1
|
Jana M, Patro CS, Sharma S, Naskar S, Biswas UK, Debnath B. Development and evaluation of solid lipid nanoparticles for enhanced peroral bioavailability of capecitabine. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-26. [PMID: 40378399 DOI: 10.1080/09205063.2025.2505344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 05/08/2025] [Indexed: 05/18/2025]
Abstract
This work aimed to improve the peroral bioavailability of capecitabine (CPB) by developing and assessing solid lipid nanoparticles (SLNs). SLNs were made using the modified nanoprecipitation method. Particle size, zeta potential, entrapment efficiency, drug loading, in-vitro drug release, TEM, in-vivo pharmacokinetic study, stability study, histopathological evaluation and cytotoxicity study were assessed. The TEM revealed that the SLNs were transparent, with a mean particle size ranging from 13.06 ± 0.09 to 86.10 ± 0.15 nm. The F-3 formulation demonstrated the highest drug entrapment efficiency at 45.49 ± 0.28. The zeta potential and polydispersity index of all SLNs ranged from -15.53 ± 0.17 to 17.55 ± 0.18 mV and from 0.1356 ± 0.11 to 0.2678 ± 0.13, respectively. The drug entrapment efficiency and drug loading of all SLNs ranged from 18.45 ± 0.36 to 45.49 ± 0.28 and from 21.75 ± 0.64 to 59.49 ± 0.38, respectively. The CPB-SLNs showed sustained drug release with prolonged plasma retention, delayed Tmax, and extended half-life compared to raw CPB. In vivo pharmacokinetic studies suggest that developed SLNs may enhance therapeutic efficacy by maintaining drug concentrations in plasma for longer periods. Toxicity was observed at 200 mg/kg/day, indicated by changes in clinical biochemistry, organ weights, and histopathology, particularly affecting the liver and kidneys. Therefore, it can be said that these developed SLNs may be among the best preparations for the delivery of anti-cancer drugs for improved therapeutic efficacy.
Collapse
Affiliation(s)
- Mayukh Jana
- School of Pharmacy, Centurion University of Technology and Management, Centurion University, Bhubaneswar, Odisha, India
| | - Chandra Sekhar Patro
- Chandra Sekhar Patro, School of Pharmacy, Centurion University of Technology and Management, Centurion University, Bhubaneswar, Odisha, India
| | - Suraj Sharma
- Sikkim Professional College of Pharmaceutical Sciences, Sikkim, India
| | - Sweet Naskar
- Institute of Pharmacy, Kalyani, Nadia, West Bengal, India
| | - Ujjwal Kumar Biswas
- Research Scholar, School of Pharmaceutical Science (SPS), Siksha O Anusandhan(SOA) University, Ghatkia, Bhubaneswar, Odisha, India
| | | |
Collapse
|
2
|
Kaya Çakir H, Eroğlu O. Investigation of the synergic effect of mocetinostat and capecitabine in a triple-negative breast neoplasms mouse model. J Investig Med 2025; 73:320-327. [PMID: 39690705 DOI: 10.1177/10815589241309603] [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: 12/19/2024]
Abstract
Combined administration of two or more drugs is emerging as a new strategy in triple-negative breast neoplasms. This is the first study to investigate the combination of the histone deacetylase inhibitor mocetinostat and the antimetabolite drug capecitabine in triple-negative mammary neoplasms in a preclinical mouse model. Thirty-five female mice were grouped into the control group, capecitabine group, mocetinostat group, and combined drugs group. At the end of the experimental period, body weight, and tumor weight were measured and tumor tissue and lung tissue were histologically examined. The results showed that the body weight of mice in the drug-treated groups was reduced by about 18%. Tumor weights were also reduced by 21% in the mocetinostat group, 27.5% in the capecitabine group, and 45% in the combined group. The combination of mocetinostat and capecitabine decreased the formation of tumors and metastases in lung tissue. In summary, the combination of mocetinostat and capecitabine was more effective than either drug alone in reducing the size of triple-negative breast neoplasms in a mouse model.
