1
|
Arab HH, Eid AH, Alsufyani SE, Ashour AM, Alnefaie AM, Alsharif NM, Alshehri AM, Almalawi AA, Alsowat AA, Abd El Aal HA, Hassan ESG, Elesawy WH, Elhemiely AA. Activation of AMPK/mTOR-Driven Autophagy and Suppression of the HMGB1/TLR4 Pathway with Pentoxifylline Attenuates Doxorubicin-Induced Hepatic Injury in Rats. Pharmaceuticals (Basel) 2024; 17:681. [PMID: 38931349 PMCID: PMC11206793 DOI: 10.3390/ph17060681] [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: 04/14/2024] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Despite being an effective chemotherapeutic agent, the clinical use of doxorubicin (DOX) is limited by several organ toxicities including hepatic injury. Pentoxifylline (PTX) is a methylxanthine derivative with marked anti-inflammatory and anti-apoptotic features. It is unknown, however, whether PTX can mitigate DOX-evoked hepatotoxicity. This study aims to explore the potential hepatoprotective impact of PTX in DOX-induced hepatic injury and the underlying molecular mechanisms. Histopathology, immunohistochemistry, and ELISA were used to examine liver tissues. The current findings revealed that PTX administration to DOX-intoxicated rats mitigated the pathological manifestations of hepatic injury, reduced microscopical damage scores, and improved serum ALT and AST markers, revealing restored hepatic cellular integrity. These favorable effects were attributed to PTX's ability to mitigate inflammation by reducing hepatic IL-1β and TNF-α levels and suppressing the pro-inflammatory HMGB1/TLR4/NF-κB axis. Moreover, PTX curtailed the hepatic apoptotic abnormalities by suppressing caspase 3 activity and lowering the Bax/Bcl-2 ratio. In tandem, PTX improved the defective autophagy events by lowering hepatic SQSTM-1/p62 accumulation and enhancing the AMPK/mTOR pathway, favoring autophagy and hepatic cell preservation. Together, for the first time, our findings demonstrate the ameliorative effect of PTX against DOX-evoked hepatotoxicity by dampening the hepatic HMGB1/TLR4/NF-κB pro-inflammatory axis and augmenting hepatic AMPK/mTOR-driven autophagy. Thus, PTX could be utilized as an adjunct agent with DOX regimens to mitigate DOX-induced hepatic injury.
Collapse
Affiliation(s)
- Hany H. Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahmed H. Eid
- Department of Pharmacology, Egyptian Drug Authority (EDA)—Formerly NODCAR, Giza 12654, Egypt
| | - Shuruq E. Alsufyani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahmed M. Ashour
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al Qura University, P.O. Box 13578, Makkah 21955, Saudi Arabia
| | | | - Nasser M. Alsharif
- College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | | | | | | | - Hayat A. Abd El Aal
- Department of Pharmacology, Egyptian Drug Authority (EDA)—Formerly NODCAR, Giza 12654, Egypt
| | - Eman S. G. Hassan
- Department of Pharmacology, Egyptian Drug Authority (EDA)—Formerly NODCAR, Giza 12654, Egypt
| | - Wessam H. Elesawy
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza 12568, Egypt
| | - Alzahraa A. Elhemiely
- Department of Pharmacology, Egyptian Drug Authority (EDA)—Formerly NODCAR, Giza 12654, Egypt
| |
Collapse
|
2
|
Chen JM, Bai JY, Yang KX. Effect of resveratrol on doxorubicin resistance in breast neoplasm cells by modulating PI3K/Akt signaling pathway. IUBMB Life 2018; 70:491-500. [PMID: 29637742 DOI: 10.1002/iub.1749] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/17/2018] [Indexed: 11/08/2022]
Abstract
In the study, we probed into the effect of Resveratrol (RES) on Doxorubicin (DOX)-resistant breast neoplasm cell line MCF-7/DOX as well as the mechanism of RES underlying the DOX-resistant breast cancer. CCK-8 assay was utilized to assess the survival rates and sensitivity of breast neoplasm cell lines MCF-7 or MDA-MB-231 to DOX and RES. DOX-resistant MCF-7 cell line was successfully cultivated with DOX dose increasing and was named MCF-7/DOX. Afterwards, wound healing and Transwell assays were performed to measure the migration and invasion capabilities of MCF-7/DOX cells, while cell propagation and apoptosis were determined by colony formation assay and flow cytometry analysis. Both western blotting and immunohistochemistry were conducted to examine the expression of proteins involved in PI3K/Akt signaling pathway. Nude mice xenograft model was constructed to further verify the effects of DOX and RES on breast neoplasm in vivo. RES restored DOX sensitivity in MCF-7/DOX cells, inhibiting biological functions of MCF-7/DOX cells and promoting cell apoptosis in vitro and impeding tumor growth in vivo. It was revealed by the mechanistic studies that MCF-7/DOX cells could regain the drug sensibility with RES treatment through inactivating the PI3K/Akt signal transduction pathway. RES could reverse DOX resistance in breast neoplasm cells and inhibited DOX-resistant breast cancer cell propagation and metastasis and facilitated cell apoptosis by modulating PI3K/Akt signaling pathway. © 2018 IUBMB Life, 70(6):491-500, 2018.
