Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Nanotechnology ; 35(46)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39146954

ABSTRACT

Triple-negative breast cancer (TNBC) is considered the most aggressive form of breast cancer owing to the negative expression of targetable bioreceptors. Epithelial to mesenchymal transition (EMT) associated with metastatic abilities is its critical feature. As an attempt to target TNBC, nanotechnology was utilised to augment the effects of drug repurposing. Concerning that, a combination therapeutic module was structured with one of the aspects being a repurposed antihistamine, promethazine hydrochloride loaded PLGA nanoparticles. The as-synthesized nanoparticles were 217 nm in size and fluoresced at 522 nm, rendering them suitable for theranostic applications too. The second feature of the module was a common histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), used as a form of pre-treatment. Experimental studies demonstrated efficient cellular internalisation and significant innate anti-proliferative potential. The use of SAHA sensitised the cells to the drug loaded nanoparticle treatment. Mechanistic studies showed increase in ROS generation, mitochondrial dysfunction followed by apoptosis. Investigations into protein expression also revealed reduction of mesenchymal proteins like vimentin by 1.90 fold; while increase in epithelial marker like E-Cadherin by 1.42 fold, thus indicating an altered EMT dynamics. Further findings also provided better insight into the benefits of SAHA potentiated targeting of tumor spheroids that mimic solid tumors of TNBC. Thus, this study paves the avenue to a more rational translational validation of combining nanotherapeutics with drug repurposing.


Subject(s)
Apoptosis , Drug Repositioning , Epithelial-Mesenchymal Transition , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , Promethazine , Triple Negative Breast Neoplasms , Vorinostat , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Vorinostat/pharmacology , Vorinostat/chemistry , Nanoparticles/chemistry , Cell Line, Tumor , Promethazine/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Apoptosis/drug effects , Female , Reactive Oxygen Species/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry , Drug Synergism , Drug Carriers/chemistry
2.
Int J Pharm ; 661: 124422, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38977163

ABSTRACT

The challenges posed by intractable relapse and metastasis in cancer treatment have led to the development of various forms of photodynamic therapy (PDT). However, traditional drug delivery systems, such as virus vectors, liposomes, and polymers, often suffer from issues like desynchronized drug release, carrier instability, and drug leakage during circulation. To address these problems, we have developed a dual-prodrug nanogel (PVBN) consisting of Pyro (Pyropheophorbide a) and SAHA (Vorinostat) bound to BSA (Bovine Serum Albumin), which facilitates synchronous and spontaneous drug release in situ within the lysosome. Detailed results indicate that PVBN-treated tumor cells exhibit elevated levels of ROS and Acetyl-H3, leading to necrosis, apoptosis, and cell cycle arrest, with PDT playing a dominant role in the synergistic therapeutic effect. Furthermore, the anti-tumor efficacy of PVBN was validated in melanoma-bearing mice, where it significantly inhibited tumor growth and pulmonary metastasis. Overall, our dual-prodrug nanogel, formed by the binding of SAHA and Pyro to BSA and releasing drugs within the lysosome, represents a novel and promising strategy for enhancing the clinical efficacy of photochemotherapy.


Subject(s)
Chlorophyll , Nanogels , Photochemotherapy , Prodrugs , Serum Albumin, Bovine , Vorinostat , Animals , Vorinostat/administration & dosage , Vorinostat/pharmacology , Vorinostat/chemistry , Photochemotherapy/methods , Chlorophyll/analogs & derivatives , Chlorophyll/chemistry , Chlorophyll/administration & dosage , Chlorophyll/pharmacology , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/administration & dosage , Cell Line, Tumor , Nanogels/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry , Mice , Apoptosis/drug effects , Drug Liberation , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Humans , Reactive Oxygen Species/metabolism , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Mice, Inbred C57BL , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Melanoma, Experimental/drug therapy , Polyethyleneimine/chemistry
3.
Chemistry ; 30(46): e202401724, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-38853639

