1
|
Kumar K, Saini M, Rani V, Mishra M, Priya, Jatin, Tiwari S, Mishra B, Chawla R. Crafting a Molecular Trojan Horse: Sialic Acid-Modified PLGA Nanoparticles for Targeted Lung Cancer Therapy. Mol Pharm 2025; 22:1816-1830. [PMID: 40021483 DOI: 10.1021/acs.molpharmaceut.4c00957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
The glycan receptors prominently expressed on the surface of lung cancer cells offers promising targets for drug delivery. The prepared gemcitabine (GB)-loaded PLGA-NPs and sialic acid (Siac)-modified PLGA-NPs exhibited a uniform polydispersity index (PDI) value below 0.3, a particle size under 200 nm, and negative zeta potentials ranging from -17.45 to -21.45 mV. Entrapment efficiency (% EE) and drug loading values exceeded 70% and 8%, respectively. SEM and TEM showed that the particles were uniformly dispersed with a spherical shape. FTIR, XRD, TGA, and DSC analyses indicated the physiochemical stability of the drug within the nanoformulations. Controlled (26.92 to 31.64% within 24 h at pH 7.4) and pH-sensitive (36.80 to 40.25% within 24 h at pH 5.5) GB release were observed for the different formulations of PLGA-NPs. The MTT cytotoxicity assay revealed IC50 values for the GB control, GB-PLGA-NPs, and GB-PLGA-Siac-NPs as 13.65 ± 1.20, 8.14 ± 1.24, and 4.16 ± 1.05 μg/mL, respectively. The Co6-GB-PLGA-Siac-NPs showed significantly higher cellular uptake than that of the Co6-GB control (p < 0.001) and Co6-GB-PLGA-NPs (p < 0.01) respectively. Pharmacokinetic profiles indicated higher AUC values (ng·h/mL) for GB-PLGA-Siac-NPs (8355.07 ± 2006.45) compared to GB-PLGA-NPs (6145.58 ± 969.25) and the GB control (1510.72 ± 81.08), resulting in higher bioavailability of GB-PLGA-Siac-NPs. Biodistribution studies confirmed superior localization of DiD-GB-PLGA-Siac-NPs, indicated by radiant efficiency signal on B[a]P induced lung cancerous tissues relative to DiD-GB-PLGA-NPs after 1 h (p < 0.001), 4 h (p < 0.01), and 12 h (p < 0.001), which could be attributed to their ability to target glycans. In vivo anticancer efficacy in a B[a]P-induced lung cancer mice model depicted that GB-PLGA-Siac-NPs effectively inhibited lung cancer cells and reduced systemic toxicity, as evidenced by the average number of lung cancer cells, body weight values, survival analysis, biochemical parameters associated with organs (such as the liver and kidney), and histopathological analysis. Therefore, GB-loaded Siac-coated PLGA nanoparticles could serve as an efficient vehicle for GB delivery via targeting glycan receptors in lung cancer therapy.
Collapse
Affiliation(s)
- Krishan Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| | - Manjit Saini
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Varsha Rani
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
- Jichi Medical University Shimostuke, Shimotsuke, Tochigi Pefecture 329-0431, Japan
| | - Mohini Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Priya
- Department of Pharmacy, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Jatin
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Siddharth Tiwari
- School of Pharmaceutical Sciences, CSJM University, Kanpur, Uttar Pradesh 208024, India
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| | - Ruchi Chawla
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
2
|
Aghanejad A, Kheiriabad S, Ghaffari M, Namvar Aghdash S, Ghafouri N, Ezzati Nazhad Dolatabadi J, Andishmand H, Hamblin MR. Targeted co-delivery nanosystem based on methotrexate, curcumin, and PAMAM dendrimer for improvement of the therapeutic efficacy in cervical cancer. Sci Rep 2025; 15:1813. [PMID: 39805840 PMCID: PMC11730290 DOI: 10.1038/s41598-024-82074-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
The simultaneous administration of multiple drugs within identical nanocarriers to cancer cells or tissues can result in the effective action of drugs at reduced concentrations. In this investigation, PAMAM dendrimers (G4-PAMAM) were employed to link with methotrexate (MTX) using DCC/NHS chemistry and followed by the entrapment of curcumin (Cur) within it. The establishment of covalent bonds between MTX and the PAMAM dendrimer led to PAMAM-MTX interaction, verified and described through FT-IR. Various techniques were employed to evaluate the structural properties of the prepared Cur-PAMAM-MTX NC. The Cur-PAMAM-MTX NC, after preparation, exhibited a particle size of 249 nm, with an encapsulation efficiency (EE) of ~ 81% for Cur. The cumulative in vitro release of Cur-loaded NC indicated a controlled release influenced by time and pH. The cell study results revealed that Cur-PAMAM-MTX NC exhibited significantly higher cytotoxicity than free MTX, Cur, and other formulations tested in vitro. The synergistic effect of co-delivery of MTX and Cur by PAMAM significantly increased cytotoxicity. Besides, the significant ROS level rising has been shown in the treated cells with MTX-PAMAM-Cur. Considering these findings, the co-delivery NC shows promise for additional in vitro investigations and possesses the capacity to function as an effective framework for the combined delivery of MTX and Cur in cervical cancer chemotherapy.
