1
|
Saharkhiz S, Zarepour A, Nasri N, Cordani M, Zarrabi A. A comparison study between doxorubicin and curcumin co-administration and co-loading in a smart niosomal formulation for MCF-7 breast cancer therapy. Eur J Pharm Sci 2023; 191:106600. [PMID: 37802230 DOI: 10.1016/j.ejps.2023.106600] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/04/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023]
Abstract
Chemotherapy agents often exhibit limited effectiveness due to their fast elimination from the body and non-targeted delivery. Emerging nanomaterials as drug delivery carriers open new expectancy to overcome these limitations in current chemotherapeutic treatments. In this study, we introduce and evaluate a smart pH-responsive niosomal formulation capable of delivering Doxorubicin (DOX) and Curcumin (CUR) in both individually and co-loaded forms. In particular, drug-loaded niosomes were prepared using thin-film hydration method and then characterized via different physicochemical analyses. The pH responsivity of the carrier was assessed by performing a drug release study in three different pH conditions (4, 6.5, and 7.4). Finally, the anticancer efficacy of the therapeutic compounds was evaluated through the MTT assay. Our results showed spherical particles with a size of about 200 nm and -2 mV surface charge. Encapsulation efficiency (EE%) of the nanocarrier was about 77.06 % and 79.08 % for DOX and CUR, respectively. The release study confirmed the pH responsivity of the carrier. The MTT assay results revealed about 39 % and 43 % of cell deaths after treatment with cur-loaded and dox-loaded niosomes, which increased to 74 % and 79 % after co-administration and co-loading forms of drugs, respectively, exhibiting increased anticancer efficacy by selectively delivering DOX and CUR individually or in combination. Overall, these findings suggest that our nanoformulation holds the potential as a targeted and highly effective approach for cancer management and therapy, overcoming the limitations of conventional chemotherapy drugs.
Collapse
Affiliation(s)
- Shaghayegh Saharkhiz
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye
| | - Negar Nasri
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid 28040, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid 28040, Spain.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye.
| |
Collapse
|
2
|
Abdel-Sattar OE, Allam RM, Al-Abd AM, El-Halawany AM, EL-Desoky AM, Mohamed SO, Sweilam SH, Khalid M, Abdel-Sattar E, Meselhy MR. Hypophyllanthin and Phyllanthin from Phyllanthus niruri Synergize Doxorubicin Anticancer Properties against Resistant Breast Cancer Cells. ACS OMEGA 2023; 8:28563-28576. [PMID: 37576627 PMCID: PMC10413485 DOI: 10.1021/acsomega.3c02953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023]
Abstract
Doxorubicin (DOX) is a cornerstone chemotherapeutic agent for the treatment of several malignancies such as breast cancer; however, its activity is ameliorated by the development of a resistant phenotype. Phyllanthus species have been studied previously for their potential anticancer properties. The current work is aimed to study the potential cytotoxicity and chemomodulatory effects of hypophyllanthin (PN4) and phyllanthin (PN5) isolated from Phyllanthus niruri to DOX against the adriamycin multidrug-resistant breast cancer cells (MCF-7ADR) and elucidate their mechanism of action. The major compounds of the active methylene chloride fraction were isolated and assessed for their potential cytotoxicity and chemomodulatory effects on DOX against naïve (MCF-7) and resistant breast (MCF-7ADR) cancer cells. The mechanism of action of both compounds in terms of their impacts on programmed/non-programmed cell death (apoptosis and autophagy/necrosis), cell cycle progression/arrest, and tumor cell migration/invasion was investigated. Both compounds PN4 and PN5 showed a moderate but similar potency against MCF-7 as well as MCF-7ADR and significantly synergized DOX-induced anticancer properties against MCF-7ADR. The chemomodulatory effect of both compounds to DOX was found to be via potentiating DOX-induced cell cycle interference and apoptosis induction. It was found that PN4 and PN5 blocked the apoptosis-escape autophagy pathway in MCF-7ADR. On the molecular level, both compounds interfered with SIRT1 expression and consequently suppressed Akt phosphorylation, and PN5 blocked apoptosis escape. Furthermore, PN4 and PN5 showed promising antimigratory and anti-invasive effects against MCF-7ADR, as confirmed by suppression of N-cadherin/β-catenin expression. In conclusion, for the first time, hypophyllanthin and phyllanthin isolated from P. niruri showed promising chemomodulatory effects to the DOX-induced chemotherapeutic activity against MCF-7ADR. Both compounds significantly synergized DOX-induced anticancer properties against MCF-7ADR. This enhanced activity was explained by further promoting DOX-induced apoptosis and suppressing the apoptosis-escape autophagy feature of the resistant breast cancer cells. Both compounds (hypophyllanthin and phyllanthin) interfered with the SIRT1/Akt pathway and suppressed the N-cadherin/β-catenin axis, confirming the observed antiproliferative, cytotoxic, and anti-invasive effects of hypophyllanthin and phyllanthin.