ABSTRACT

The clinical use of many potent anticancer agents is limited by their non-selective toxicity to healthy tissue. One of these examples is vorinostat (SAHA), a pan histone deacetylase inhibitor, which shows high cytotoxicity with limited discrimination for cancerous over healthy cells. In an attempt to improve tumor selectivity, we exploited the properties of cobalt(III) as a redox-active metal center through stabilization with cyclen and cyclam tetraazamacrocycles, masking the anticancer activity of SAHA and other hydroxamic acid derivatives to allow for the complex to reach the hypoxic microenvironment of the tumor. Biological assays demonstrated the desired low in vitro anticancer activity of the complexes, suggesting effective masking of the activity of SAHA. Once in the tumor, the bioactive moiety may be released through the reduction of the CoIII center. Investigations revealed long-term stability of the complexes, with cyclic voltammetry and chemical reduction experiments supporting the design hypothesis of SAHA release through the reduction of the CoIII prodrug. The results highlight the potential for further developing this complex class as novel anticancer agents by masking the high cytotoxicity of a given drug, however, the cellular uptake needs to be improved.


Subject(s)
Antineoplastic Agents , Cobalt , Coordination Complexes , Hydroxamic Acids , Oxidation-Reduction , Vorinostat , Cobalt/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Vorinostat/chemistry , Vorinostat/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Cell Line, Tumor , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology
4.
J Pharm Sci ; 113(8): 2584-2594, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38801974

ABSTRACT

Vorinostat (VST) is a chemotherapeutic agent administrated for various types of cancers. However, it suffers from side effects and chemoresistance that reduce its application. Different nanoniosomes comprised Span 20, 60, 65 and 80 were prepared by the thin film hydration method and loaded with VST. The nanoniosomes were physicochemically characterized using particle size analysis and field emission scanning electron microscopy. The best formulation that was prepared using Span 65 (VST-NN-S65) included vesicle size of 127 nm with a narrow size distribution. VST-NN-S65 had an entrapment efficiency and loading capacity of 81.3 ± 5.1 and 32.0 ± 3.9 %, respectively. Drug release rate measurements showed that 90 % of VST was liberated within 1 h. Cytotoxicity assessments of VST-NN-S65 in HeLa and MCF7 cells indicated significant improvement in the effectiveness of VST, compared to the VST suspension. For VST-NN-S65, IC50 values of 26.3 and 6.6 µg mL-1 were obtained for HeLa and MCF7 cell lines, respectively. In situ apoptosis detection by the TUNEL assay revealed that apoptosis mainly occurred in the cell lines.


Subject(s)
Antineoplastic Agents , Apoptosis , Drug Carriers , Hydroxamic Acids , Liposomes , Particle Size , Vorinostat , Humans , Vorinostat/pharmacology , Vorinostat/administration & dosage , Vorinostat/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , HeLa Cells , MCF-7 Cells , Apoptosis/drug effects , Hydroxamic Acids/chemistry , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , Drug Carriers/chemistry , Nanoparticles/chemistry , Drug Liberation , Cell Survival/drug effects
5.
Eur J Med Chem ; 229: 114058, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34954595

ABSTRACT

In view of histone deacetylases (HDACs) as a promising target for cancer therapy, a series of phthalazino[1,2-b]-quinazolinone units were hybrided with ortho-aminoanilide or hydroxamic acid to serve as multi-target HDAC inhibitors for the treatment of solid tumors. Among the target compounds, 8h possessed nano-molar IC50 values toward the tested cancer cells and HDAC subtypes, which was more potent than the HDAC inhibitor SAHA (vorinostat). Mechanism study revealed that compound 8h could suppress the HepG2 cell proliferation via prompting the acetylation of histone 3 (H3) and α-tubulin, and activating the p53 signal pathway as designed. In addition, compound 8h exhibited much stronger in vivo antitumor efficacy than SAHA in the HepG2 xenograft tumor model with negligible toxicity. As a novel multi-target HDAC inhibitor, compound 8h deserves further development as a potential anticancer agent.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Histone Deacetylase Inhibitors , Histone Deacetylases , Liver Neoplasms , Phthalazines , Quinazolinones , Tumor Suppressor Protein p53 , Animals , Humans , Male , Mice , Acetylation , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Hydroxamic Acids/chemistry , Liver Neoplasms/drug therapy , Molecular Docking Simulation , Phthalazines/chemistry , Quinazolinones/chemical synthesis , Quinazolinones/pharmacology , Signal Transduction , Structure-Activity Relationship , Tubulin/metabolism , Tumor Suppressor Protein p53/metabolism , Vorinostat/chemistry
6.
Bioorg Chem ; 115: 104868, 2021 10.
Article in English | MEDLINE | ID: mdl-34523507

