1
|
Steers GJ, O’Leary BR, Du J, Wagner BA, Carroll RS, Domann FE, Goswami PC, Buettner GR, Cullen JJ. Pharmacologic Ascorbate and DNMT Inhibitors Increase DUOX Expression and Peroxide-Mediated Toxicity in Pancreatic Cancer. Antioxidants (Basel) 2023; 12:1683. [PMID: 37759986 PMCID: PMC10525653 DOI: 10.3390/antiox12091683] [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: 07/13/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Recent studies have demonstrated an important role for vitamin C in the epigenetic regulation of cancer-related genes via DNA demethylation by the ten-eleven translocation (TET) methylcytosine dioxygenase enzymes. DNA methyltransferase (DNMT) reverses this, increasing DNA methylation and decreasing gene expression. Dual oxidase (DUOX) enzymes produce hydrogen peroxide (H2O2) in normal pancreatic tissue but are silenced in pancreatic cancer (PDAC). Treatment of PDAC with pharmacologic ascorbate (P-AscH-, intravenous, high dose vitamin C) increases DUOX expression. We hypothesized that inhibiting DNMT may act synergistically with P-AscH- to further increase DUOX expression and cytotoxicity of PDAC. PDAC cells demonstrated dose-dependent increases in DUOX mRNA and protein expression when treated with DNMT inhibitors. PDAC cells treated with P-AscH- + DNMT inhibitors demonstrated increased DUOX expression, increased intracellular oxidation, and increased cytotoxicity in vitro and in vivo compared to either treatment alone. These findings suggest a potential therapeutic, epigenetic mechanism to treat PDAC.
Collapse
Affiliation(s)
- Garett J. Steers
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, Iowa City, IA 52242, USA; (G.J.S.); (B.R.O.); (J.D.); (B.A.W.); (R.S.C.); (F.E.D.); (P.C.G.); (G.R.B.)
- The Department of Surgery, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Brianne R. O’Leary
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, Iowa City, IA 52242, USA; (G.J.S.); (B.R.O.); (J.D.); (B.A.W.); (R.S.C.); (F.E.D.); (P.C.G.); (G.R.B.)
- The Department of Surgery, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Juan Du
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, Iowa City, IA 52242, USA; (G.J.S.); (B.R.O.); (J.D.); (B.A.W.); (R.S.C.); (F.E.D.); (P.C.G.); (G.R.B.)
- The Department of Surgery, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Brett A. Wagner
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, Iowa City, IA 52242, USA; (G.J.S.); (B.R.O.); (J.D.); (B.A.W.); (R.S.C.); (F.E.D.); (P.C.G.); (G.R.B.)
| | - Rory S. Carroll
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, Iowa City, IA 52242, USA; (G.J.S.); (B.R.O.); (J.D.); (B.A.W.); (R.S.C.); (F.E.D.); (P.C.G.); (G.R.B.)
- The Department of Surgery, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Frederick E. Domann
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, Iowa City, IA 52242, USA; (G.J.S.); (B.R.O.); (J.D.); (B.A.W.); (R.S.C.); (F.E.D.); (P.C.G.); (G.R.B.)
| | - Prabhat C. Goswami
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, Iowa City, IA 52242, USA; (G.J.S.); (B.R.O.); (J.D.); (B.A.W.); (R.S.C.); (F.E.D.); (P.C.G.); (G.R.B.)
| | - Garry R. Buettner
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, Iowa City, IA 52242, USA; (G.J.S.); (B.R.O.); (J.D.); (B.A.W.); (R.S.C.); (F.E.D.); (P.C.G.); (G.R.B.)
| | - Joseph J. Cullen
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, Iowa City, IA 52242, USA; (G.J.S.); (B.R.O.); (J.D.); (B.A.W.); (R.S.C.); (F.E.D.); (P.C.G.); (G.R.B.)