Collapse
Affiliation(s)
- Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Kheiriabad
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Maryam Ghaffari
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Namvar Aghdash
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Neda Ghafouri
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hashem Andishmand
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Department of Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| |
Collapse
|
3
|
Babar M, Aslam B, Faisal MN, Malik A, Akhtar S, Fatima S, Majeed W, Umer A, Farooq MA. Phytochemical characterization and anti-arthritic potential of green-synthesized CuO nanoparticles derived from the Bistorta amplexicaulis root extract. Front Pharmacol 2024; 15:1474592. [PMID: 39741627 PMCID: PMC11685013 DOI: 10.3389/fphar.2024.1474592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/22/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Rheumatoid arthritis is an autoimmune disease that mainly causes joint damage. The patient experiences loss of appetite, pain, fever, and fatigue. The present study was designed to phytochemically characterize and evaluate the anti-arthritic activity of green-synthesized copper oxide (CuO) nanoparticles (NPs) using the hydroalcoholic extract of Bistorta amplexicaulis roots in an adjuvant-induced arthritic rat model. Material and Methods For this purpose, crude powdered plant material was used for proximate analysis, and the plant extract was assessed for qualitative phytochemical analysis, mineral contents, and flavonoid and phenolic contents, as well as quantitative phytochemical analysis through reversed-phase high-performance liquid chromatography (RP-HPLC) and Fourier-transform infrared (FTIR) spectroscopy. The in vitro antioxidant activity of both extracts was determined by the 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay. The biosynthesized CuO NPs from the Bistorta amplexicaulis extract showed anti-arthritic activity due to the presence of flavonoids and phenols, which showed a pain reliever effect by blocking the cyclo-oxygenase enzyme and has immune suppressant activity, thus securing the joint from destruction. The nanoparticles were characterized by zeta size, zeta potential, scanning electron microscopy (SEM), and FTIR spectroscopy. Forty-eight albino rats were divided randomly into six treatment groups. Results and Disscussion The zeta size and zeta potential of the nanoparticles were 186.8 nm and -9.23 mV, respectively. Joint stiffness, spleen weight, thymus weight, and paw thickness showed a significant decrease after treatment with NPs. The hematological parameters such as red blood cells (RBCs) and hemoglobin showed a significant increase, while platelets and white blood cells (WBCs) showed a significant decrease in NP-treated groups. C-reactive protein (CRP), rheumatoid factor (RF), liver and kidney function biomarkers, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) levels showed a significant decrease at both low and high doses of green-synthesized CuO nanoparticles from the Bistorta amplexicaulis root extract. The final data were analyzed by one way and two-way analysis of variance (ANOVA) and Tukey's multi-comparison test. Conclusion So, from this study, it was concluded that both the plant root extract and green-synthesized CuO nanoparticles have anti-arthritic potential, but CuO NPs showed remarkable results.
Collapse
Affiliation(s)
- Mahrukh Babar
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Bilal Aslam
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Naeem Faisal
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Suhail Akhtar
- Department of Biochemistry, A.T. Still University of Health Sciences, Kirksville, MO, United States
| | - Sabiha Fatima
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Wafa Majeed
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Asher Umer
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
4
|
Madani F, Morovvati H, Webster TJ, Najaf Asaadi S, Rezayat SM, Hadjighassem M, Khosravani M, Adabi M. Combination chemotherapy via poloxamer 188 surface-modified PLGA nanoparticles that traverse the blood-brain-barrier in a glioblastoma model. Sci Rep 2024; 14:19516. [PMID: 39174603 PMCID: PMC11341868 DOI: 10.1038/s41598-024-69888-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024] Open
Abstract
The effect of chemotherapy for anti-glioblastoma is limited due to insufficient drug delivery across the blood-brain-barrier. Poloxamer 188-coated nanoparticles can enhance the delivery of nanoparticles across the blood-brain-barrier. This study presents the design, preparation, and evaluation of a combination of PLGA nanoparticles (PLGA NPs) loaded with methotrexate (P-MTX NPs) and PLGA nanoparticles loaded with paclitaxel (P-PTX NPs), both of which were surface-modified with poloxamer188. Cranial tumors were induced by implanting C6 cells in a rat model and MRI demonstrated that the tumors were indistinguishable in the two rats with P-MTX NPs + P-PTX NPs treated groups. Brain PET scans exhibited a decreased brain-to-background ratio which could be attributed to the diminished metabolic tumor volume. The expression of Ki-67 as a poor prognosis factor, was significantly lower in P-MTX NPs + P-PTX NPs compared to the control. Furthermore, the biodistribution of PLGA NPs was determined by carbon quantum dots loaded into PLGA NPs (P-CQD NPs), and quantitative analysis of ex-vivo imaging of the dissected organs demonstrated that 17.2 ± 0.6% of the NPs were concentrated in the brain after 48 h. The findings highlight the efficacy of combination nanochemotherapy in glioblastoma treatment, indicating the need for further preclinical studies.
