved by repeated treatment cycles and by combining alpha1-oleate with low-dose chemotherapy. Rapidly growing bladder tumors were treated by intravesical instillation of alpha1-oleate, Epirubicin or Mitomycin C alone or in combination. One treatment cycle arrested tumor growth, with a protective effect lasting at least 4 weeks in mice receiving 8.5 mM of alpha1-oleate alone or 1.7 mM of alpha-oleate combined with Epirubicin or Mitomycin C. Repeated treatment cycles extended protection, defined by a lack of bladder pathology and a virtual absence of bladder cancer-specific gene expression. Synergy with Epirubicin was detected at the lower alpha1-oleate concentration and in vitro, alpha1-oleate was shown to enhance the uptake and nuclear translocation of Epirubicin, by tumor cells. Effects at the chromatin level affecting cell proliferation were further suggested by reduced BrdU incorporation. In addition, alpha1-oleate triggered DNA fragmentation, defined by the TUNEL assay. The results suggest that bladder cancer development may be prevented long-term in the murine model, by alpha1-oleate alone or in combination with low-dose Epirubicin. In addition, the combination of alpha1-oleate and Epirubicin reduced the size of established tumors. Exploring these potent preventive and therapeutic effects will be of immediate interest in patients with bladder cancer.
Collapse
Affiliation(s)
- Tran Thi Hien
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ines Ambite
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Murphy Lam Yim Wan
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Michele Cavalera
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Parisa Esmaeili
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Arunima Chaudhuri
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Samudra Sabari
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Marek Babjuk
- Department of Urology, Motol Hospital and Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
9
|
Huang C, Zhang Z, Gu J, Li D, Gao S, Zhang R, Shi R, Sun J. Combined Therapy of Experimental Autoimmune Uveitis by a Dual-Drug Nanocomposite Formulation with Berberine and Dexamethasone. Int J Nanomedicine 2023; 18:4347-4363. [PMID: 37545873 PMCID: PMC10402891 DOI: 10.2147/ijn.s417750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
Purpose Autoimmune uveitis is a kind of sight-threatening ocular and systemic disorders. Recent treatments on autoimmune uveitis still remain many limitations due to extreme complexity and undetermined pathogenesis. In this study, a novel dual-drug nanocomposite formulation is developed to treat experimental autoimmune uveitis by a combined and sustained therapy method. Methods The dual-drug nanocomposite formulation is constructed by integrating berberine (BBR)-loaded mesoporous silica nanoparticles (MSNs) into dexamethasone (DEX)-loaded thermogel (BBR@MSN-DEX@Gel). The BBR@MSN-DEX@Gel is characterized by transmission electron microscopy, dynamic light scattering, Fourier transform infrared spectrometer and rheometer. The in vitro drug release profile, cytotoxicity and anti-inflammation effectiveness of BBR@MSN-DEX@Gel on lipopolysaccharide-stimulated human conjunctival epithelial cells are investigated. After the in vivo drug release profile and biosafety of the dual-drug nanocomposite formulation are confirmed, its treatment effectiveness is fully assessed based on the induced experimental autoimmune uveitis (EAU) Lewis rat's model. Results The dual-drug nanocomposite formulation has good injectability and thermosensitivity, suitable for administration by an intravitreal injection. The BBR@MSN-DEX@Gel has been found to sustainably release both drugs for up to 4 weeks. The carrier materials have minimal in vitro cytotoxicity and high in vivo biosafety. BBR@MSN-DEX@Gel presents obviously anti-inflammatory effectiveness in vitro. After administration of BBR@MSN-DEX@Gel into Lewis rat's eye with EAU by an intravitreal injection, the nanocomposite formulation significantly suppresses inflammatory reaction of autoimmune uveitis via a dual-drug combined and sustained therapy method, compared with the equivalent dose of single-component formulations. Conclusion BBR@MSN-DEX@Gel serves as a promising dual-drug nanocomposite formulation for future treatment of autoimmune uveitis.
Collapse
Affiliation(s)
- Chang Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, People’s Republic of China
| | - Zhutian Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, People’s Republic of China
| | - Jifeng Gu
- Department of Pharmacy, Eye & ENT Hospital, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, 200031, People’s Republic of China
| | - Dan Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, People’s Republic of China
| | - Shunxiang Gao
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, People’s Republic of China
| | - Rong Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, People’s Republic of China
| | - Rong Shi
- Science and Technology Experimental Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Jianguo Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, People’s Republic of China
| |
Collapse
|
10
|
Kharazmi A, Attaran N. Evaluation of the parameters affecting the loading of anticancer drug Paclitaxel on coated gold nanoparticles for breast cancer treatment. IET Nanobiotechnol 2023; 17:234-245. [PMID: 36849875 DOI: 10.1049/nbt2.12121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 03/01/2023] Open
Abstract
The purpose of this study is the design and synthesis of gold nanoparticles (GNPs) conjugated with paclitaxel and to investigate the parameters affecting the stability of synthesised nanoparticles with drug delivery capability. Here, synthesised GNPs were coated with polyethylene glycol. Then these particles were conjugated with paclitaxel under different conditions and the physical and structural characteristics, as well as the factors affecting the loading of paclitaxel on nanoparticles, were evaluated by ultraviolet spectrophotometer, fourier transform infrared spectroscopy, transmission electron microscopy, dynamic light scattering and zeta potential apparatus. It was found that pegylated GNPs have a limited loading capacity at the time of 24 h of incubation and the Paclitaxel loading was observed to be pH dependent. The use of these particles in the treatment of breast cancer (MCF7) was also investigated using the MTT test. It was determined that the survival percentage of MCF7 cells in the presence of paclitaxel-bound nanoparticles decreases to about 55% at the maximum measured concentration (690 μM).