Collapse
Affiliation(s)
- Ju-Min Chen
- Department of Breast and Thyroid Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
| | - Jun-Yun Bai
- Department of Geriatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Kun-Xian Yang
- Department of Breast and Thyroid Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
| |
Collapse
|
3
|
Castellanos-Esparza YC, Wu S, Huang L, Buquet C, Shen R, Sanchez-Gonzalez B, García Latorre EA, Boyer O, Varin R, Jiménez-Zamudio LA, Janin A, Vannier JP, Li H, Lu H. Synergistic promoting effects of pentoxifylline and simvastatin on the apoptosis of triple-negative MDA-MB-231 breast cancer cells. Int J Oncol 2018; 52:1246-1254. [PMID: 29436616 DOI: 10.3892/ijo.2018.4272] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/14/2017] [Indexed: 11/06/2022] Open
Abstract
Pentoxifylline (PTX), a xanthine family molecule and simvastatin (SIM), an anti-hypercholesterolemic agent, have recently been considered as sensitizers to chemotherapy and radiotherapy. The present in vitro study evaluated their antitumor synergistic effects on MDA‑MB‑231 breast cancer cells characterized by the triple‑negative phenotype (TNP). The anti-proliferative effects of these two agents were evaluated by MTT and clonogenic assays. Cell cycle progression was examined using propidium iodide staining. Apoptosis was investigated by Annexin V labeling, and by examining caspase 3 activity and DNA fragmentation. Autophagic vesicles and reactive oxygen species (ROS) levels were monitored by flow cytometry. Western blot analysis was performed to evaluate molecular targets. Our results revealed that when used alone, PTX and SIM exerted antitumor effects. Nevertheless, used in combination, the inhibition of cell proliferation was synergistically superior (80% vs 42%) than that observed following treatment with each agent alone after 48 h. PTX alone (0.5 mM) induced both apoptosis (25%) and autophagy (25%); however, when used in combination with SIM (0.5 µM), the balance between these processes was disrupted and the cells underwent apoptosis (>65%) as opposed to autophagy (<13%). This imbalance was associated with an increase in ERK1/2 and AKT activation, but not with an increase in mTOR phosphorylation, and with the suppression of the NF-κB pathway. In addition, in the cells treated with both agents, almost 78% of the cells were arrested at the G0/G1 phase and lost their colony-forming ability (38±5%) compared to the cells treated with PTX alone (115±5%). On the whole, these results suggest that the induction of autophagy may be a protective mechanism preventing MDA‑MB‑231 cancer cell death. The combined use of PTX and SIM may drive dormant autophagic cancer cells to undergo apoptosis and thus this may be a novel treatment strategy for breast cancer characterized by the TNP.