Collapse
Affiliation(s)
- Hacer Kaya Çakir
- Department of Molecular Biology and Genetics, Faculty of Science, Bilecik Seyh Edebali University, Bilecik, Turkey
| | - Onur Eroğlu
- Department of Molecular Biology and Genetics, Faculty of Science, Bilecik Seyh Edebali University, Bilecik, Turkey
| |
Collapse
|
3
|
Zehra K, Banu A, Can E, Hülya C. Fisetin and/or capecitabine causes changes in apoptosis pathways in capecitabine-resistant colorectal cancer cell lines. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7913-7926. [PMID: 38748229 PMCID: PMC11449987 DOI: 10.1007/s00210-024-03145-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 10/04/2024]
Abstract
Capecitabine is recommended as one of the first-line chemotherapy treatments for advanced or metastatic colorectal cancer. Researches have been conducted on capecitabine's impact on the viability of human colon cancer cells and its potential to induce apoptosis. However, even in cases initially responsive to treatment, the development of acquired resistance significantly limits its efficacy. Challenges still exist in effectively treating patients with chemotherapy, and developing new cytotoxic drugs is hindered by drug resistance. Fisetin alters the cell cycle, inducing apoptosis, inhibiting cancer cell proliferation, and enhancing the therapeutic effectiveness of chemotherapy drugs. This work aims to create a plan for reversing capecitabine resistance. For this purpose, the role of capecitabine and/or fisetin combinations in cell proliferation and apoptosis has been determined in both wild-type and capecitabine-resistant HT29 cells (CR/HT29). We developed capecitabine-resistant cell line from wild-type HT29 cells. This study demonstrated the effects of capecitabine, fisetin, and their combinations on both resistant and wild-type cells through experiments including cell survival skills, cell proliferation, wound healing, colony formation, hoechst staining, and western blot analysis. We established capecitabine-resistant cell lines. P-gp expression increased in CR/HT29 cells. Capecitabine effects on a CR/HT29 cells less than wild-type HT29 cells. The combination of fisetin and capecitabine in cell proliferation caused greater reductions in wild-type HT29 cells than in capecitabine-resistant cells. Fisetin has also additive effects on the apoptotic pathway in CR/HT29 cells. This study provides new perspectives on the combination of capecitabine and/or flavonoid treatment in resistant cells.
Collapse
Affiliation(s)
- Kanli Zehra
- Institute of Health Sciences, Marmara University, Basibuyuk-Maltepe, Istanbul, 34854, Turkey
| | - Aydin Banu
- School of Medicine, Department of Biophysics, Marmara University, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey
| | - Erzik Can
- School of Medicine, Department of Medical Biology, Marmara University, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey
| | - Cabadak Hülya
- School of Medicine, Department of Biophysics, Marmara University, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey.
| |
Collapse
|
4
|
Li H, Wang Y, Zhang M, Wang H, Cui A, Zhao J, Ji W, Chen YG. Establishment of porcine and monkey colonic organoids for drug toxicity study. CELL REGENERATION 2021; 10:32. [PMID: 34599392 PMCID: PMC8486901 DOI: 10.1186/s13619-021-00094-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/02/2021] [Indexed: 12/27/2022]
Abstract
Pig and monkey are widely used models for exploration of human diseases and evaluation of drug efficiency and toxicity, but high cost limits their uses. Organoids have been shown to be promising models for drug test as they reasonably preserve tissue structure and functions. However, colonic organoids of pig and monkey are not yet established. Here, we report a culture medium to support the growth of porcine and monkey colonic organoids. Wnt signaling and PGE2 are important for long-term expansion of the organoids, and their withdrawal results in lineage differentiation to mature cells. Furthermore, we observe that porcine colonic organoids are closer to human colonic organoids in terms of drug toxicity response. Successful establishment of porcine and monkey colonic organoids would facilitate the mechanistic investigation of the homeostatic regulation of the intestine of these animals and is useful for drug development and toxicity studies.
Collapse
Affiliation(s)
- Haonan Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yalong Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Mengxian Zhang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Hong Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Along Cui
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianguo Zhao
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China. .,Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510700, China.