Collapse
Affiliation(s)
- Afrooz Kharazmi
- Department of Biomedical Engineering, Applied Biophotonics Research Center, Tehran Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Neda Attaran
- Department of Medical Nanotechnology, Applied Biophotonics Research Center, Tehran Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
11
|
Hamrang R, Moniri E, Heydarinasab A, Safaeijavan R. In vitro evaluation of copper sulfide nanoparticles decorated with folic acid/chitosan as a novel pH-sensitive nanocarrier for the efficient controlled targeted delivery of cytarabine as an anticancer drug. Biotechnol Appl Biochem 2023; 70:330-343. [PMID: 35561253 DOI: 10.1002/bab.2355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 04/21/2022] [Indexed: 12/07/2022]
Abstract
Nanoparticles (NPs) have gained more attention as drug delivery systems. Folic acid (FA)-chitosan (CS) conjugates, because of their biodegradability, low toxicity, and better stability, offer a pharmaceutical drug delivery tool. The aim of this work was to fabricate CuS NPs modified by CS followed by grafting FA as a nanocarrier for the delivery of cytarabine (CYT) as an anticancer drug. In this work, CuS NPs modified by CS and FA were successfully synthesized. The structural properties of the nanocarrier were characterized by using scanning electron microscopy, Fourier transform infrared, X-ray diffraction, thermogravimetric analysis, and Brunauer-Emmett-Teller. The adsorption mechanism of CYT by adsorption isotherms, kinetics, and thermodynamics was deliberated and modeled. The in vitro CYT release behavior for the nanocarrier was 99% and 61% at pH 5.6 and 7.4, respectively. The adsorption behavior of CYT by CuS NPs -CS-FA was well explored by pseudo-second-order kinetic and Langmuir isotherm models by the coefficient of determination (R2 > 0.99). Thermodynamic results showed that the uptake of CYT by CuS NPs-CS-FA was endothermic and spontaneous. The experimental results showed that CYT/CuS NPs -CS-FA can be proposed as an efficient nanocarrier for the targeted delivery of anticancer drugs.
Collapse
Affiliation(s)
- Roya Hamrang
- Department of Chemical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Elham Moniri
- Department of Chemistry, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Amir Heydarinasab
- Department of Chemical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Raheleh Safaeijavan
- Department of Biochemistry and Biophysics, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| |
Collapse
|
12
|
Shao Z, Li R, Shao D, Tang H, Han Y. Albumin-Based Zn (II)-Quercetin Enzyme Mimic Scavenging ROS for Protection against Cardiotoxicity Induced by Doxorubicin. Pharmaceuticals (Basel) 2022; 15:ph15121524. [PMID: 36558975 PMCID: PMC9781925 DOI: 10.3390/ph15121524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent that can cause cardiotoxicity leading to progressive, chronic, life-threatening cardiomyopathy, called DOX-induced cardiomyopathy (DIC). DIC is a fatal cardiomyopathy with a worse prognosis compared to other cardiomyopathies and limits the use of DOX in malignancies due to its cardiotoxicity. DIC has been proven to be associated with reactive oxygen species (ROS)-induced side effect damage in cardiac myocytes. Currently, scavenging of reactive oxygen species is a practical strategy to reduce chemotherapy-associated DIC. Although quercetin has already been reported to have superior antioxidant activity, its clinical application is severely limited due to its rapid degradation and poor tissue absorption. Herein, we reported the preparation of a novel enzyme mimic via coordinated albumin, Zinc Ion (Zn2+) and quercetin. The enzyme mimics were capable of simultaneously increasing the biocompatibility and efficiently overcame the drawbacks of free quercetin, and were achieved by long circulation in vivo. Most importantly, these quercetin-based enzyme mimics had no effect on the antioxidant activity of quercetin. These beneficial therapeutic properties, together with high drug-carrying capacity and redox stimuli, will significantly improve quercetin's alleviation of chemotherapeutic cardiotoxicity without causing significant side effects. Therefore, nanoparticles of albumin-based Zn (II)-Quercetin have a promising clinical application as an effective agent for mitigating the cardiotoxicity of chemotherapy.
Collapse
Affiliation(s)
- Zehua Shao
- Heart Center of Zhengzhou University People’s Hospital, Fuwai Central China Cardiovascular Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China
| | - Ran Li
- Heart Center of Zhengzhou University People’s Hospital, Fuwai Central China Cardiovascular Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou 451464, China
| | - Dongxing Shao
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou 451464, China
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Hao Tang
- Heart Center of Zhengzhou University People’s Hospital, Fuwai Central China Cardiovascular Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou 451464, China
- Correspondence: (H.T.); (Y.H.)
| | - Yu Han
- Heart Center of Zhengzhou University People’s Hospital, Fuwai Central China Cardiovascular Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Correspondence: (H.T.); (Y.H.)