Collapse
Affiliation(s)
- Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Morovvati
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Thomas J Webster
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, China
- Program in Materials Science, UFPI, Teresina, Brazil
| | - Sareh Najaf Asaadi
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Yekeler HB, Guler E, Beato PS, Priya S, Abobakr FKM, Dogan M, Uner B, Kalaskar DM, Cam ME. Design and in vitro evaluation of curcumin-loaded PLGA nanoparticle-embedded sodium alginate/gelatin 3D printed scaffolds for Alzheimer's disease. Int J Biol Macromol 2024; 268:131841. [DOI: 6.https:/doi.org/10.1016/j.ijbiomac.2024.131841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2025]
|
6
|
Yekeler HB, Guler E, Beato PS, Priya S, Abobakr FKM, Dogan M, Uner B, Kalaskar DM, Cam ME. Design and in vitro evaluation of curcumin-loaded PLGA nanoparticle-embedded sodium alginate/gelatin 3D printed scaffolds for Alzheimer's disease. Int J Biol Macromol 2024; 268:131841. [PMID: 38679260 DOI: 10.1016/j.ijbiomac.2024.131841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Targeted nanoparticles (NPs) are aimed at improving clinical outcomes by enhancing the diagnostic and therapeutic efficacy of drugs in the treatment of Alzheimer's disease (AD). METHODS Curcumin (CUR)-loaded poly-lactic-co-glycolic acid (PLGA) NPs (CNPs) were produced to demonstrate a prolonged release and successfully embedded into 3D printed sodium alginate (SA)/gelatin (GEL) scaffolds that can dissolve rapidly sublingually. Characterization and in vitro activity of the NPs and scaffolds were evaluated. RESULTS Based on the in vitro drug release studies, 99.6 % of the encapsulated CUR was released in a controlled manner within 18 days for the CNPs. In vitro cell culture studies showed that all samples exhibited cell viability above 84.2 % and no significant cytotoxic effect on SH-SY5Y cells. The samples were analyzed through 2 different pathways by PCR analysis. Real-time PCR results indicated that CNP and CNP-embedded SA/GEL scaffolds (CNPSGS) may show neuroprotective effects by modulating the Wnt/β-catenin pathway. The gene expression level of β-catenin slightly increased compared to the gene expression levels of other proteins and enzymes with these treatments. However, the PI3K/Akt/GSK-3β signaling pathway was regulated at the same time because of the crosstalk between these 2 pathways. CONCLUSION CNPSGS might be an effective therapeutic alternative for AD treatment.
Collapse
Affiliation(s)
- Humeyra Betul Yekeler
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Istanbul 34854, Türkiye; Center for Nanotechnology and Biomaterials Application and Research, Marmara University, Istanbul 34722, Türkiye; UCL Division of Surgery and Interventional Science, Royal Free Hospital Campus, University College London, Rowland Hill Street, NW3 2PF, UK; MecNano Technologies, Cube Incibation, Teknopark Istanbul, Istanbul 34906, Türkiye
| | - Ece Guler
- Center for Nanotechnology and Biomaterials Application and Research, Marmara University, Istanbul 34722, Türkiye; UCL Division of Surgery and Interventional Science, Royal Free Hospital Campus, University College London, Rowland Hill Street, NW3 2PF, UK; MecNano Technologies, Cube Incibation, Teknopark Istanbul, Istanbul 34906, Türkiye; Department of Pharmacology, Faculty of Pharmacy, Istanbul Kent University, Kagithane 34406, Istanbul, Türkiye
| | - Patricia Santos Beato
- UCL Division of Surgery and Interventional Science, Royal Free Hospital Campus, University College London, Rowland Hill Street, NW3 2PF, UK
| | - Sushma Priya
- UCL Division of Surgery and Interventional Science, Royal Free Hospital Campus, University College London, Rowland Hill Street, NW3 2PF, UK
| | | | - Murat Dogan
- Department of Pharmaceutical Biotechnology, Cumhuriyet University, Sivas 58140, Türkiye; Cancer Survivorship Institute, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 625 N. Michigan Ave., Suite 2100, Chicago, IL, 60611, USA
| | - Burcu Uner
- Department of Pharmaceutical and Administrative Sciences, University of Health Science and Pharmacy in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul Kent University, Kagithane 34406, Istanbul, Türkiye
| | - Deepak M Kalaskar
- UCL Division of Surgery and Interventional Science, Royal Free Hospital Campus, University College London, Rowland Hill Street, NW3 2PF, UK
| | - Muhammet Emin Cam
- Center for Nanotechnology and Biomaterials Application and Research, Marmara University, Istanbul 34722, Türkiye; UCL Division of Surgery and Interventional Science, Royal Free Hospital Campus, University College London, Rowland Hill Street, NW3 2PF, UK; MecNano Technologies, Cube Incibation, Teknopark Istanbul, Istanbul 34906, Türkiye; Department of Pharmacology, Faculty of Pharmacy, Istanbul Kent University, Kagithane 34406, Istanbul, Türkiye; Biomedical Engineering Department, University of Aveiro, Aveiro 3810-193, Portugal; Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, Istanbul 34854, Türkiye.