ABSTRACT

Reviewed herein are key research accomplishments of Professor Ronald Charles D. Breslow (1931-2017) throughout his more than 60 year research career. These accomplishments span a wide range of topics, most notably physical organic chemistry, medicinal chemistry, and bioorganic chemistry. These topics are reviewed, as are topics of molecular electronics and origin of chirality, which combine to make up the bulk of this review. Also reviewed briefly are Breslow's contributions to the broader chemistry profession, including his work for the American Chemical Society and his work promoting gender equity. Throughout the article, efforts are made to put Breslow's accomplishments in the context of other work being done at the time, as well as to include subsequent iterations and elaborations of the research.


Subject(s)
Chemistry, Pharmaceutical/history , Amino Acids/chemistry , Catalysis , Cyclodextrins/chemistry , History, 20th Century , Humans , Stereoisomerism , Thiamine/chemistry , Vorinostat/chemistry
7.
Pharmacol Res ; 171: 105787, 2021 09.
Article in English | MEDLINE | ID: mdl-34314859

ABSTRACT

We adopted a novel strategy by combining histone deacetylase (HDAC) inhibitors with traditional chemotherapeutics to treat solid tumors. However, chemotherapeutics often have a narrow therapeutic index and need multiple administrations with undesired side effects that lead to the intolerance. To reduce the non-specificity of chemotherapeutics, targeted therapy was introduced to restrict such agents in the tumor with minimum effects on other tissues. We developed bioinspired artificial exosomes (AE), which enabled to deliver chemotherapeutics to the tumors effectively after systemic administration. AE were produced by incorporating membrane proteins from cancer cells into phospholipid liposomes that mimicked the plasma membrane. The synthesized AE were used for the delivery of broad-spectrum chemotherapeutic doxorubicin (DOX) and vorinostat (SAHA), an epigenetic inhibitor. The combination of DOX and SAHA showed synergistic effects on suppressing non-small cell lung cancer cells and xenograft tumors without apparent adverse effects. AE facilitated the delivery of drugs to tumor tissue and extended the retention time of drugs within tumors. Taken together, these studies suggest that the bioengineered artificial exosomes may serve as novel delivery strategy for chemotherapeutics to treat non-small cell lung cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Exosomes , Histone Deacetylase Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , Vorinostat/administration & dosage , Animals , Antineoplastic Agents/chemistry , Cell Line , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Liberation , Epigenesis, Genetic , Humans , Lung Neoplasms/pathology , Mice, Inbred BALB C , Tumor Burden/drug effects , Vorinostat/chemistry
8.
Bioorg Chem ; 114: 105132, 2021 09.
Article in English | MEDLINE | ID: mdl-34229198

ABSTRACT

Hydroxamic acid derivatives constitute an interesting novel class of antitumor agents. Three of them, including vorinostat, are approved drugs for the treatment of malignancies, while several others are currently under clinical trials. In this work, we present new vorinostat analogs containing the benzoxazole ring as the cap group and various linkers. The benzoxazole-based analogs were synthesized starting either from 2-aminobenzoxazole, through conventional coupling, or from benzoxazole, through a metal-free oxidative amination. All the synthesized compounds were evaluated for their antiproliferative activity on three diverse human cancer cell lines (A549, Caco-2 and SF268), in comparison to vorinostat. Compound 12 (GK601), carrying a benzoxazole ring replacement for the phenyl ring of vorinostat, was the most potent inhibitor of the growth of three cell lines (IC50 1.2-2.1 µΜ), similar in potency to vorinostat. Compound 12 also inhibited human HDAC1, HDAC2 and HDAC6 like vorinostat. This new analog also showed antiproliferative activity against two colon cancer cell lines genetically resembling pseudomyxoma peritonei (PMP), namely HCT116 GNAS R201C/+ and LS174T (IC50 0.6 and 1.4 µΜ, respectively) with potency comparable to vorinostat (IC50 1.1 and 2.1 µΜ, respectively).