| |
Collapse
|
13
|
Jadhav M, Prasad R, Gandhi M, Srivastava R. Erythrocyte nanovesicles as chemotherapeutic drug delivery platform for cancer therapy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
14
|
Arjama M, Mehnath S, Jeyaraj M. Self-assembled hydrogel nanocube for stimuli responsive drug delivery and tumor ablation by phototherapy against breast cancer. Int J Biol Macromol 2022; 213:435-446. [PMID: 35661669 DOI: 10.1016/j.ijbiomac.2022.05.190] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 12/14/2022]
Abstract
The shape and responsiveness of nanoengineered delivery carriers are crucial characteristics for the rapid and efficient delivery of therapeutics. We report on a novel type of micrometer-sized hydrogel particles of controlled shape with dual pH and redox sensitivity for intracellular delivery of anticancer drugs and phototherapy. The cubical HA-DOP-CS-PEG networks with disulfide links are obtained by cross-linking HA-DOP-CS-PEG with cystamine. The pH-triggered hydrogel swelling/shrinkage was not only affords effective doxorubicin release. It also actively provides the endosomal/lysosomal escape, redox-triggered drug release. The hydrogels degrade rapidly to low molecular weight chains in the presence of the typical intracellular concentration of glutathione. Drug-loaded cube particles found to be 12% more cytotoxic. ICG and DOX-loaded hydrogel cubes demonstrate 90% cytotoxicity when incubated with MCF-7 cancer cells for 24 and 48 h, respectively. This approach integrates the advantages of pH sensitivity, enzymatic degradation, and shape-regulated internalization for novel types of "intelligent" three-dimensional networks with programmable behavior for controlled delivery of therapeutics.
Collapse
Affiliation(s)
- Mukherjee Arjama
- National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai 25, Tamil Nadu, India
| | - Sivaraj Mehnath
- National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai 25, Tamil Nadu, India
| | - Murugaraj Jeyaraj
- National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai 25, Tamil Nadu, India.
| |
Collapse
|
15
|
Habibullah MM, Mohan S, Syed NK, Makeen HA, Jamal QMS, Alothaid H, Bantun F, Alhazmi A, Hakamy A, Kaabi YA, Samlan G, Lohani M, Thangavel N, Al-Kasim MA. Human Growth Hormone Fragment 176–191 Peptide Enhances the Toxicity of Doxorubicin-Loaded Chitosan Nanoparticles Against MCF-7 Breast Cancer Cells. Drug Des Devel Ther 2022; 16:1963-1974. [PMID: 35783198 PMCID: PMC9249349 DOI: 10.2147/dddt.s367586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/15/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Numerous drugs with potent toxicity against cancer cells are available for treating malignancies, but therapeutic efficacies are limited due to their inefficient tumor targeting and deleterious effects on non-cancerous tissue. Therefore, two improvements are mandatory for improved chemotherapy 1) novel delivery techniques that can target cancer cells to deliver anticancer drugs and 2) methods to specifically enhance drug efficacy within tumors. The loading of inert drug carriers with anticancer agents and peptides which are able to bind (target) tumor-related proteins to enhance tumor drug accumulation and local cytotoxicity is a most promising approach. Objective To evaluate the anticancer efficacy of Chitosan nanoparticles loaded with human growth hormone hGH fragment 176–191 peptide plus the clinical chemotherapeutic doxorubicin in comparison with Chitosan loaded with doxorubicin alone. Methods Two sets of in silico experiments were performed using molecular docking simulations to determine the influence of hGH fragment 176–191 peptide on the anticancer efficacy of doxorubicin 1) the binding affinities of hGH fragment 176–191 peptide to the breast cancer receptors, 2) the effects of hGH fragment 176–191 peptide binding on doxorubicin binding to these same receptors. Further, the influence of hGH fragment 176–191 peptide on the anticancer efficacy of doxorubicin was validated using viability assay in Human MCF-7 breast cancer cells. Results In silico analysis suggested that addition of the hGH fragment to doxorubicin-loaded Chitosan nanoparticles can enhance doxorubicin binding to multiple breast cancer protein targets, while photon correlation spectroscopy revealed that the synthesized dual-loaded Chitosan nanoparticles possess clinically favorable particle size, polydispersity index, as well as zeta potential. Conclusion These dual-loaded Chitosan nanoparticles demonstrated greater anti-proliferative activity against a breast cancer cell line (MCF-7) than doxorubicin-loaded Chitosan. This dual-loading strategy may enhance the anticancer potency of doxorubicin and reduce the clinical side effects associated with non-target tissue exposure.
Collapse
Affiliation(s)
- Mahmoud M Habibullah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
- Correspondence: Mahmoud M Habibullah, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Al Maarefah Road, Jazan, Saudi Arabia, Tel +966 556644205, Email
| | - Syam Mohan
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, Saudi Arabia
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Nabeel Kashan Syed
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Farkad Bantun
- Department of Microbiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Alaa Alhazmi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
| | - Ali Hakamy
- SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
- Respiratory Therapy Department, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Yahia A Kaabi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Ghalia Samlan
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohtashim Lohani
- Emergency Medical Services Department, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Neelaveni Thangavel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mohamed Ahmed Al-Kasim
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
16
|
Abbasi M, Sohail M, Minhas MU, Iqbal J, Mahmood A, Shaikh AJ. Folic acid-functionalized nanoparticles-laden biomaterials for the improved oral delivery of hydrophobic drug in colorectal cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
17
|
Xu X, Tian K, Lou X, Du Y. Potential of Ferritin-Based Platforms for Tumor Immunotherapy. Molecules 2022; 27:2716. [PMID: 35566065 PMCID: PMC9104857 DOI: 10.3390/molecules27092716] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/14/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
Ferritin is an iron storage protein that plays a key role in iron homeostasis and cellular antioxidant activity. Ferritin has many advantages as a tumor immunotherapy platform, including a small particle size that allows for penetration into tumor-draining lymph nodes or tumor tissue, a unique structure consisting of 24 self-assembled subunits, cavities that can encapsulate drugs, natural targeting functions, and a modifiable outer surface. In this review, we summarize related research applying ferritin as a tumor immune vaccine or a nanocarrier for immunomodulator drugs based on different targeting mechanisms (including dendritic cells, tumor-associated macrophages, tumor-associated fibroblasts, and tumor cells). In addition, a ferritin-based tumor vaccine expected to protect against a wide range of coronaviruses by targeting multiple variants of SARS-CoV-2 has entered phase I clinical trials, and its efficacy is described in this review. Although ferritin is already on the road to transformation, there are still many difficulties to overcome. Therefore, three barriers (drug loading, modification sites, and animal models) are also discussed in this paper. Notwithstanding, the ferritin-based nanoplatform has great potential for tumor immunotherapy, with greater possibility of clinical transformation.