Collapse
Affiliation(s)
- Yessica Cristina Castellanos-Esparza
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Shuang Wu
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Limin Huang
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Catherine Buquet
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Rong Shen
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Berenice Sanchez-Gonzalez
- Immunochemistry Laboratory I, Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City 11340, Mexico
| | - Ethel Awilda García Latorre
- Immunochemistry Laboratory I, Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City 11340, Mexico
| | - Olivier Boyer
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Remi Varin
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Luis Antonio Jiménez-Zamudio
- Immunochemistry Laboratory I, Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City 11340, Mexico
| | - Anne Janin
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Jean-Pierre Vannier
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Hong Li
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - He Lu
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| |
Collapse
|
4
|
Zhao M, Ding XF, Shen JY, Zhang XP, Ding XW, Xu B. Use of liposomal doxorubicin for adjuvant chemotherapy of breast cancer in clinical practice. J Zhejiang Univ Sci B 2017; 18:15-26. [PMID: 28070993 DOI: 10.1631/jzus.b1600303] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Breast cancer is one of the malignant tumors with the highest morbidity and mortality. It is helpful to reduce the rate of tumor recurrence and metastasis by treating breast cancer with adjuvant chemotherapy, so as to increase the cure rate or survival of patients. In recent years, liposomes have been regarded as a kind of new carrier for targeted drugs. Being effective for enhancing drug efficacy and reducing side effects, they have been widely used for developing anticancer drugs. As a kind of anthracycline with high anticancer activity, doxorubicin can treat or alleviate a variety of malignant tumors effectively when it is used on its own or in combination with other anticancer drugs. Although liposomal doxorubicin has been extensively used in the adjuvant chemotherapy of breast cancer, its exact therapeutic efficacy and side effects have not been definitely proven. Various clinical studies have adopted different combined regimes, dosages, and staging, so their findings differ to certain extent. This paper reviews the clinical application of liposomal doxorubicin in the adjuvant chemotherapy of breast cancer and illustrates therapeutic effects and side effects of pegylated liposomal doxorubicin (PLD) and non-PLD (NPLD) in clinical research, in order to discuss the strategies for applying these drugs in such adjuvant chemotherapy, looking forward to providing references for related research and clinical treatment in terms of dosage, staging, combined regimes, and analysis methods and so on.
Collapse
Affiliation(s)
- Ming Zhao
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xian-Feng Ding
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Jian-Yu Shen
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xi-Ping Zhang
- Department of Tumor Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Xiao-Wen Ding
- Department of Tumor Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Bin Xu
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| |
Collapse
|
5
|
Pentoxifylline affects idarubicin binding to DNA. Bioorg Chem 2016; 65:118-25. [DOI: 10.1016/j.bioorg.2016.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 01/21/2023]
|
6
|
Gołuński G, Borowik A, Wyrzykowski D, Woziwodzka A, Piosik J. Pentoxifylline as a modulator of anticancer drug doxorubicin. Part I: Reduction of doxorubicin DNA binding. Chem Biol Interact 2015; 242:291-8. [PMID: 26499448 DOI: 10.1016/j.cbi.2015.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 10/07/2015] [Accepted: 10/08/2015] [Indexed: 12/26/2022]
Abstract
Pentoxifylline--biologically active aromatic compound--has a well established capability to sequester aromatic ligands, such as an anticancer drug--doxorubicin--in mixed stacking aggregates. Formation of such hetero-complexes may influence biological activity of secluded drug. Presented work shows assessment of pentoxifylline influence on doxorubicin direct interactions with DNA employing biophysical methods. Achievement of this goal required statistical-thermodynamical model allowing numerical four-parameter analysis of experimental mixture--an issue that was successfully tackled by merging McGhee--von Hippel and Kapuscinski--Kimmel models. Results obtained with new model are well in agreement with data obtained with separate experiments with each of these two models and show reduction of doxorubicin in free (monomeric, dimeric) and complexed with DNA forms in favor of doxorubicin-pentoxifylline complexes with increasing pentoxifylline concentration. Developed model appears to be a universal tool allowing numerical analysis of mixtures containing self-aggregating ligand, DNA, and modulating agent.