Subject(s)
Antineoplastic Agents/pharmacology , Benzoxazoles/pharmacology , Vorinostat/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzoxazoles/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured , Vorinostat/chemical synthesis , Vorinostat/chemistry
9.
J Med Chem ; 64(11): 7468-7482, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34043359

ABSTRACT

Nowadays, simultaneous inhibition of multiple targets through drug combination is an important anticancer strategy owing to the complex mechanism behind tumorigenesis. Recent studies have demonstrated that the inhibition of histone deacetylases (HDACs) will lead to compensated activation of a notorious cancer-related drug target, signal transducer and activator of transcription 3 (STAT3), in breast cancer through a cascade, which probably limits the anti-proliferation effect of HDAC inhibitors in solid tumors. By incorporating the pharmacophore of the HDAC inhibitor SAHA (vorinostat) into the STAT3 inhibitor pterostilbene, a series of potent pterostilbene hydroxamic acid derivatives with dual-target inhibition activity were synthesized. An excellent hydroxamate derivate, compound 14, inhibited STAT3 (KD = 33 nM) and HDAC (IC50 = 23.15 nM) with robust potency in vitro. Compound 14 also showed potent anti-proliferation ability in vivo and in vitro. Our study provides the first STAT3 and HDAC dual-target inhibitor for further exploration.


Subject(s)
Antineoplastic Agents/chemistry , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Half-Life , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/chemistry , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Molecular Docking Simulation , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/antagonists & inhibitors , Stilbenes/chemistry , Stilbenes/metabolism , Structure-Activity Relationship , Vorinostat/chemistry , Vorinostat/metabolism
10.
Biol Chem ; 402(4): 501-512, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33938179

ABSTRACT

Triple-negative breast cancers (TNBC) that produce nitric oxide (NO) are more aggressive, and the expression of the inducible form of nitric oxide synthase (NOS2) is a negative prognostic indicator. In these studies, we set out to investigate potential therapeutic strategies to counter the tumor-permissive properties of NO. We found that exposure to NO increased proliferation of TNBC cells and that treatment with the histone deacetylase inhibitor Vorinostat (SAHA) prevented this proliferation. When histone acetylation was measured in response to NO and/or SAHA, NO significantly decreased acetylation on histone 3 lysine 9 (H3K9ac) and SAHA increased H3K9ac. If NO and SAHA were sequentially administered to cells (in either order), an increase in acetylation was observed in all cases. Mechanistic studies suggest that the "deacetylase" activity of NO does not involve S-nitrosothiols or soluble guanylyl cyclase activation. The observed decrease in histone acetylation by NO required the interaction of NO with cellular iron pools and may be an overriding effect of NO-mediated increases in histone methylation at the same lysine residues. Our data revealed a novel pathway interaction of Vorinostat and provides new insight in therapeutic strategy for aggressive TNBCs.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Nitric Oxide/metabolism , Triple Negative Breast Neoplasms/drug therapy , Vorinostat/pharmacology , Acetylation/drug effects , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors/chemistry , Humans , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Vorinostat/chemistry
11.
ACS Appl Mater Interfaces ; 13(5): 5999-6010, 2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33506682