Collapse
Affiliation(s)
- Xiaoling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (X.X.); (K.T.)
| | - Kewei Tian
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (X.X.); (K.T.)
| | - Xuefang Lou
- School of Medicine, Zhejiang University City College, Hangzhou 310015, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
18
|
Kuruppu AI, Turyanska L, Bradshaw TD, Manickam S, Galhena BP, Paranagama P, De Silva R. Apoferritin and Dps as drug delivery vehicles: Some selected examples in oncology. Biochim Biophys Acta Gen Subj 2022; 1866:130067. [PMID: 34896255 DOI: 10.1016/j.bbagen.2021.130067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/27/2021] [Accepted: 12/02/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND The ideal nanoparticle should be able to encapsulate either pharmaceutical agents or imaging probes so that it could treat or image clinical tumours by targeting the cancer site efficiently. Further, it would be an added advantage if it demonstrates: small size, built in targeting, biocompatibility and biodegradability. Ferritin, which is an endogenous self-assembling protein, stores iron and plays a role in iron homeostasis. When iron atoms are removed apoferritin (AFt) is formed which consists of a hollow shell where it can be used to load guest molecules. Due to its unique architecture, AFt has been investigated as a versatile carrier for tumour theranostic applications. DNA-binding protein from starved cells (Dps), which also belongs to the ferritin family, is a protein found only in prokaryotes. It is used to store iron and protect chromosomes from oxidative damage; because of its architecture, Dps could also be used as a delivery vehicle. CONCLUSIONS Both these nano particles are promising in the field of oncology, especially due to their stability, solubility and biocompatibility features. Further their exterior surface can be modified for better tumour-targeting ability. More studies, are warranted to determine the immunogenicity, biodistribution, and clearance from the body. GENERAL PERSPECTIVE This review discusses a few selected examples of the remarkable in vitro and in vivo studies that have been carried out in the recent past with the use of AFt and Dps in targeting and delivery of various pharmaceutical agents, natural products and imaging probes in the field of oncology.
Collapse
Affiliation(s)
- Anchala I Kuruppu
- Institute for Combinatorial Advanced Research & Education, General Sir John Kotelawala Defence University, Sri Lanka.
| | | | | | - Sivakumar Manickam
- Petroleum and Chemical Engineering, Faculty of Engineering, Universiti Teknologi Brunei, Brunei Darussalam
| | - Bandula Prasanna Galhena
- Department Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Kelaniya, Sri Lanka
| | - Priyani Paranagama
- Department of Chemistry, Faculty of Science, University of Kelaniya, Sri Lanka; Institute of Indigenous Medicine, University of Colombo, Sri Lanka
| | - Ranil De Silva
- Institute for Combinatorial Advanced Research & Education, General Sir John Kotelawala Defence University, Sri Lanka
| |
Collapse
|
19
|
Luiz MT, Dutra JAP, Di Filippo LD, Junior AGT, Tofani LB, Marchetti JM, Chorilli M. Epirubicin: Biological Properties, Analytical Methods, and Drug Delivery Nanosystems. Crit Rev Anal Chem 2021; 53:1080-1093. [PMID: 34818953 DOI: 10.1080/10408347.2021.2007469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Epirubicin (EPI) is a chemotherapeutic agent belonging to the anthracycline drug class indicated for treating several tumors. It acts by suppressing the DNA and RNA synthesis by intercalating between their base pair. However, several side effects are associated with this therapy, including cardiotoxicity and myelosuppression. Therefore, EPI delivery in nanosystems has been an interesting strategy to overcome these limitations and improve the safety and efficacy of EPI. Thus, analytical methods have been used to understand and characterize these nanosystems, including spectrophotometric, spectrofluorimetric, and chromatography. Spectrophotometric and spectrofluorimetric methods have been used to quantify EPI in less complex matrices due to their efficiency, low cost, and green chemistry character. By contrast, high-performance liquid chromatography is a suitable method for detecting EPI in more complex matrices (e.g., plasm and urine) owing to its high sensitivity. This review summarizes physicochemical and pharmacokinetic properties of EPI, its application in drug delivery nanosystems, and the analytical methods employed in its quantification in different matrices, including blood, plasm, urine, and drug delivery nanosystems.