Collapse
Affiliation(s)
- Grzegorz Gołuński
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology UG-MUG, Kładki 24, 80-822, Gdańsk, Poland
| | - Agnieszka Borowik
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology UG-MUG, Kładki 24, 80-822, Gdańsk, Poland
| | - Dariusz Wyrzykowski
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Anna Woziwodzka
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology UG-MUG, Kładki 24, 80-822, Gdańsk, Poland.
| | - Jacek Piosik
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology UG-MUG, Kładki 24, 80-822, Gdańsk, Poland.
| |
Collapse
|
7
|
Pentoxifylline during steroid window phase at induction to remission increases apoptosis in childhood with acute lymphoblastic leukemia. Clin Transl Oncol 2015; 18:369-74. [PMID: 26329293 DOI: 10.1007/s12094-015-1376-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/04/2015] [Indexed: 01/22/2023]
Abstract
PURPOSE Pentoxifylline (PTX) has been shown to increase chemotherapy-induced apoptosis. A clinical trial was developed to evaluate the effect of the addition of PTX to the induction steroid window phase in children with acute lymphoblastic leukemia (ALL). METHODS Thirty-two children were enrolled on this study. Children with a new diagnosis of ALL were randomly assigned to receive prednisone (PRD) 40 mg/m(2)/day only during the 7-day treatment pre-phase (PRD group, 11 patients) or to receive PRD with PTX (10 mg/kg/day) (PTX group, 11 patients); the control group included children with normal bone marrow (10 patients). Bone marrow aspiration (BMA) was performed at diagnosis (day -7) in all groups, and at day 0 (end of PRD window) for patients with ALL (PRD and PTX groups). Apoptosis was evaluated by flow cytometry (FC) using Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) stains. Statistical analysis was performed using the Mann-Whitney U test. RESULTS Apoptotic index at day -7 was similar in all groups. However, at day 0 post-treatment, apoptosis was significantly higher in the PTX group than in the PRD group (p < 0.001). There were no serious adverse effects associated with PTX. CONCLUSIONS PTX potentiates blast apoptosis induced by PRD in children with ALL during steroid window phase.
Collapse
|
8
|
Nidhyanandan S, Boreddy TS, Chandrasekhar KB, Reddy ND, Kulkarni NM, Narayanan S. Phosphodiesterase inhibitor, pentoxifylline enhances anticancer activity of histone deacetylase inhibitor, MS-275 in human breast cancer in vitro and in vivo. Eur J Pharmacol 2015. [PMID: 26209365 DOI: 10.1016/j.ejphar.2015.07.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
MS-275, a histone deacetylase inhibitor (HDACi), is undergoing clinical trials for treatment of various cancers. Pentoxifylline, a nonselective phosphodiesterase (PDE) inhibitor, has been shown to increase the effectiveness of antitumor chemotherapy. In the present study, the potential anti-cancer activity of MS-275 in combination with pentoxifylline in panel of cell lines and human breast cancer xenograft model were examined. A Panel of cancer cell lines were treated with MS-275 and pentoxifylline to determine their impact on cellular proliferation, cell cycle regulation, apoptosis, anti-angiogenesis. The in vivo activities of MS-275 and pentoxifylline were assessed in a Matrigel plug angiogenesis model and human breast cancer (MDA-MB-231) xenograft model. Combination of MS-275 with pentoxifylline showed enhanced anti-proliferative activity in a panel of cancer cell lines (HCT 116, MCF-7, PC3 and MDA-MB-231). Apoptotic studies performed using, Hoechst staining and cell cycle analysis reveal that this combination at the lower concentrations induces apoptosis downstream of the HDAC inhibition and PDE regulation. Further, combination showed enhanced antiangiogenic activity in Matrigel tube formation assay using HUVECs and in Matrigel plug assay in vivo. A significant inhibition (P<0.001) of tumor growth was observed in mice bearing MDA-MB-231 breast cancer xenograft treated with the combination of MS-275 (5mg/kg p.o.) and pentoxifylline (60 mg/kg i.p.) than treatments alone, without much signs of toxicity. Taken together, our study demonstrated enhanced anticancer activity of MS-275 and pentoxifylline combination both in vitro and in vivo with reduced toxicity. However, further studies are required to understand the mechanism for this combination effect.