ABSTRACT

Cellular FLIP (cFLIP) is a crucial player of apoptosis-regulated pathways that is frequently overexpressed in solid cancers. To inhibit c-FLIP, pre- and post-transcriptionally, a multifunctional nanoparticle (NP) was created to deliver cFLIP-specific small interfering RNA (siRNA) into cancer cells. Specifically, Vorinostat (Vor)-loaded mesoporous silica nanoparticles (MSN) were conjugated with polyethylenimine-biotin (PB), followed by electrostatically binding with cFLIP siRNA (Vor/siR@MSN-PB). To stabilize and prolong the circulation time of nanoparticles, a bialdehyde-modified poly(ethylene glycol) (PEG) was cross-linked onto the polyethylenimine (PEI) backbone via the formation of the imine linkage (Schiff base) (Vor/siR@MSN-PB-PEG). The Schiff base is highly stable at physiological pH 7.4 but labile under slightly acidic pH conditions. In the acidic tumor microenvironment (TME), the PEG outer layer could be rapidly cleaved, resulting in the switching of the nanoparticle surface charge to positive, which specifically enhances internalization of the NPs to the biotin-positive tumor cells. Our results demonstrated the successful preparation of Vor/siR@MSN-PB-PEG NPs, in which the siRNA was effectively protected in serum and regulated the expression of cFlip, post-transcriptionally. The presence of the PEG layer resulted in high tumor accumulation and high efficacy in tumor inhibition, which was a result of the efficient cFLIP suppression. Furthermore, in the low-dose regimen of Vorinostat-the pre-transcriptional cFLIP suppressor, treatment with Vor/siR@MSN-PB-PEG NPs was found to be safe with the treated mice, indicating a promising combination regimen for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/antagonists & inhibitors , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Nanoparticles/chemistry , RNA, Small Interfering/pharmacology , Vorinostat/pharmacology , Animals , Antineoplastic Agents/chemistry , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Particle Size , Polyethylene Glycols/chemistry , RNA, Small Interfering/chemistry , Surface Properties , Vorinostat/chemistry
12.
Anticancer Agents Med Chem ; 21(10): 1301-1308, 2021.
Article in English | MEDLINE | ID: mdl-33023457

ABSTRACT

BACKGROUND AND OBJECTIVE: Curcumin is an effective anti-cancer agent used in thyroid cancer treatments. However, its use in clinical applications is limited due to low solubility and bioavailability. In this study, a novel combination strategy was applied by combining curcumin with Suberoylanilide Hydroxamic Acid (SAHA) to increase both bioavailability of curcumin and the efficiency of SAHA, which have limited efficiency when used alone. METHODS: MTT assay was used to determine the cell viability of B-CPAP cells upon treatment with SAHA, curcumin and their combinations. Synergistic interactions between two agents were analyzed by Calcusyn software. Apoptosis and cell cycle assays were measured by flow cytometry. Expressions of apoptotic and cell cyclerelated proteins (PARP, P21/CDKN1A/WAF1, P27/KIP1) were examined by western blot analysis. Broth microdilution assay was performed to determine Minimum Inhibitory Concentration (MIC) values against S. aureus. RESULTS: Based on MTT assay, IC50 values for SAHA and curcumin were determined as 0.91µM and 20.97µM, respectively. The combination index CI value was determined as 0.891 in B-CPAP cells, which demonstrate synergistic activity. The apoptotic effect was achieved by combination treatment (51.85%) on B-CPAP cells using half of the dose required for SAHA and curcumin alone. Combination treatment showed a significant increase in the percentage of B-CPAP cells in the S-phase due to cell arrest. Cleaved-PARP, P21/CDKN1A/ WAF1 and P27/KIP1 protein expressions were upregulated. Curcumin was found to have better anti-microbial activity than SAHA as having a lower MIC value, and checkerboard synergy analysis revealed that the two compounds co-operate synergistically for the in vitro killing of S. aureus. CONCLUSION: In the present study, synergistic combinations of SAHA and curcumin were shown to have both anti-cancer and antibacterial activities that would provide a novel thyroid cancer treatment strategy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Biological Products/pharmacology , Curcumin/pharmacology , Flavonoids/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Vorinostat/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcumin/chemical synthesis , Curcumin/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flavonoids/chemical synthesis , Flavonoids/chemistry , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Humans , Microbial Sensitivity Tests , Molecular Structure , Staphylococcus aureus/drug effects , Structure-Activity Relationship , Tumor Cells, Cultured , Vorinostat/chemical synthesis , Vorinostat/chemistry
13.
Molecules ; 25(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33227941