Collapse
Affiliation(s)
- Marcela Tavares Luiz
- School of Pharmaceutical Science of Ribeirao Preto, University of São Paulo (USP), Ribeirao Preto, São Paulo, Brazil
| | | | | | | | - Larissa Bueno Tofani
- School of Pharmaceutical Science of Ribeirao Preto, University of São Paulo (USP), Ribeirao Preto, São Paulo, Brazil
| | - Juliana Maldonado Marchetti
- School of Pharmaceutical Science of Ribeirao Preto, University of São Paulo (USP), Ribeirao Preto, São Paulo, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Science of São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
20
|
Mohammed ASY, Dyab AKF, Taha F, Abd El-Mageed AIA. Encapsulation of folic acid (vitamin B 9) into sporopollenin microcapsules: Physico-chemical characterisation, in vitro controlled release and photoprotection study. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112271. [PMID: 34474830 DOI: 10.1016/j.msec.2021.112271] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/09/2021] [Accepted: 06/21/2021] [Indexed: 02/03/2023]
Abstract
Folic acid (FA) is a crucial vitamin for all living creatures. However, it is susceptible to degradation under pH, heat, ultraviolet (UV) and day sunlight conditions, resulting in lowering its bioavailability. Therefore, a versatile protective encapsulation system for FA is highly required to overcome its inherent instability. We report the use of the robust Lycopodium clavatum sporopollenin (LCS) microcapsules, extracted from their natural micrometer-sized raw spores, for FA microencapsulation. The physico-chemical characterisation of the LCS microcapsules are comprehensively investigated before and after the microencapsulation using SEM, elemental, CLSM, FTIR, TGA/DTG and XRD analyses, revealing a successful FA encapsulation within the LCS in an amorphous form. The phenylpropanoid acids, responsible for the UV protection and the autofluorescence of the LCS, were found in the LCS as evidenced by FTIR analysis. TGA/DTG results revealed that the hemi-cellulose and cellulose are the major component of the LCS. A controlled and sustained release of FA from FA-loaded LCS were achieved where the release profile of FA-loaded LCS was found to be pH-dependent. The percentages of cumulative FA released after 10 h at 37 ± 0.5 °C were 45.5% and 76.1% in pH 1.2 and 7.4, respectively, ensuring controlled and slow release in simulated physiological conditions. The FA release kinetic studies indicated the prevalence of the Fickian diffusion mechanism in pH 1.2, while anomalous non-Fickian transport was ascribed for FA release in pH 7.4. The in vitro cytotoxicity assay revealed that the obtained formulations were biocompatible against the human skin fibroblast (HSF) cell line. The versatile LCS microcapsules exhibited intriguing photostability for FA under UV or sunlight irradiation. Concretely, the obtained FA sustained delivery and photoprotection properties of these LCS microcapsules validate their multifunctional characteristics, opening up intriguing applications in oral and topical drug delivery as well as in food industry.
Collapse
Affiliation(s)
- Al-Shymaa Y Mohammed
- Colloids & Advanced Materials Group, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt
| | - Amro K F Dyab
- Colloids & Advanced Materials Group, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt.
| | - Fouad Taha
- Colloids & Advanced Materials Group, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt
| | - Ahmed I A Abd El-Mageed
- Colloids & Advanced Materials Group, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt; Nanoscience and Technology, Faculty of Science, GALALA University, Galala City, Suez 43711, Egypt
| |
Collapse
|
21
|
Amer Ridha A, Kashanian S, Rafipour R, Hemati Azandaryani A, Zhaleh H, Mahdavian E. A promising dual-drug targeted delivery system in cancer therapy: nanocomplexes of folate-apoferritin-conjugated cationic solid lipid nanoparticles. Pharm Dev Technol 2021; 26:673-681. [PMID: 33896342 DOI: 10.1080/10837450.2021.1920037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Various nano-sized protein and lipid complexes are being investigated as drug delivery systems. The encapsulation of more than one drug in a single nanocomplex carrier could enhance the therapeutic potency and afford synergistic therapeutic effects. In this study, we developed a novel protein-lipid nanocomplex as a controlled drug delivery system for two important cancer drugs, doxorubicin (DOX) and mitoxantrone (MTO). Apoferritin (AFr) functionalized with folic acid (FA) was used to encapsulate DOX to create the targeted protein nanocomplexes (TPNs). The second drug, MTO, was loaded into the cationic solid lipid nanoparticles (cSLN) to form the liposomal drug nanocomplex particles (MTO-cSLNs). Two complexes were then assembled by tight coupling through ionic interactions to obtain the final drug delivery system, the dual-targeted protein-lipid nanocomplexes (DTPLNs). UV-Vis and fluorescence spectroscopy were used for structural characterization of TPNs and DTPLNs. Transmission electron microscopy (TEM) was used for comprehensive analysis of the final DTPLNs. We confirmed that the DTPLNs display desired time-dependent and pH-dependent drug release behaviors. We also demonstrated the improved anti-cancer efficacy of DOX and MTO in their encapsulated DTPLNs as compared to their free forms. Our results provide promising prospects for the application of the DTPLNs as efficient drug delivery systems.
Collapse
Affiliation(s)
- Abbas Amer Ridha
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran.,Iraqi Ministry of Health, Baghdad, Iraq
| | - Soheila Kashanian
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran.,Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ronak Rafipour
- Department of Chemistry, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | | | - Hossein Zhaleh
- Substance Abuse Prevention Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elahe Mahdavian
- Department of Chemistry and Physics, Louisiana State University in Shreveport, Shreveport, LA, USA
| |
Collapse
|
22
|
Rafipour R, Mousavi A, Mansouri K. Apoferritin nanocages for targeted delivery of idarubicin against breast cancer cells. Biotechnol Appl Biochem 2021; 69:1061-1067. [PMID: 33929766 DOI: 10.1002/bab.2177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/20/2021] [Indexed: 11/10/2022]
Abstract
In recent years, nanotechnology has attracted attention for its capability to diagnose and remedy diverse tumors successfully. Protein nanocarriers as a platform of targeted drug delivery can be used to reduce toxicity and improve the effect of anticancer drugs. Idarubicin (IDR) is a chemotherapy drug that is classified as an anthracycline antitumor. In this study, IDR was encapsulated within horse spleen apoferritin (HsAFr) nanocarriers. Encapsulation was obtained through disassembling apoferritin into subunits at pH 2 and subsequently reassembling it at pH 7.4 in the presence of IDR. Transmission electron microscopy, UV-vis, and fluorescence spectroscopy techniques showed that drug molecules are loaded within apoferritin. Intrinsic fluorescence information exhibited that the encapsulation does not have any effects on the tertiary structure of the protein. Drug loading and entrapment efficiency were found to be 7.15% and 84.75%, respectively. Comparison of anticancer activities in HsAFr-IDR and free drug IDR was made via the MTT viability technique in a human breast cancer cell line (MCF-7).