Collapse
Affiliation(s)
- Ola E. Abdel-Sattar
- Pharmacognosy Department,
Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
| | - Rasha M. Allam
- Pharmacology
Department, Medical Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Ahmed M. Al-Abd
- Pharmacology
Department, Medical Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Ali M. El-Halawany
- Pharmacognosy Department,
Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
| | - Ahmed M. EL-Desoky
- Department of Molecular Biology,
Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City (USC), Sadat City 32958, Egypt
| | - Shanaz O. Mohamed
- School of Pharmaceutical
Sciences, Universiti Sains Malaysia, Gelugor, Penang 11700, Malaysia
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of
Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo 11829, Egypt
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Essam Abdel-Sattar
- Pharmacognosy Department,
Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
| | - Meselhy R. Meselhy
- Pharmacognosy Department,
Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
| |
Collapse
|
3
|
Amalina ND, Salsabila IA, Zulfin UM, Jenie RI, Meiyanto E. In vitro synergistic effect of hesperidin and doxorubicin downregulates epithelial-mesenchymal transition in highly metastatic breast cancer cells. J Egypt Natl Canc Inst 2023; 35:6. [PMID: 36967442 DOI: 10.1186/s43046-023-00166-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 03/15/2023] [Indexed: 03/28/2023] Open
Abstract
Abstract
Background
We previously reported that in highly metastatic breast cancer cells, doxorubicin (DOX) at non-toxic concentrations promoted cell migration and invasion. Hesperidin (30, 5, 9-dihydroxy-40-methoxy-7-orutinosyl flavanone) is a flavonoid glycoside isolated from citrus/lemon plant that possesses a cytotoxic effect in several cancer cells. In this study, we investigate whether DOX efficacy is enhanced by hesperidin (Hsd) and the molecular pathway involved in highly metastatic breast cancer, 4T1.
Methods
Combined cytotoxicity of Hsd and DOX was evaluated with MTT assay and was analyzed using Chou-Talalay’s method. To better understand the underlying mechanism, several factors, including apoptosis and cell cycle arrest were analyzed by flow cytometry. In addition, antimigration activity was evaluated by scratch wound healing assay, MMP-9 expression by ELISA and gelatin zymography, and Rac-1 protein level using western blot. The data on survival rate and expression level of MMP-9 and Rac-1 were obtained from Gene Expression OMNIBUS (GEO).
Results
Under MTT assay, Hsd showed a cytotoxic effect in a concentration-dependent manner with an IC50 value of 284 µM on 4T1 cells. Hsd synergistically enhanced the cytotoxic effect of DOX which seemed to correlate with an increase in apoptotic cell death, G2/M cell cycle arrest and blocked the migration of 4T1 cells. At 10 nM, doxorubicin induced lamellipodia formation, and increased the level of Rac-1 and metalloproteinase-9 (MMP-9) expression. Interestingly, combined treatment of DOX and Hsd dramatically downregulated the expression of MMP-9 and Rac-1. These results indicated that Hsd block the cell migration induced by DOX under in vitro studies.
Conclusion
These findings strongly suggest that Hsd possesses a potential synergistic effect that can be developed to enhance the anticancer efficacy of DOX and reduce the risks of chemotherapy use in highly metastatic breast cancer.
Collapse
|
4
|
Hartanto MM, Prajoko YW, Putra A, Amalina ND. The Combination of Mesenchymal Stem Cells and Bovine Colostrum in Reducing α-SMA Expression and NLR Levels in Wistar Rats After 50% Fibrotic Liver Resection. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.10557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Liver fibrogenesis will produce α-smooth muscle actin (α-SMA) expression and a continuous inflammatory process, seen through the neutrophil lymphocyte ratio (NLR). The combination of mesenchymal stem cells and bovine colostrum is a novel strategy for repairing hepatic fibrosis tissue. To assess the combination of mesenchymal stem cells and bovine colostrum to reduce α-SMA expression and NLR levels in Wistar rats after 50% fibrotic liver resection.
Methods: Thirty-six Wistar male rats were randomly divided into 6 groups (sham, control, colostrum, MSCs, and colostrum and MSCs combination). Rats were injected with CCl4 for 8 weeks to induce liver fibrosis then underwent liver resection. NLR levels was determined using Hematology Analyzer, α-SMA expression of myofibroblast was analyzed by immunofluorescence staining.
Results: A significant reduction in NLR levels on day 3 in the treatment group I (1.10), treatment II (0.83), treatment III (0.93) compared to the control group. A significant reduction in NLR levels on day 10 in the treatment group I (0.76), treatment II (0.64), treatment III (0.54) compared to the control group. A significant decrease in α-SMA in treatment group I (0.134), treatment II (0.68), treatment III (0.42) compared to the control group.
Conclusion: In this study, it was found that α-SMA expression, NLR levels on the 3rd and 10th day of administration were reduced in group receiving combination of mesenchymal stem cells and bovine colostrum in the liver of post-resection Wistar rats by 50%.
Collapse
|
5
|
Emerging role of exosomes as biomarkers in cancer treatment and diagnosis. Crit Rev Oncol Hematol 2021; 169:103565. [PMID: 34871719 DOI: 10.1016/j.critrevonc.2021.103565] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is a leading cause of death worldwide and cancer incidence and mortality are rapidly growing. These massive amounts of cancer patients require rapid diagnosis and efficient treatment strategies. However, the currently utilized methods are invasive and cost-effective. Recently, the effective roles of exosomes as promising diagnostic, prognostic, and predictive biomarkers have been revealed. Exosomes are membrane-bound extracellular vesicles containing RNAs, DNAs, and proteins, and are present in a wide array of body fluids. Exosomal cargos have shown the potential to detect various types of cancers at early stages with high sensitivity and specificity. They can also delivery therapeutic agents efficiently. In this article, an overview of recent advances in the research of exosomal biomarkers and their applications in cancer diagnosis and treatment has been provided. Furthermore, the advantages and challenges of exosomes as liquid biopsy targets are discussed and the clinical implications of using exosomal miRNAs have been revealed.
Collapse
|