- The Department of Surgery, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
2
|
Raja Arul GL, Toruner MD, Gatenby RA, Carr RM. Ecoevolutionary biology of pancreatic ductal adenocarcinoma. Pancreatology 2022; 22:730-740. [PMID: 35821188 DOI: 10.1016/j.pan.2022.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common histological subtype of pancreatic cancer, is an aggressive disease predicted to be the 2nd cause of cancer mortality in the US by 2040. While first-line therapy has improved, 5-year overall survival has only increased from 5 to ∼10%, and surgical resection is only available for ∼20% of patients as most present with advanced disease, which is invariably lethal. PDAC has well-established highly recurrent mutations in four driver genes including KRAS, TP53, CDKN2A, and SMAD4. Unfortunately, these genetic drivers are not currently therapeutically actionable. Despite extensive sequencing efforts, few additional significantly recurrent and druggable drivers have been identified. In the absence of targetable mutations, chemotherapy remains the mainstay of treatment for most patients. Further, the role of the above driver mutations on PDAC initiation and early development is well-established. However, these mutations alone cannot account for PDAC heterogeneity nor discern early from advanced disease. Taken together, management of PDAC is an example highlighting the shortcomings of the current precision medicine paradigm. PDAC, like other malignancies, represents an ecoevolutionary process. Better understanding the disease through this lens can facilitate the development of novel therapeutic strategies to better control and cure PDAC. This review aims to integrate the current understanding of PDAC pathobiology into an ecoevolutionary framework.
Collapse
Affiliation(s)
| | - Merih D Toruner
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Robert A Gatenby
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Ryan M Carr
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Zhao Y, Yang M, Wang S, Abbas SJ, Zhang J, Li Y, Shao R, Liu Y. An Overview of Epigenetic Methylation in Pancreatic Cancer Progression. Front Oncol 2022; 12:854773. [PMID: 35296007 PMCID: PMC8918690 DOI: 10.3389/fonc.2022.854773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Over the past decades, the aberrant epigenetic modification, apart from genetic alteration, has emerged as dispensable events mediating the transformation of pancreatic cancer (PC). However, the understanding of molecular mechanisms of methylation modifications, the most abundant epigenetic modifications, remains superficial. In this review, we focused on the mechanistic insights of DNA, histone, and RNA methylation that regulate the progression of PC. The methylation regulators including writer, eraser and reader participate in the modification of gene expression associated with cell proliferation, invasion and apoptosis. Some of recent clinical trials on methylation drug targeting were also discussed. Understanding the novel regulatory mechanisms in the methylation modification may offer alternative opportunities to improve therapeutic efficacy to fight against this dismal disease.
Collapse
Affiliation(s)
- Yuhao Zhao
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
| | - Mao Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
| | - Shijia Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
| | - Sk Jahir Abbas
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, China
| | - Junzhe Zhang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
| | - Yongsheng Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
| | - Rong Shao
- Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yingbin Liu, ; Rong Shao,
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- *Correspondence: Yingbin Liu, ; Rong Shao,
| |
Collapse
|
4
|
Interstitial Control-Released Polymer Carrying a Targeting Small-Molecule Drug Reduces PD-L1 and MGMT Expression in Recurrent High-Grade Gliomas with TMZ Resistance. Cancers (Basel) 2022; 14:cancers14041051. [PMID: 35205800 PMCID: PMC8870243 DOI: 10.3390/cancers14041051] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/08/2022] [Accepted: 02/13/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary This study reports a potential new drug—Cerebraca wafer—that is designed to deliver its active pharmaceutical ingredient, (Z)-n-butylidenephthalide (BP), directly into the surgical cavity created when a brain tumor is resected. The therapeutic mechanism of Cerebraca wafer was shown to