| |
Collapse
|
7
|
Malekpour MR, Hosseindoost S, Madani F, Kamali M, Khosravani M, Adabi M. Combination nanochemotherapy of brain tumor using polymeric nanoparticles loaded with doxorubicin and paclitaxel: An in vitro and in vivo study. Eur J Pharm Biopharm 2023; 193:175-186. [PMID: 37926270 DOI: 10.1016/j.ejpb.2023.11.002] [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: 05/20/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023]
Abstract
This study aims to overcome physiological barriers and increase the therapeutic index for the treatment of glioblastoma (GBM) tumors by using Paclitaxel (PTX) loaded Poly(lactic co-glycolic acid) nanoparticles (PTX-PLGA-NPs) and Doxorubicin (DOX) loaded Poly (lactic co-glycolic acid) nanoparticles (DOX-PLGA-NPs). The hydrodynamic diameter of nanoparticles (NPs) was characterized by dynamic light scattering (DLS) which was 94 ± 4 nm and 133 ± 6 nm for DOX-PLGA-NPs, and PTX-PLGA-NPs, respectively. The zeta potential for DOX-PLGA-NPs and PTX-PLGA-NPs were -15.2 ± 0.18 mV and -17.3 ± 0.34 mV, respectively. The cytotoxicity of PTX-PLGA-NPs and DOX-PLGA-NPs was augmented compared to DOX and PTX on C6 GBM cells. The Lactate dehydrogenase (LDH) tests for various formulations were carried out. The results indicated that the amount of released LDH was 262 ± 7.84 U.L-1 at the concentration of 2 mg/mL in the combination therapy, which was much higher than other groups (DOX-PLGA-NPs (210 ± 6.92 U.L-1), PTX-PLGA-NPs (201 ± 8.65 U.L-1), DOX (110 ± 9.81 U.L-1), PTX (95 ± 5.02 U.L-1) and PTX + DOX (67 ± 4.89 U.L-1)). MRI results of the combination therapy of PTX-PLGA-NPs and DOX-PLGA-NPs indicated that GBM tumor size decreased considerably compared to the other formulations. Also, combination therapy of PTX-PLGA-NPs and DOX-PLGA-NPs demonstrated a longer median survival of more than 80 days compared to PTX (38 days), DOX (37 days) and PTX + DOX (48 days), PTX-NPs (58 days) and DOX-NPs (62 days). The results of locomotion, body weight, rearing and grooming assays indicated that combination therapy of PTX-PLGA-NPs and DOX-PLGA-NPs had the most positive effect on the movements of rats compared to the other formulations.