Collapse
Affiliation(s)
- Ronak Rafipour
- Department of Chemistry, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Asma Mousavi
- Department of Chemistry, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
23
|
Veroniaina H, Pan X, Wu Z, Qi X. Apoferritin: a potential nanocarrier for cancer imaging and drug delivery. Expert Rev Anticancer Ther 2021; 21:901-913. [PMID: 33844625 DOI: 10.1080/14737140.2021.1910027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Introduction: As a protein-based biomaterial for potential cancer targeting delivery, apoferritin has recently attracted interest.Areas covered: In this review, we discuss the development of this cage-like protein as an endogenous nanocarrier that can hold molecules in its cavity. We present the specific characterizations and formulations of apoferritin nanocarriers, and outline the recent progress of the protein as an appropriate tumor-delivery vehicle in different therapeutic strategies to treat solid tumors. Finally, we propose how the application for cancer drug repurposing delivery within apoferritin could expand cancer treatment in the future.Expert opinion: Being a ubiquitous iron storage protein that exists in many living organisms, apoferritin is promising as a cancer tumor-targeting nanocarrier. By exploiting its versatility, apoferritin could be used for cancer repurposed drug delivery and could reduce the high cost of new drug discovery development and shorten the formulation process.
Collapse
Affiliation(s)
| | - Xiuhua Pan
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
24
|
Ștefan G, Hosu O, De Wael K, Lobo-Castañón MJ, Cristea C. Aptamers in biomedicine: Selection strategies and recent advances. Electrochim Acta 2021. [DOI: 10.1016/j.electacta.2021.137994] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
Zhang Y, Zhou T, Li J, Xu N, Cai M, Zhang H, Zhao Q, Wang S. Au Catalyzing Control Release NO in vivo and Tumor Growth-Inhibiting Effect in Chemo-Photothermal Combination Therapy. Int J Nanomedicine 2021; 16:2501-2513. [PMID: 33824588 PMCID: PMC8018432 DOI: 10.2147/ijn.s270466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/27/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Aim to obtain a NO donor that can control released NO in vivo with the high efficacy of tumor suppression and targeting, a nanoplatform consisting of FA-Fe3O4@mSiO2-Au/DOX was constructed. METHODS In vitro, the nanoplatform catalyzed NO's release with the maximum value of 4.91 μM within 60 min at 43°C pH=5.0, which was increased by 1.14 times when the temperature was 37°C. In vivo, 11.7 μg Au in the tumor tissue was found to catalyze S-nitrosoglutathione continuously, and 54 μM NO was checked out in the urine. RESULTS AND DISCUSSION The high concentration of NO was found to increase the apoptotic rate and to reduce tumor proliferation. In the chemo-photothermal combination therapy, the tumor inhibition rate was increased up to 94.3%, and Au's contribution from catalyzing NO release NO was 8.17%.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Tianfu Zhou
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Jian Li
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Nuo Xu
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Mingze Cai
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Hong Zhang
- Van ’T Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, 1098 XH, the Netherlands
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| |
Collapse
|
26
|
Bayat P, Pakravan P, Salouti M, Ezzati Nazhad Dolatabadi J. Lysine Decorated Solid Lipid Nanoparticles of Epirubicin for Cancer Targeting and Therapy. Adv Pharm Bull 2021; 11:96-103. [PMID: 33747856 PMCID: PMC7961234 DOI: 10.34172/apb.2021.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/29/2020] [Accepted: 04/19/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose: Cancer is an example of the most important growing diseases in human society and scientists are trying to treat it without considerable side effects on patient’s health. Solid lipids are colloidal nanoparticles that were used in drug delivery due to their several advantages. Methods: In this work, surface modified targeted solid lipid nanoparticles (SLNs) were fabricated by nano-homogenizer using tripalmitin glyceride and stearic acid as lipid constituents. The size of nanoparticles and morphological evaluations were surveyed using particle size analyzer, scanning electron microscopy; Fourier transforms infrared spectroscopy (FT-IR) and differential scanning calorimetry (DSC). Results: The particle size of 148.5 and appropriate polydispersity index were achieved for lipid nanoparticles with an entrapment efficiency of 86.1%. The FT-IR analysis confirmed the coupling of lysine to the free functional group of SLNs. DSC proved the conjugation of amino acid to the surface of carriers. The in vitro epirubicin (EPI) release test exhibited the further controlled release phenomenon for the lysine conjugated nanoparticles. The cytotoxicity assay showed lower IC50 of lysine conjugated SLNs of EPI on the investigated cell line. Conclusion: These studies showed that the fabricated targeted carrier has a very remarkable anticancer effect on breast cancer cell lines in comparison with pure drug.