| |
Collapse
|
5
|
Singh DD, Yadav DK. TNBC: Potential Targeting of Multiple Receptors for a Therapeutic Breakthrough, Nanomedicine, and Immunotherapy. Biomedicines 2021; 9:biomedicines9080876. [PMID: 34440080 PMCID: PMC8389539 DOI: 10.3390/biomedicines9080876] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/08/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous, recurring cancer associated with a high rate of metastasis, poor prognosis, and lack of therapeutic targets. Although target-based therapeutic options are approved for other cancers, only limited therapeutic options are available for TNBC. Cell signaling and receptor-specific targets are reportedly effective in patients with TNBC under specific clinical conditions. However, most of these cancers are unresponsive, and there is a requirement for more effective treatment modalities. Further, there is a lack of effective biomarkers that can distinguish TNBC from other BC subtypes. ER, PR, and HER2 help identify TNBC and are widely used to identify patients who are most likely to respond to diverse therapeutic strategies. In this review, we discuss the possible treatment options for TNBC based on its inherent subtype receptors and pathways, such as p53 signaling, AKT signaling, cell cycle regulation, DNA damage, and programmed cell death, which play essential roles at multiple stages of TNBC development. We focus on poly-ADP ribose polymerase 1, androgen receptor, vascular endothelial growth factor receptor, and epidermal growth factor receptor as well as the application of nanomedicine and immunotherapy in TNBC and discuss their potential applications in drug development for TNBC.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur 303002, India;
| | - Dharmendra Kumar Yadav
- Department of Pharmacy and Gachon Institute of Pharmaceutical Science, College of Pharmacy, Gachon University, Hambakmoeiro 191, Yeonsu-gu, Incheon 21924, Korea
- Correspondence: ; Tel.: +82-32-820-4948
| |
Collapse
|
6
|
Zhao K, Wang M, Kang H, Wu A. A prognostic five long-noncoding RNA signature for patients with rectal cancer. J Cell Biochem 2020; 121:3854-3860. [PMID: 31709598 DOI: 10.1002/jcb.29549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/24/2019] [Indexed: 01/24/2023]
Abstract
This study aimed to identify prognostic long noncoding RNAs (lncRNAs) signature for predicting the prognosis of patients with rectal cancer. LncRNA-sequencing data and clinicopathological data of patients with rectal cancer were retrieved from The Cancer Genome Atlas database. Univariate and multivariate Cox proportional hazards regression analysis, the least absolute shrinkage, and selection operator analysis and the Kaplan-Meier curve method were employed to identify prognostic lncRNAs and construct multi-lncRNA signature. Finally, five lncRNAs (AC079789.1, AC106900.2, AL121987.1, AP004609.1, and LINC02163) were identified to construct a five-lncRNA signature. According to the five-lncRNA signature, patients with rectal cancer were divided into a high-risk group and low-risk group. Patients with rectal cancer had significantly poorer overall survival in the high-risk group than in the low-risk group. We used a time-dependent receiver operating characteristic curve to assess the power of the five-lncRNA signature by calculating the area under the curve (AUC). The AUCs for predicting 3-year survival and 5-year survival were 0.742 and 0.935, respectively, which indicated a good performance of the five-lncRNA signature. The five-lncRNA signature was independently associated with the prognosis of patients with rectal cancer through using univariate and multivariate Cox regression analysis. The biological function of the five lncRNAs was enriched in some cancer-related biological processes and pathways by performing functional enrichment analysis of their correlated protein-coding genes. In conclusion, we developed a five-lncRNA signature as a potential indicator for rectal cancer.
Collapse
Affiliation(s)
- Kankan Zhao
- Department of General Surgery, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Mengchuan Wang
- Department of General Surgery, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Houlong Kang
- Department of General Surgery, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Aiguo Wu
- Department of General Surgery, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Jin W. Regulation of Src Family Kinases during Colorectal Cancer Development and Its Clinical Implications. Cancers (Basel) 2020; 12:cancers12051339. [PMID: 32456226 PMCID: PMC7281431 DOI: 10.3390/cancers12051339] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/11/2022] Open
Abstract
Src family kinases (SFKs) are non-receptor kinases that play a critical role in the pathogenesis of colorectal cancer (CRC). The expression and activity of SFKs are upregulated in patients with CRC. Activation of SFKs promotes CRC cell proliferation, metastases to other organs and chemoresistance, as well as the formation of cancer stem cells (CSCs). The enhanced expression level of Src is associated with decreased survival in patients with CRC. Src-mediated regulation of CRC progression involves various membrane receptors, modulators, and suppressors, which regulate Src activation and its downstream targets through various mechanisms. This review provides an overview of the current understanding of the correlations between Src and CRC progression, with a special focus on cancer cell proliferation, invasion, metastasis and chemoresistance, and formation of CSCs. Additionally, this review discusses preclinical and clinical strategies to improve the therapeutic efficacy of drugs targeting Src for treating patients with CRC.
Collapse
Affiliation(s)
- Wook Jin
- Laboratory of Molecular Disease and Cell Regulation, Department of Biochemistry, School of Medicine, Gachon University, Incheon 406-840, Korea
| |
Collapse
|