Collapse
Affiliation(s)
- Mohammad Reza Malekpour
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saereh Hosseindoost
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Kamali
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Yu F, Wei Z, Chen J, Long Y, Qing Q, Li B, Zhang X, Chen H, Lan T, Zhu P, Shen P, Zeng W, Lin J, Qi Z, Hong X, Chen XD. Preparation of curcumin-loaded MPEG-PTMC nanoparticles: Physicochemical properties, antioxidant activity, and in vivo pharmacokinetic behavior. POWDER TECHNOL 2023. [DOI: 10.1016/j.powtec.2023.118492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
9
|
Hu H, Liao Z, Xu M, Wan S, Wu Y, Zou W, Wu J, Fan Q. Fabrication, Optimization, and Evaluation of Paclitaxel and Curcumin Coloaded PLGA Nanoparticles for Improved Antitumor Activity. ACS OMEGA 2023; 8:976-986. [PMID: 36643566 PMCID: PMC9835547 DOI: 10.1021/acsomega.2c06359] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
Codelivery of chemotherapeutic drugs in nanoparticles can enhance the therapeutic effects against tumors. However, their anticancer properties and physiochemical characteristics can be severely influenced by many formulation parameters during the preparation process. It is a complicated development phase to select the optimal parameters for preparation of nanoparticles based on the commonly used one single parameter method, which consumes a lot of money, time, and effort, and sometimes even fails. Therefore, the statistical analysis based on Box-Behnken design (BBD) has attracted much attention in bioengineering fields because it can illustrate the influence of parameters, build mathematical models, and predict the optimal combinational factors in a decreased number of experiments. In this study, we used a three-factor three-level BBD design to optimize the preparation of poly(lactic-co-glycolic acid) (PLGA) nanoparticles coloaded with two anticancer drugs curcumin and paclitaxel (PLGA-CUR-PTX nanoparticles). The surfactant concentration, polymer concentration, and oil-water ratio were selected as independent variables. An optimized model of the formulation for PLGA-CUR-PTX nanoparticles was validated. The optimal nanoparticles possessed a uniform spherical shape, with an average size of 99.94 nm, and the drug encapsulation efficiencies of CUR and PTX were 63.53 and 80.64%, respectively. The drug release from nanoparticles showed a biphasic release behavior, with a release mechanism via diffusion and fundamentally quasi-Fickian diffusion. The optimized nanoparticles demonstrated an enhanced cytotoxicity effect with lower IC50 values to 4T1 and MCF-7 breast cancer cell lines compared to free drugs. In summary, BBD optimization of CUR and PTX coloaded nanoparticles yielded a favorable drug carrier that holds potential as an alternative treatment for anticancer therapy.
Collapse
Affiliation(s)
- Haiyang Hu
- Department
of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan610075, China
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
| | - Zuyue Liao
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
| | - Mengyao Xu
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
| | - Shengli Wan
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
| | - Yuesong Wu
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
| | - Wenjun Zou
- Department
of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan610075, China
| | - Jianming Wu
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
- School
of Basic Medical Sciences, Southwest Medical
University, Luzhou646000, China
| | - Qingze Fan
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
| |
Collapse
|
10
|
Recent Trends in Nanomedicine-Based Strategies to Overcome Multidrug Resistance in Tumors. Cancers (Basel) 2022; 14:cancers14174123. [PMID: 36077660 PMCID: PMC9454760 DOI: 10.3390/cancers14174123] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is the leading cause of economic and health burden worldwide. The commonly used approaches for the treatment of cancer are chemotherapy, radiotherapy, and surgery. Chemotherapy frequently results in undesirable side effects, and cancer cells may develop resistance. Combating drug resistance is a challenging task in cancer treatment. Drug resistance may be intrinsic or acquired and can be due to genetic factors, growth factors, the increased efflux of drugs, DNA repair, and the metabolism of xenobiotics. The strategies used to combat drug resistance include the nanomedicine-based targeted delivery of drugs and genes using different nanocarriers such as gold nanoparticles, peptide-modified nanoparticles, as well as biomimetic and responsive nanoparticles that help to deliver payload at targeted tumor sites and overcome resistance. Gene therapy in combination with chemotherapy aids in this respect. siRNA and miRNA alone or in combination with chemotherapy improve therapeutic response in tumor cells. Some natural substances, such as curcumin, quercetin, tocotrienol, parthenolide, naringin, and cyclosporin-A are also helpful in combating the drug resistance of cancer cells. This manuscript summarizes the mechanism of drug resistance and nanoparticle-based strategies used to combat it.
Collapse
|
11
|
Mohamadian M, Ahmadi SS, Bahrami A, Ferns GA. Review on the Therapeutic Potential of Curcumin and its Derivatives on Glioma Biology. Neurochem Res 2022; 47:2936-2953. [PMID: 35790698 DOI: 10.1007/s11064-022-03666-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/15/2022] [Accepted: 06/18/2022] [Indexed: 11/24/2022]
Abstract
Gliomas are common and aggressive brain tumors that carry a poor prognosis. The current multimodal therapeutic option for glioma includes surgery subsequently temozolomide chemotherapy and/or radiation; but gliomas are often associated with multidrug resistance, intensive adverse events, and tumor relapse. Thus, novel interventions that can enhance successful chemo-prevention and overcome therapeutic resistance are urgently needed. Phytochemicals have several biological properties with multi-target sites and relatively limited degrees of toxicity. Curcumin is a natural polyphenolic compound with several anti-tumor effects which potentially inhibit tumor growth, development, proliferation, invasion, dissemination, and angiogenesis in different human malignancies. Experimental model studies have demonstrated that curcumin attenuates glioma cell viability by G2/M cell cycle arrest, apoptosis, induction of autophagy, gene expression alteration, and disruption of multi-molecular pathways. Moreover, curcumin has been reported to re-sensitize cancer to chemotherapeutics as well as augment the effect of radiotherapy on glioma cells. In this review, we have provided an update on the in vitro and in vivo effects of curcumin-based therapy on gliomas. We have also discussed the use of curcumin in combination therapies, its effectiveness on drug-resistant cells, and new formulations of curcumin in the treatment of gliomas.