Collapse
Affiliation(s)
- Saranya Nidhyanandan
- Department of Biology, Drug Discovery Research, Orchid Chemicals and Pharmaceuticals Ltd., Old Mahabalipuram Road, Sozhanganallur, Chennai 600119, Tamil Nadu, India; Jawaharlal Nehru Technological University Anantapur, Anantapur, 515 002 Andhra Pradesh, India.
| | - Thippeswamy S Boreddy
- Department of Biomedical Science, College of Pharmacy, Shaqra University, Al-Dawadmi, Kingdom of Saudi Arabia
| | | | - Neetinkumar D Reddy
- Department of Biology, Drug Discovery Research, Orchid Chemicals and Pharmaceuticals Ltd., Old Mahabalipuram Road, Sozhanganallur, Chennai 600119, Tamil Nadu, India
| | - Nagaraj M Kulkarni
- Department of Biology, Drug Discovery Research, Orchid Chemicals and Pharmaceuticals Ltd., Old Mahabalipuram Road, Sozhanganallur, Chennai 600119, Tamil Nadu, India
| | - Shridhar Narayanan
- Foundation for Neglected Disease Research, Sir M Visvesvaraya Institute of Technology, International Airport Road, Yelahanka, Bangaluru 562157, India
| |
Collapse
|
9
|
Cancer multidrug resistance-targeted therapy in both cancer and cardiovascular system with cardiovascular drugs. Int J Cardiol 2014; 176:1306-8. [PMID: 25131921 DOI: 10.1016/j.ijcard.2014.07.158] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 07/27/2014] [Indexed: 02/06/2023]
|
10
|
Jain D, Bajaj A, Athawale R, Shrikhande S, Goel PN, Nikam Y, Gude R, Patil S, Prashant Raut P. Surface-coated PLA nanoparticles loaded with temozolomide for improved brain deposition and potential treatment of gliomas: development, characterization and in vivo studies. Drug Deliv 2014; 23:999-1016. [PMID: 25026415 DOI: 10.3109/10717544.2014.926574] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Hydrophobicity of PLA nanoparticles makes them a good substrate for macrophageal and reticulo-endothelial system uptake. Long-circulating properties can be imparted to these particles by coating them with hydrophilic stabilizers. Surface-modified PLA nanoparticles loaded with anti-cancer agent temozolomide were fabricated by solvent evaporation method and coated with surface modifiers. Selection of the surface modifier was based upon uptake of nanoparticles by K9 cells (liver cells). The particles were prepared and characterized for various physicochemical properties using transmission electron microscopy, differential scanning calorimetry, powder X-ray diffraction and in vitro dissolution studies. In vitro BBB permeation studies were performed using the co-culture model developed by using Madin-Darby canine kidney and C6 glioma cells as endothelial and glial cells, respectively. In vitro C6 glioma cell cytotoxicity, cellular proliferation, cellular migration and cellular uptake studies due to developed nanoparticles was assessed. In vivo studies such as pharmacokinetics, qualitative and quantitative biodistribution studies were performed for the developed nanoparticles. Drug-loaded nanoparticles with entrapment efficiency of 50% were developed. PEG-1000 and polysorbate-80 coated nanoparticles were least taken up by the liver cells. Characterization of the nanoparticles revealed formation of spherical shape nanoparticles, with no drug and excipient interaction. In vivo pharmacokinetics of developed nanoparticles depicted enhancement of half-life, area under the curve and mean residence time of the drug. Qualitative and quantitative biodistribution studies confirmed enhanced permeation of the drug into the brain upon loading into nanoparticles with less deposition in the highly perfused organs like lung, liver, spleen, heart and kidney.
Collapse
Affiliation(s)
- Darshana Jain
- a Department of Pharmaceutics , CU Shah College of Pharmacy , Mumbai , Maharashtra , India
| | - Amrita Bajaj
- b SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , Maharashtra , India
| | - Rajani Athawale
- a Department of Pharmaceutics , CU Shah College of Pharmacy , Mumbai , Maharashtra , India
| | - Shruti Shrikhande
- a Department of Pharmaceutics , CU Shah College of Pharmacy , Mumbai , Maharashtra , India
| | - Peeyush N Goel
- c ACTREC, Tata Memorial Cancer Centre , Mumbai , Maharashtra , India , and
| | - Yuvraj Nikam
- c ACTREC, Tata Memorial Cancer Centre , Mumbai , Maharashtra , India , and
| | - Rajiv Gude
- c ACTREC, Tata Memorial Cancer Centre , Mumbai , Maharashtra , India , and
| | | | | |
Collapse
|