Collapse
Affiliation(s)
- Parvaneh Bayat
- Department of Chemistry, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | - Parvaneh Pakravan
- Department of Chemistry, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | - Mojtaba Salouti
- Nanobiotechnology Research Center, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | | |
Collapse
|
27
|
Blue light-triggered Fe 2+-release from monodispersed ferrihydrite nanoparticles for cancer iron therapy. Biomaterials 2021; 271:120739. [PMID: 33690102 DOI: 10.1016/j.biomaterials.2021.120739] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/23/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Site-specific Fe2+ generation is promising for tumor therapy. Up to now, reported materials or systems for Fe2+ delivery do not naturally exist in the body, and their biological safety and toxicity are concerned. Herein, inspired by the natural biomineral ferrihydrite in ferritin, we synthesized monodispersed ferrihydrite nanoparticles and demonstrated a light triggered Fe2+ generation on tumor sites. Ferrihydrite nanoparticles of 20-30 nm in diameter possessed high cellular uptake efficiency and good biocompatibility. Under common blue light illumination, a large amount of Fe2+ could be released from ferrihydrite and promote the iron/reactive oxygen species (ROS)-related irreversible DNA fragmentation and glutathione peroxidase 4 (GPX4) inhibition, which led to the apoptosis- and ferroptosis-depended cancer cell proliferation inhibition. On mice, this method induced tumor associated macrophage (TAM) polarization from the tumor-promoting M2 type to the tumor-killing M1 type. With the intravenous pre-injection of ferrihydrite, the combinational effects of the light/Fe2+-approach attenuated pulmonary metastasis on mice. These results demonstrated a novel external light controlled Fe2+-generation approach based on biomineral, which will fully tap the anti-cancer potential of Fe2+ in chemo-dynamic, photo-dynamic and immune-activating therapies.
Collapse
|
28
|
Wang B, Guo C, Liu Y, Han G, Li Y, Zhang Y, Xu H, Chen D. Novel nano-pomegranates based on astragalus polysaccharides for targeting ERα-positive breast cancer and multidrug resistance. Drug Deliv 2021; 27:607-621. [PMID: 32308054 PMCID: PMC7191906 DOI: 10.1080/10717544.2020.1754529] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Chemotherapy is an important method for treating breast cancer. However, multidrug resistance is one of the major challenges in breast cancer chemotherapy. There is an urgent need to develop novel, effective antitumor strategies that will perfect existing therapeutic regimens. In this study, the double-targeted nanocarrier, Quercetin-3'3-dithiodipropionic acid-Astragalus polysaccharides-Folic acid (QDAF), was successfully synthesized and self-assembled into a neoteric nano-targeted delivery strategy, named nano-pomegranates, and which were utilized to effectively inhibit multidrug resistance in estrogen receptor α (ERα)-positive breast tumor. The outstanding abilities of nano-pomegranates to release the drug in a reducing environment was determined by in vitro release assay. The cellular studies in MCF-7 cells were examined that nano-pomegranates have remarkable efficiencies of enhancing cellular uptake, inhibition and necrosis and apoptosis. In vivo antitumor experiments showed that nano-pomegranates have better anti-tumor effects and lower systemic toxicity than free Cur. In conclusion, nano-pomegranates have great potential in anti-breast cancer treatment.
Collapse
Affiliation(s)
- Bingjie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P.R. China
| | - Chunjing Guo
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P.R. China
| | - Yanhui Liu
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, P. R. China
| | - Guangting Han
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, P. R. China
| | - Yi Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P.R. China.,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, P. R. China
| | - Yanchun Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Daquan Chen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P.R. China.,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, P. R. China
| |
Collapse
|
29
|
Kurzątkowska K, Pazos MA, Herschkowitz JI, Hepel M. Cancer-Targeted Controlled Delivery of Chemotherapeutic Anthracycline Derivatives Using Apoferritin Nanocage Carriers. Int J Mol Sci 2021; 22:ijms22031362. [PMID: 33572999 PMCID: PMC7866407 DOI: 10.3390/ijms22031362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
The interactions of chemotherapeutic drugs with nanocage protein apoferritin (APO) are the key features in the effective encapsulation and release of highly toxic drugs in APO-based controlled drug delivery systems. The encapsulation enables mitigating the drugs' side effects, collateral damage to healthy cells, and adverse immune reactions. Herein, the interactions of anthracycline drugs with APO were studied to assess the effect of drug lipophilicity on their encapsulation excess n and in vitro activity. Anthracycline drugs, including doxorubicin (DOX), epirubicin (EPI), daunorubicin (DAU), and idarubicin (IDA), with lipophilicity P from 0.8 to 15, were investigated. We have found that in addition to hydrogen-bonded supramolecular ensemble formation with n = 24, there are two other competing contributions that enable increasing n under strong polar interactions (APO(DOX)) or under strong hydrophobic interactions (APO(IDA) of the highest efficacy). The encapsulation/release processes were investigated using UV-Vis, fluorescence, circular dichroism, and FTIR spectroscopies. The in vitro cytotoxicity/growth inhibition tests and flow cytometry corroborate high apoptotic activity of APO(drugs) against targeted MDA-MB-231 adenocarcinoma and HeLa cells, and low activity against healthy MCF10A cells, demonstrating targeting ability of nanodrugs. A model for molecular interactions between anthracyclines and APO nanocarriers was developed, and the relationships derived compared with experimental results.