Collapse
Affiliation(s)
- Malihe Mohamadian
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsane Bahrami
- Clinical Research Development Unit, Faculty of Medicine, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran. .,Clinical Research Development Unit of Akbar Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Medical Education, Falmer, Brighton, BN1 9PH, Sussex, UK
| |
Collapse
|
12
|
Hasan Khudhair D, Al-Gareeb AI, Al-kuraishy HM, El-Kadem AH, Elekhnawy E, Negm WA, Saber S, Cavalu S, Tirla A, Alotaibi SS, Batiha GES. Combination of Vitamin C and Curcumin Safeguards Against Methotrexate-Induced Acute Liver Injury in Mice by Synergistic Antioxidant Effects. Front Med (Lausanne) 2022; 9:866343. [PMID: 35492324 PMCID: PMC9047671 DOI: 10.3389/fmed.2022.866343] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
Methotrexate (MTX), an antineoplastic and immunosuppressive drug, widely used in the treatment of different types of cancers and the management of chronic inflammatory diseases. However, its use is associated with hepatotoxicity. Vitamin C (VC) and curcumin (CUR) exhibit anti-inflammatory and antioxidant effects. Thus, we aimed to investigate the potential hepatoprotective effects of VC and CUR pretreatment alone and in combination against MTX-induced hepatotoxicity. Albino mice were randomly divided into 7 groups: the control group, which received only normal saline; MTX group; VC group, pretreated with VC (100 or 200 mg/kg/day orally) for 10 days; CUR group, pretreated with CUR (10 or 20 mg/kg/day orally); and combination group, which received VC (100 mg/kg) and CUR (10 mg/kg). MTX was administered (20 mg/kg, intraperitoneally) to all the groups on the tenth day to induce hepatotoxicity. Forty eight hours after MTX administration, the mice were anesthetized. Blood samples were collected, the liver was removed for biochemical analysis, and a part of the tissue was preserved in formalin for histopathological analysis. The results indicated that pretreatment with a combination of VC and CUR induced a more significant decrease in the serum levels of alanine transaminase, aspartate transaminase, alkaline phosphatase, and lactic dehydrogenase and a significant increase in the tissue level of superoxide dismutase and glutathione; furthermore, it induced a significant decrease in malondialdehyde levels and improvement in histopathological changes in the liver tissues, confirming the potential hepatoprotective effects of the combination therapy on MTX-induced liver injury. To conclude, MTX-induced hepatotoxicity is mediated by induction of oxidative stress as evident by increased lipid peroxidation and reduction of antioxidant enzyme activity. Pretreatment with VC, CUR or their combination reduces the MTX-induced hepatotoxicity by antioxidant and anti-inflammatory effects. However, the combined effect of VC and CUR provided a synergistic hepatoprotective effect that surpasses pretreatment with CUR alone but seems to be similar to that of VC 200 mg/kg/day. Therefore, VC and CUR combination or a large dose of VC could be effective against MTX-induced hepatotoxicity. In this regard, further studies are warranted to confirm the combined hepatoprotective effect of VC and CUR against MTX-induced hepatotoxicity.
Collapse
Affiliation(s)
- Dhekra Hasan Khudhair
- Department of Clinical Pharmacology and Medicine, College of Medicine, University of Al-Mustansiriyah, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, University of Al-Mustansiriyah, Baghdad, Iraq
| | - Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, University of Al-Mustansiriyah, Baghdad, Iraq
| | - Aya H. El-Kadem
- Department of Pharmacology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Walaa A. Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Adrian Tirla
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Saqer S. Alotaibi
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
13
|
Mousazadeh N, Gharbavi M, Rashidzadeh H, Nosrati H, Danafar H, Johari B. Anticancer evaluation of methotrexate and curcumin coencapsulated niosomes against colorectal cancer cell line. Nanomedicine (Lond) 2022; 17:201-217. [PMID: 35037483 DOI: 10.2217/nnm-2021-0334] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: The aim of the present investigation was to develop niosomes containing both curcumin (CUR) and methotrexate (MTX). Also, the combinational effect of CUR and MTX in both free and niosomal forms on growth inhibition potential and induction of apoptosis in the HCT-116 cell line were exploited. Materials & methods: Niosomes were prepared by the thin-film hydration method and their physicochemical properties were determined by various techniques. Cellular uptake, cell apoptosis, wound healing and MTT assay were conducted to ascertain niosomes' feasibility for cancer therapy. Results: The combination of CUR and MTX in niosomal formulation showed more toxicity than their combination in free form. Conclusion: The nanocarrier-based approach was effective for the codelivery of CUR and MTX against cancer cells in vitro.