involve the following: (1) an IC50 of BP against tumor stem cells four times lower than that of bis-chloroethylnitrosourea (BCNU); (2) a synergistic effect between BP and temozolomide (TMZ), as demonstrated by a reduction in O6-methylguanine-DNA-methyltransferase (MGMT) expression level; (3) BP inhibition of programmed cell death-ligand 1 (PD-L1) protein levels, thereby activating T-cell cytotoxicity and increasing interferon-gamma (IFN-γ) secretion. The implantation of Cerebraca wafer is safe, no drug-related adverse events (AEs) and serious AEs (SAEs) were found. The median overall survival (OS) of patients receiving high-dose Cerebraca wafer have exceeded 17.4 months, and a 100% progression-free survival (PFS) rate at six month was achieved. In sum, these findings demonstrate that the Cerebraca wafer has superior therapeutic effects to Gliadel wafer in recurrent high-grade gliomas. Abstract In recurrent glioblastoma, Gliadel wafer implantation after surgery has been shown to result in incomplete chemical removal of residual tumor and development of brain edema. Furthermore, temozolomide (TMZ) resistance caused by O6-methylguanine-DNA-methyltransferase (MGMT) activation and programmed cell death-ligand 1 (PD-L1) expression leads to immune-cold lesions that result in poorer prognosis. Cerebraca wafer, a biodegradable polymer containing (Z)-n-butylidenephthalide (BP), is designed to eliminate residual tumor after glioma resection. An open-label, one-arm study with four dose cohorts, involving a traditional 3 + 3 dose escalation clinical trial, of the Cerebraca wafer combined with TMZ on patients with recurrent high-grade glioma, was conducted. Of the 12 patients who receive implantation of Cerebraca wafer, there were no drug-related adverse events (AEs) or serious AEs (SAEs). The median overall survival (OS) of patients receiving low-dose Cerebraca wafer was 12 months in the group with >25% wafer coverage of the resected tumor, which is longer than OS duration in previously published studies (Gliadel wafer, 6.4 months). Patients who received high-dose Cerebraca wafer treatment had not yet died at the data cut-off date; a 100% progression-free survival (PFS) rate at six month was achieved, indicating the median OS of cohort IV was more than 17.4 months. In vitro study of the primary cells collected from the patients revealed that the IC50 of BP against tumor stem cells was four times lower than that of bis-chloroethylnitrosourea (BCNU). A synergistic effect between BP and TMZ was demonstrated by a reduction in MGMT expression. Furthermore, BP inhibited PD-L1 expression, thereby activating T-cell cytotoxicity and increasing interferon-gamma (IFN-γ) secretion. The better therapeutic effect of Cerebraca wafer on recurrent high-grade glioma could occur through re-sensitization of TMZ and reduction of PD-L1.
Collapse
|
5
|
Diverse and precision therapies open new horizons for patients with advanced pancreatic ductal adenocarcinoma. Hepatobiliary Pancreat Dis Int 2022; 21:10-24. [PMID: 34538570 DOI: 10.1016/j.hbpd.2021.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 08/31/2021] [Indexed: 02/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a common cause of cancer-related death, and most patients are with advanced disease when diagnosed. At present, despite a variety of treatments have been developed for PDAC, few effective treatment options are available; on the other hand, PDAC shows significant resistance to chemoradiotherapy, targeted therapy, and immunotherapy due to its heterogeneous genetic profile, molecular signaling pathways, and complex tumor immune microenvironment. Nevertheless, over the past decades, there have been many new advances in the key theory and understanding of the intrinsic mechanisms and complexity of molecular biology and molecular immunology in pancreatic cancer, based on which more and more diverse new means and reasonable combination strategies for PDAC treatment have been developed and preliminary breakthroughs have been made. With the continuous exploration, from surgical local treatment to comprehensive medical management, the research-diagnosis-management system of pancreatic cancer is improving. This review focused on the variety of treatments for advanced PDAC, including traditional chemotherapy, targeted therapy, immunotherapy, microenvironment matrix regulation as well as the treatment targeting epigenetics, metabolism and cancer stem cells. We pointed out the current research bottlenecks and future exploration directions.