Collapse
Affiliation(s)
- Katarzyna Kurzątkowska
- Department of Chemistry, State University of New York at Potsdam, Potsdam, NY 13676, USA
- Department of Biosensors, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland
- Correspondence: (K.K.); (M.H.); Tel.: +1-315-267-2267 (M.H.)
| | - Manuel A. Pazos
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Rensselaer, NY 12222, USA; (M.A.P.II); (J.I.H.)
| | - Jason I. Herschkowitz
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Rensselaer, NY 12222, USA; (M.A.P.II); (J.I.H.)
| | - Maria Hepel
- Department of Chemistry, State University of New York at Potsdam, Potsdam, NY 13676, USA
- Correspondence: (K.K.); (M.H.); Tel.: +1-315-267-2267 (M.H.)
| |
Collapse
|
30
|
Li XT, Jing M, Cai FY, Yao XM, Kong L, Wang XB. Enhanced antitumour efficiency of R 8GD-modified epirubicin plus tetrandrine liposomes in treatment of gastric cancer via inhibiting tumour metastasis. J Liposome Res 2020; 31:145-157. [PMID: 32223361 DOI: 10.1080/08982104.2020.1748647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Tumour metastasis is a major cause of cancer treatment failure and death, and chemotherapy efficiency for gastric cancer patients is usually unsatisfactory due to tumour cell metastasis, poor targeting and serious adverse reactions. In this study, a kind of R8GD-modified epirubicin plus tetrandrine liposomes was prepared to enhance the antitumor efficiency via killing tumour cells, destroying tumour metastasis and inhibiting energy supply for tumour cells. In order to investigate the antitumour efficiency of the targeting liposomes, morphology observation, intracellular uptake, cytotoxic effects, and inhibition on tumour metastasis and energy supply were carried out in vitro, and tumour-bearing mice models were established to investigate the antitumour efficiency in vivo. In vitro results showed that R8GD-modified epirubicin plus tetrandrine liposomes with ideal physicochemical properties could kill the most tumour cells, inhibit tumour metastasis and cut-off energy supply for tumour cells. In vivo results exhibited that R8GD-modified epirubicin plus tetrandrine liposomes could enhance the accumulation in tumour site and display an obvious antitumor efficiency. Therefore, R8GD-modified epirubicin plus tetrandrine liposomes could be used as a potential therapy for treatment of gastric cancer.
Collapse
Affiliation(s)
- Xue-Tao Li
- Department of Pharmacy, Chinese People's Liberation Army Logistics Support Force No. 967 Hospital, Dalian, China.,School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Ming Jing
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Fu-Yi Cai
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xue-Min Yao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xiao-Bo Wang
- Department of Pharmacy, Chinese People's Liberation Army Logistics Support Force No. 967 Hospital, Dalian, China
| |
Collapse
|
31
|
Alqaraghuli HGJ, Kashanian S, Rafipour R. A Review on Targeting Nanoparticles for Breast Cancer. Curr Pharm Biotechnol 2020; 20:1087-1107. [PMID: 31364513 DOI: 10.2174/1389201020666190731130001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 12/11/2022]
Abstract
Chemotherapeutic agents have been used extensively in breast cancer remedy. However, most anticancer drugs cannot differentiate between cancer cells and normal cells, leading to toxic side effects. Also, the resulted drug resistance during chemotherapy reduces treatment efficacy. The development of targeted drug delivery offers great promise in breast cancer treatment both in clinical applications and in pharmaceutical research. Conjugation of nanocarriers with targeting ligands is an effective therapeutic strategy to treat cancer diseases. In this review, we focus on active targeting methods for breast cancer cells through the use of chemical ligands such as antibodies, peptides, aptamers, vitamins, hormones, and carbohydrates. Also, this review covers all information related to these targeting ligands, such as their subtypes, advantages, disadvantages, chemical modification methods with nanoparticles and recent published studies (from 2015 to present). We have discussed 28 different targeting methods utilized for targeted drug delivery to breast cancer cells with different nanocarriers delivering anticancer drugs to the tumors. These different targeting methods give researchers in the field of drug delivery all the information and techniques they need to develop modern drug delivery systems.
Collapse
Affiliation(s)
- Hasanain Gomhor J Alqaraghuli
- Faculty of Chemistry, Razi University, Kermanshah, Iran.,Department of Sciences, College of Basic Education, Al- Muthanna University, Al-Muthanna, Iraq
| | - Soheila Kashanian
- Faculty of Chemistry, Sensor and Biosensor Research Center (SBRC) & Nanoscience and Nanotechnology Research Center (NNRC), Razi University, Kermanshah, Iran.,Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ronak Rafipour
- Department of Chemistry, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| |
Collapse
|
32
|
Yang W, Veroniaina H, Qi X, Chen P, Li F, Ke PC. Soft and Condensed Nanoparticles and Nanoformulations for Cancer Drug Delivery and Repurpose. ADVANCED THERAPEUTICS 2020; 3:1900102. [PMID: 34291146 PMCID: PMC8291088 DOI: 10.1002/adtp.201900102] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Indexed: 12/24/2022]
Abstract
Drug repurpose or reposition is recently recognized as a high-performance strategy for developing therapeutic agents for cancer treatment. This approach can significantly reduce the risk of failure, shorten R&D time, and minimize cost and regulatory obstacles. On the other hand, nanotechnology-based delivery systems are extensively investigated in cancer therapy due to their remarkable ability to overcome drug delivery challenges, enhance tumor specific targeting, and reduce toxic side effects. With increasing knowledge accumulated over the past decades, nanoparticle formulation and delivery have opened up a new avenue for repurposing drugs and demonstrated promising results in advanced cancer therapy. In this review, recent developments in nano-delivery and formulation systems based on soft (i.e., DNA nanocages, nanogels, and dendrimers) and condensed (i.e., noble metal nanoparticles and metal-organic frameworks) nanomaterials, as well as their theranostic applications in drug repurpose against cancer are summarized.
Collapse
Affiliation(s)
- Wen Yang
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | | | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | - Feng Li
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn AL 36849, USA
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| |
Collapse
|