ABSTRACT

The stabilisation of G-quadruplexes (G4s) by small-molecule compounds is an effective approach for causing cell growth arrest, followed by cell death. Some of these compounds are currently being developed for the treatment of human cancers. We have previously developed a substituted naphthalene diimide G4-binding molecule (CM03) with selective potency for pancreatic cancer cells, including gemcitabine-resistant cells. We report here that CM03 and the histone deacetylase (HDAC) inhibitor SAHA (suberanilohydroxamic acid) have synergistic effects at concentrations close to and below their individual GI50 values, in both gemcitabine-sensitive and resistant pancreatic cancer cell lines. Immunoblot analysis showed elevated levels of γ-H2AX and cleaved PARP proteins upon drug combination treatment, indicating increased levels of DNA damage (double-strand break events: DSBs) and apoptosis induction, respectively. We propose that the mechanism of synergy involves SAHA relaxing condensed chromatin, resulting in higher levels of G4 formation. In turn, CM03 can stabilise a greater number of G4s, leading to the downregulation of more G4-containing genes as well as a higher incidence of DSBs due to torsional strain on DNA and chromatin structure.


Subject(s)
Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , G-Quadruplexes , Histone Deacetylase Inhibitors/therapeutic use , Pancreatic Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Vorinostat/therapeutic use , Cell Line, Tumor , DNA Damage , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Synergism , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Pancreatic Neoplasms/pathology , Vorinostat/chemistry , Vorinostat/pharmacology , Gemcitabine
14.
Biomark Med ; 14(12): 1069-1084, 2020 08.
Article in English | MEDLINE | ID: mdl-32969243

ABSTRACT

Aim: To explore the mechanism of gastric carcinogenesis by mining potential hub genes and to search for promising small-molecular compounds for gastric cancer (GC). Materials & methods: The microarray datasets were downloaded from Gene Expression Omnibus database and the genes and compounds were analyzed by bioinformatics-related tools and software. Results: Six hub genes (MKI67, PLK1, COL1A1, TPX2, COL1A2 and SPP1) related to the prognosis of GC were confirmed to be upregulated in GC and their high expression was correlated with poor overall survival rate in GC patients. In addition, eight candidate compounds with potential anti-GC activity were identified, among which resveratrol was closely correlated with six hub genes. Conclusion: Six hub genes identified in the present study may contribute to a more comprehensive understanding of the mechanism of gastric carcinogenesis and the predicted potential of resveratrol may provide valuable clues for the future development of targeted anti-GC inhibitors.


Subject(s)
Gene Expression Profiling , Genes, Neoplasm , Neoplasm Proteins/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Amiodarone/chemistry , Cell Cycle Proteins/genetics , Clomipramine/chemistry , Collagen Type I/genetics , Databases, Genetic , Datasets as Topic , Gene Expression Regulation, Neoplastic , Humans , Ki-67 Antigen/genetics , Levallorphan/chemistry , Microtubule-Associated Proteins/genetics , Osteopontin/genetics , Piroxicam/chemistry , Procaine/chemistry , Procaine/pharmacology , Procaine/therapeutic use , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Resveratrol/chemistry , Resveratrol/pharmacology , Small Molecule Libraries/therapeutic use , Ursodeoxycholic Acid/chemistry , Vorinostat/chemistry , Polo-Like Kinase 1
15.
Bioorg Med Chem ; 28(17): 115639, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32773090

ABSTRACT

Histone deacetylases (HDACs) have been found to be biomarkers of cancers and the corresponding inhibitors have attracted much attention these years. Herein we reported a near-infrared fluorescent HDAC inhibitor based on vorinostat (SAHA) and a NIR fluorophore. This newly designed inhibitor showed similar inhibitory activity to SAHA against three HDAC isoforms (HDAC1, 3, 6). The western blot assay showed significant difference in compared with the negative group. When used as probe for further kinematic imaging, Probe 1 showed enhanced retention in tumor cells and the potential of HDAC inhibitors in drug delivery was firstly brought out. The cytotoxicity assay showed Probe 1 had some anti-proliferation activities with corresponding IC50 values of 9.20 ± 0.96 µM on Hela cells and 5.91 ± 0.57 µM on MDA-MB-231 cells. These results indicated that Probe 1 could be used as a potential NIR fluorescent in the study of HDAC inhibitors and lead compound for the development of visible drugs.