Collapse
|
6
|
Fang X, Poulsen R, Zhao L, Wang J, Rivkees SA, Wendler CC. Knockdown of DNA methyltransferase 1 reduces DNA methylation and alters expression patterns of cardiac genes in embryonic cardiomyocytes. FEBS Open Bio 2021. [PMID: 34235895 PMCID: PMC8329956 DOI: 10.1002/2211-5463.13252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/14/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
We previously found that DNA methyltransferase 3a (DNMT3a) plays an important role in regulating embryonic cardiomyocyte gene expression, morphology, and function. In this study, we investigated the role of the most abundant DNMT in mammalian cells, DNMT1, in these processes. It is known that DNMT1 is essential for embryonic development, during which it is involved in regulating cardiomyocyte DNA methylation and gene expression. We used siRNA to knock down DNMT1 expression in primary cultures of mouse embryonic cardiomyocytes. Immunofluorescence staining and multielectrode array were, respectively, utilized to evaluate cardiomyocyte growth and electrophysiology. RNA sequencing (RNA‐Seq) and multiplex bisulfite sequencing were, respectively, performed to examine gene expression and promoter methylation. At 72 h post‐transfection, reduction of DNMT1 expression decreased the number and increased the size of embryonic cardiomyocytes. Beat frequency and the amplitude of field action potentials were decreased by DNMT1 siRNA. RNA‐Seq analysis identified 801 up‐regulated genes and 494 down‐regulated genes in the DNMT1 knockdown cells when compared to controls. Pathway analysis of the differentially expressed genes revealed pathways that were associated with cell death and survival, cell morphology, cardiac function, and cardiac disease. Alternative splicing analysis identified 929 differentially expressed exons, including 583 up‐regulated exons and 308 down‐regulated exons. Moreover, decreased methylation levels were found in the promoters of cardiac genes Myh6, Myh7, Myh7b, Tnnc1, Tnni3, Tnnt2, Nppa, Nppb, mef2c, mef2d, Camta2, Cdkn1A, and Cdkn1C. Of these 13 genes, 6 (Myh6, Tnnc1, Tnni3, Tnnt2, Nppa, Nppb) and 1 (Cdkn1C) had increased or decreased gene expression, respectively. Altogether, these data show that DNMT1 is important in embryonic cardiomyocytes by regulating DNA methylation, gene expression, gene splicing, and cell function.
Collapse
Affiliation(s)
- Xiefan Fang
- Department of Pediatrics, Child Health Research Institute, College of Medicine, University of Florida, Gainesville, FL, USA.,Charles River Laboratories, Inc., Reno, NV, USA
| | - Ryan Poulsen
- Department of Pediatrics, Child Health Research Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lu Zhao
- Charles River Laboratories, Inc., Reno, NV, USA
| | | | - Scott A Rivkees
- Department of Pediatrics, Child Health Research Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Christopher C Wendler
- Department of Pediatrics, Child Health Research Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
7
|
Wong KK. DNMT1 as a therapeutic target in pancreatic cancer: mechanisms and clinical implications. Cell Oncol (Dordr) 2020; 43:779-792. [PMID: 32504382 DOI: 10.1007/s13402-020-00526-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/09/2020] [Accepted: 04/18/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic cancer or pancreatic ductal adenocarcinoma (PDAC) is one of the most devastating cancer types with a 5-year survival rate of only 9%. PDAC is one of the leading causes of cancer-related deaths in both genders. Epigenetic alterations may lead to the suppression of tumor suppressor genes, and DNA methylation is a predominant epigenetic modification. DNA methyltransferase 1 (DNMT1) is required for maintaining patterns of DNA methylation during cellular replication. Accumulating evidence has implicated the oncogenic roles of DNMT1 in various malignancies including PDACs. CONCLUSIONS Herein, the expression profiles, oncogenic roles, regulators and inhibitors of DNMT1 in PDACs are presented and discussed. DNMT1 is overexpressed in PDAC cases compared with non-cancerous pancreatic ducts, and its expression gradually increases from pre-neoplastic lesions to PDACs. DNMT1 plays oncogenic roles in suppressing PDAC cell differentiation and in promoting their proliferation, migration and invasion, as well as in induction of the self-renewal capacity of PDAC cancer stem cells. These effects are achieved via promoter hypermethylation of tumor suppressor genes, including cyclin-dependent kinase inhibitors (e.g., p14, p15, p16, p21 and p27), suppressors of epithelial-mesenchymal transition (e.g., E-cadherin) and tumor suppressor miRNAs (e.g., miR-148a, miR-152 and miR-17-92 cluster). Pre-clinical investigations have shown the potency of novel non-nucleoside DNMT1 inhibitors against PDAC cells. Finally, phase I/II clinical trials of DNMT1 inhibitors (azacitidine, decitabine and guadecitabine) in PDAC patients are currently underway, where these inhibitors have the potential to sensitize PDACs to chemotherapy and immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
8
|
Network Pharmacology-Based Prediction and Verification of the Targets and Mechanism for Panax Notoginseng Saponins against Coronary Heart Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:6503752. [PMID: 31354855 PMCID: PMC6636530 DOI: 10.1155/2019/6503752] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/08/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022]
Abstract
Coronary heart disease (CHD) is the worldwide leading cause for cardiovascular death. Panax notoginseng saponin (PNS), which is the main bioactive compound of panax notoginseng, has been generally accepted to exert a remarkable effect on CHD for a long time. However, to reveal the underlying treatment target and corresponding mechanism of PNS against CHD is still a substantial challenge. In this work, the targets and mechanism of PNS against CHD were successfully achieved by pharmacology-based prediction and experimental verification. 36 common targets were screened out through integrating the gene expression profile of CHD and the chemical-protein data of PNS. Then, two key nodes were further selected for verification by experiment after analyzing GO function, KEGG pathway, coexpression, and topology analysis. Results showed that PNS has protected the human umbilical vein endothelial cells from H2O2-induced oxidative stress by inhibiting early cell apoptosis via upregulating VEGFA mRNA expression. Therefore, our research has successfully pointed out one treatment target and apoptotic inhibition caused by PNS with method of integrating bioinformatics prediction and experimental verification, which has partially explained the pharmacological mechanism of PNS against CHD.