Collapse
Affiliation(s)
- Navid Mousazadeh
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahmoud Gharbavi
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamid Rashidzadeh
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamed Nosrati
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Joint Ukraine-Azerbaijan International Research & Education Center of Nanobiotechnology & Functional Nanosystems, Drohobych, Ukraine, Baku, Azerbaijan
| | - Hossein Danafar
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Behrooz Johari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
14
|
Jenifer J, Upputuri RTP. In vitro release mechanism and cytotoxic behavior of curcumin loaded casein nanoparticles. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
15
|
Maleki H, Hosseini Najafabadi MR, Webster TJ, Hadjighassem MR, Sadroddiny E, Ghanbari H, Khosravani M, Adabi M. Effect of Paclitaxel/etoposide co-loaded polymeric nanoparticles on tumor size and survival rate in a rat model of glioblastoma. Int J Pharm 2021; 604:120722. [PMID: 34022255 DOI: 10.1016/j.ijpharm.2021.120722] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/18/2022]
Abstract
The aim of this work is to co-load paclitaxel (PTX) and etoposide (ETP) in methoxy poly(ethylene glycol)-poly(lactic-co-glycolic acid) nanoparticles (mPEG-PLGA NPs) to overcome pharmacokinetics and physiological limitations and enhance therapeutic efficacy for treating intracranial glioblastoma. Both drugs were loaded into mPEG-PLGA NPs by a nano-precipitation method. The resultant NPs demonstrated an enhanced cytotoxic effect indicated by lower IC50 values and augmented cell apoptosis to U87 and C6 glioma cell lines compared to both free drugs. Additionally, blood compatibility assays showed that the PTX/ETP co-loaded mPEG-PLGA NPs did not induce blood hemolysis, blood clotting, or platelet aggregation. In vivo anti-glioma efficacy evaluation in rats bearingintracranialC6glioma revealed a superior anti-glioma activity for the treatment with PTX/ETP co-loaded mPEG-PLGA NPs compared to other formulations, particularly a significantly longer median survival, 76 days compared to 36 days for free PTX and 37 days for free ETP treatment, respectively, and higher tumor regression, proved by magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Hassan Maleki
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Hosseini Najafabadi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Thomas J Webster
- Chemical Engineering Department, Northeastern University, Boston, MA 02115, USA
| | - Mahmoud Reza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Effect of Curcumin-Hydroxypropyl-$$\beta$$-Cyclodextrin Complex and the Complex Loaded Gelatin Carrageenan Microparticles on the Various Chemical and Biological Properties. J Pharm Innov 2021. [DOI: 10.1007/s12247-021-09559-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
17
|
Kour G, Haq SA, Bajaj BK, Gupta PN, Ahmed Z. Phytochemical add-on therapy to DMARDs therapy in rheumatoid arthritis: In vitro and in vivo bases, clinical evidence and future trends. Pharmacol Res 2021; 169:105618. [PMID: 33878447 DOI: 10.1016/j.phrs.2021.105618] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/25/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
The use of biologically active compounds derived from plants i.e. phytochemicals, have been known for ages for their pharmacological activities in the treatment of autoimmune disorders like rheumatoid arthritis (RA). Besides enormous scientific evidence, the therapeutic potential of phytochemicals is often undervalued. The treatment in RA involves the use of synthetic and biological disease modifying anti-rheumatic drugs (DMARDs). However, the long-term treatment in RA is associated with the risk of gastrointestinal, liver, pulmonary and renal toxicities and serious infections including latent tuberculosis, pneumococcus influenza, herpes zoster and hepatitis. These adverse effects sometimes lead to discontinuation of the therapy. A relatively new vision based on the combination of DMARDs with phytochemicals exhibiting anti-inflammatory, anti-arthritic, anti-oxidant, hepatoprotective and nephroprotective properties for the treatment of RA has achieved substantial importance in the last decade. From this perspective, the present review focuses on the combination of DMARDs (primarily MTX) with phytochemicals that have shown synergistic therapeutic effects while decreasing the toxic repercussions of current RA therapy. The review covers recent evidences of such combination studies that have shown promising results both in experimental arthritic models and clinical arthritis. Few of the combinations including resveratrol, sinomenine, coenzyme Q10 exhibited considerable interest because of their efficacy as an adjuvant to the MTX/standard DMARDs therapy in clinical trials. Besides giving an overview of such combination studies the review also critically discusses the limitations with the use of phytochemicals (e.g. solubility, permeability and bioavailability) compromising their clinical application. Additionally, it stresses upon the need of novel delivery systems and pharmaceutical technologies to increase the therapeutic efficacy of the combination therapy. Overall, the review unveils the potential of phytochemicals in combination with DMARDs with increased tolerability and superior efficacy in further refining the future of the RA therapy.