Subject(s)
Drug Design , Fluorescent Dyes/chemistry , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , HeLa Cells , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Microscopy, Fluorescence , Vorinostat/chemistry
16.
Nat Commun ; 11(1): 3841, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737323

ABSTRACT

Histone deacetylases (HDACs) are key enzymes in epigenetics and important drug targets in cancer biology. Whilst it has been established that HDACs regulate many cellular processes, far less is known about the regulation of these enzymes themselves. Here, we show that HDAC8 is allosterically regulated by shifts in populations between exchanging states. An inactive state is identified, which is stabilised by a range of mutations and resembles a sparsely-populated state in equilibrium with active HDAC8. Computational models show that the inactive and active states differ by small changes in a regulatory region that extends up to 28 Å from the active site. The regulatory allosteric region identified here in HDAC8 corresponds to regions in other class I HDACs known to bind regulators, thus suggesting a general mechanism. The presented results pave the way for the development of allosteric HDAC inhibitors and regulators to improve the therapy for several disease states.


Subject(s)
Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/chemistry , Hydroxamic Acids/chemistry , Indoles/chemistry , Repressor Proteins/chemistry , Vorinostat/chemistry , Allosteric Regulation , Allosteric Site , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Enzyme Activation , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/metabolism , Indoles/metabolism , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Substrate Specificity , Thermodynamics , Vorinostat/metabolism
17.
Sci Rep ; 10(1): 12864, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32733053

ABSTRACT

Pharmacological modulation of class I histone deacetylases (HDAC) has been evaluated as a therapeutic strategy for pulmonary hypertension (PH) in experimental models of PH. However, information of their expression, regulation and transcriptional targets in human PH and the therapeutic potential of isoform-selective enzyme modulation are lacking. Comprehensive analysis of expression and regulation of class I HDACs (HDAC1, HDAC2, HDAC3 and HDAC8) was performed in cardiopulmonary tissues and adventitial fibroblasts isolated from pulmonary arteries (PAAF) of idiopathic pulmonary arterial hypertension (IPAH) patients and healthy donors. Cellular functions and transcriptional targets of HDAC enzymes were investigated. Therapeutic effects of pan-HDAC (Vorinostat), class-selective (VPA) and isoform-selective (CAY10398, Romidepsin, PCI34051) HDAC inhibitors were evaluated ex vivo (IPAH-PAAF, IPAH-PASMC) and in vivo (rat chronic hypoxia-induced PH and zebrafish angiogenesis). Our screening identifies dysregulation of class I HDAC isoforms in IPAH. Particularly, HDAC1 and HDAC8 were consistently increased in IPAH-PAs and IPAH-PAAFs, whereas HDAC2 and HDAC8 showed predominant localization with ACTA2-expressing cells in extensively remodeled IPAH-PAs. Hypoxia not only significantly modulated protein levels of deacetylase (HDAC8), but also significantly caused dynamic changes in the global histone lysine acetylation levels (H3K4ac, H3K9/K14ac and H3K27ac). Importantly, isoform-specific RNA-interference revealed that HDAC isoforms regulate distinct subset of transcriptome in IPAH-PAAFs. Reduced transcript levels of KLF2 in IPAH-PAAFs was augmented by HDAC8 siRNA and HDAC inhibitors, which also attenuated IPAH-associated hyperproliferation and apoptosis-resistance ex vivo, and mitigated chronic hypoxia-induced established PH in vivo, at variable degree. Class I HDAC isoforms are significantly dysregulated in human PAH. Isoform-selective HDAC inhibition is a viable approach to circumvent off-target effects.