Collapse
|
9
|
Chen YS, Chiu YH, Li YS, Lin EY, Hsieh DK, Lee CH, Huang MH, Chuang HM, Lin SZ, Harn HJ, Chiou TW. Integration of PEG 400 into a self-nanoemulsifying drug delivery system improves drug loading capacity and nasal mucosa permeability and prolongs the survival of rats with malignant brain tumors. Int J Nanomedicine 2019; 14:3601-3613. [PMID: 31190814 PMCID: PMC6530554 DOI: 10.2147/ijn.s193617] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/21/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction: Kolliphor® EL (K-EL) is among the most useful surfactants in the preparation of emulsions. However, it is associated with low hydrophobic drug loading in the resulting emulsified formulation. Methods: In this study, a formulation for intranasal administration of butylidenephthalide (Bdph), a candidate drug against glioblastoma (GBM), was prepared. Physical characteristics of the formulation such as particle size, zeta potential, conductivity, and viscosity were assessed, as well as its cytotoxicity and permeability, in order to optimize the formulation and improve its drug loading capacity. Results: The optimized formulation involved the integration of polyethylene glycol 400 (PEG 400) in K-EL to encapsulate Bdph dissolved in dimethyl sulfoxide (DMSO), and it exhibited higher drug loading capacity and drug solubility in water than the old formulation, which did not contain PEG 400. Incorporation of PEG 400 as a co-surfactant increased Bdph loading capacity to up to 50% (v/v), even in formulations using Kolliphor® HS 15 (K-HS15) as a surfactant, which is less compatible with Bdph than K-EL. The optimized Bdph formulation presented 5- and 2.5-fold higher permeability and cytotoxicity, respectively, in human GBM than stock Bdph. This could be attributed to the high drug loading capacity and the high polarity index due to DMSO, which increases the compatibility between the drug and the cell. Rats bearing a brain glioma treated with 160 mg/kg intranasal emulsified Bdph had a mean survival of 37 days, which is the same survival time achieved by treatment with 320 mg/kg stock Bdph. This implies that the optimized emulsified formulation required only half the Bdph dose to achieve an efficacy similar to that of stock Bdph in the treatment of animals with malignant brain tumor.
Collapse
Affiliation(s)
- Yu-Shuan Chen
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China.,Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China.,Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China
| | - Yu-Han Chiu
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China
| | - Yuan-Sheng Li
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China
| | - En-Yi Lin
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China.,Department of Chemistry, National Dong Hwa University, Hualien, Taiwan, Republic of China
| | - Dean-Kuo Hsieh
- Department and Graduate Institute of Applied Chemistry, Chaoyang University of Technology, Taichung, Taiwan, Republic of China
| | - Chia-Hung Lee
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China
| | - Mao-Hsuan Huang
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
| | - Hong-Meng Chuang
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China.,Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China
| | - Horng-Jyh Harn
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, Republic of China.,Department of Pathology, Hualien Tzu Chi Hospital, Tzu Chi University, Hualien, Taiwan, Republic of China
| | - Tzyy-Wen Chiou
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, Republic of China
| |
Collapse
|