Collapse
Affiliation(s)
- Gurleen Kour
- Inflammation Pharmacology Division, CSIR, Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, J&K, India; School of Biotechnology, University of Jammu, Baba Saheb Ambedkar Road, Jammu Tawi, 180006 J&K, India
| | - Syed Assim Haq
- Formulation & Drug Delivery Division, CSIR, Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, J&K, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bijender Kumar Bajaj
- School of Biotechnology, University of Jammu, Baba Saheb Ambedkar Road, Jammu Tawi, 180006 J&K, India
| | - Prem N Gupta
- Formulation & Drug Delivery Division, CSIR, Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, J&K, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Zabeer Ahmed
- Inflammation Pharmacology Division, CSIR, Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, J&K, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
18
|
McCord E, Pawar S, Koneru T, Tatiparti K, Sau S, Iyer AK. Folate Receptors' Expression in Gliomas May Possess Potential Nanoparticle-Based Drug Delivery Opportunities. ACS OMEGA 2021; 6:4111-4118. [PMID: 33623837 PMCID: PMC7893640 DOI: 10.1021/acsomega.0c05500] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/22/2021] [Indexed: 05/05/2023]
Abstract
Brain cancer effected around estimated 23 890 adults and 3540 children under the age of 15 in 2020. The chemotherapeutic agents that are already approved by the FDA for brain cancer are proving to be not highly effective because of the interference from the tumor microenvironment as well as their own toxicities. Added to this is the impedance presented by the extremely restrictive permeability of the blood brain barrier (BBB). Targeted nanoparticulate drug delivery systems offer a good opportunity to traverse the BBB and selectively target the tumor cells. Folate receptors are found to be one of the most useful targets for drug delivery to the brain. Hence, this Mini-Review discusses the folate receptors and their application in the treatment of brain cancers using targeted nanoparticles.
Collapse
Affiliation(s)
- Eva McCord
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
- Grosse
Pointe South High School, Grosse
Pointe Farms, Michigan 48236, United States
| | - Shreya Pawar
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
- Troy
High School, Troy, Michigan 48098, United States
| | - Tejaswi Koneru
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
- Walled
Lake Central High School, Commerce Charter Township, Walled Lake, Michigan 48390, United States
| | - Katyayani Tatiparti
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
| | - Samaresh Sau
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
| | - Arun K. Iyer
- Use-inspired
Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory,
Department of Pharmaceutical Sciences, Eugene Applebaum College of
Pharmacy and Health Sciences, Wayne State
University, Detroit, Michigan 48201, United States
- Molecular
Imaging Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| |
Collapse
|
19
|
Esnaashari SS, Muhammadnejad S, Amanpour S, Amani A. A Combinational Approach Towards Treatment of Breast Cancer: an Analysis of Noscapine-Loaded Polymeric Nanoparticles and Doxorubicin. AAPS PharmSciTech 2020; 21:166. [PMID: 32504144 DOI: 10.1208/s12249-020-01710-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 05/13/2020] [Indexed: 01/10/2023] Open
Abstract
Our aim in this study was to clarify the combination anticancer effect of Noscapine (Nos) loaded in a polymeric nanocarrier with Doxorubicin (Dox) on breast cancer cells. Nanoprecipitation method was used to prepare methoxy polyethylene glycol (mPEG), poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) containing Nos. Transmission electron microscopy (TEM) and dynamic light scattering (DLS) were used to characterize the prepared Nos NPs. The anticancer activity of Nos NPs alone and in combination with Dox was assessed on 4T1 breast cancer cell line and in mice model. Spherical-shaped Nos NPs were prepared, with size of 101 ± 4.80 nm and zeta potential of - 15.40 ± 1 mV. Fourier transform infrared (FTIR) spectroscopy results demonstrated that Nos chemical structure was kept stable during preparation process. However, differential scanning calorimetric (DSC) thermogram proved that crystalline state of Nos changed to amorphous state in Nos NPs. The entrapment efficacy % (EE%) and drug loading % (DL%) of Nos NPs were about 87.20 ± 3.50% and 12.50 ± 2.30%, respectively. Synergistic anticancer effects of Nos both in free form (in hydrochloride form, Nos HCl) and Nos NPs form with Dox hydrochloride (Dox HCl) were observed on 4T1 cells. Combination of Nos NPs and Dox HCl inhibited tumor growth (68.50%) in mice more efficiently than Nos NPs (55.10%) and Dox HCl (32%) alone. Immunohistochemical (IHC) analysis of the tumor tissues confirmed antiangiogenic effect of Nos NPs. The findings highlighted efficacy of Nos NPs alone and in combination with Dox HCl on breast cancer tumors.
Collapse
|
20
|
Maleki H, Naghibzadeh M, Amani A, Adabi M, Khosravani M. Preparation of Paclitaxel and Etoposide Co-loaded mPEG-PLGA Nanoparticles: an Investigation with Artificial Neural Network. J Pharm Innov 2019. [DOI: 10.1007/s12247-019-09419-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|