Subject(s)
Histone Deacetylases/therapeutic use , Hypertension, Pulmonary/drug therapy , Animals , Cells, Cultured , Depsipeptides/chemistry , Depsipeptides/pharmacology , Depsipeptides/therapeutic use , Histone Deacetylases/chemistry , Histone Deacetylases/pharmacology , Humans , In Vitro Techniques , Isoenzymes , Rats , Structure-Activity Relationship , Transcriptome/drug effects , Vorinostat/chemistry , Vorinostat/pharmacology , Vorinostat/therapeutic use , Zebrafish
18.
Chem Commun (Camb) ; 56(56): 7753-7756, 2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32613216

ABSTRACT

We utilized albumin as a reducing agent to establish novel copper-based and pH-sensitive nanocarrier CuNPs with abundant Cu+, which can encapsulate histone deacetylase (HDAC) inhibitor vorinostat to form uniform and stable nanomedicine V-CuNPs for synergistic chromatin remodelling and chemodynamic therapy.


Subject(s)
Copper/chemistry , Hydrogen-Ion Concentration , Metal Nanoparticles/chemistry , Molecular Targeted Therapy/methods , Vorinostat/pharmacology , Animals , Cell Line, Tumor , Coordination Complexes/chemistry , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Vorinostat/chemistry , Vorinostat/pharmacokinetics
19.
J Biomol Struct Dyn ; 38(2): 597-603, 2020 02.
Article in English | MEDLINE | ID: mdl-30744531

ABSTRACT

AbbreviationsSAHAsuberoylanilide hydroxamic acidEhHDACHistone Deacetylase from Entamoeba histolyticaRgRadius of gyrationRMSDroot-mean-square deviationRMSFroot-mean-square fluctuationMDSmolecular dynamics simulationVMDVisual Molecular DynamicsNAMDNanoscale Molecular DynamicsPBCperiodic boundary conditionsPMEParticle Mesh Ewald3Dthree-dimensionalCαalpha carbonFDAFood and Drug AdministrationnsnanosecondsGPU CUDAGraphics Processing Unit Compute Unified Device ArchitectureCommunicated by Ramaswamy H. Sarma.


Subject(s)
Amebiasis/drug therapy , Amebiasis/parasitology , Entamoeba histolytica/physiology , Metronidazole/therapeutic use , Vorinostat/therapeutic use , Entamoeba histolytica/drug effects , Entamoeba histolytica/enzymology , Histone Deacetylases/chemistry , Metronidazole/chemistry , Metronidazole/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Phylogeny , Structural Homology, Protein , Trophozoites/drug effects , Trophozoites/physiology , Vorinostat/chemistry , Vorinostat/pharmacology
20.
Biomater Sci ; 7(12): 5301-5311, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31616866

ABSTRACT

Nanocarriers with high local curvatures hold a great potential of inducing effective penetration of intracellular barriers and cytosolic delivery of membrane-impermeable drugs. However, the fine control of the sharp edges and their morphological effects inside cells remains largely unexplored. Herein, a nanocarrier system of hybrid mesoporous nanorods with six-arm star-shaped end faces and groove-patterned lateral faces was developed to maximize surface regions with high local curvatures for enhancing membrane destabilization. Specifically, twisted (right-handed) nanorods (TNR, diameter ∼120, aspect ratio 4-5) with a hexagon cross-section from a templated synthesis were modified by amino groups to promote surface coating of a wet-adhesive polymer, i.e. polydopamine (PDA). An edge-preferential deposition of PDA by local curvature effects led to the protective etching of silica, and in turn, the formation of nanorods with varying groove depths at different volumes of the aqueous coating solution. Finally, branched polyethylene imine (PEI) was grafted on the exterior surface of the nanorods for enhancing the dispersity and cellular uptake rate. As verified by elaborate in vitro investigations, the configuration of nanorods with the sharpest edges/deepest grooves can be rotated to a lying-down/upright mode in order to minimize/maximize the membrane tension during the interaction with membranes, which consequently resulted in highly efficient lysosomal escape despite the relatively lower uptake degree. The successful delivery of vorinostat (SAHA, a FDA-approved histone deacetylase inhibitor) and inhibition of cancer cells demonstrated the attractive ability of the nanocarriers in drug delivery.


Subject(s)
Cytosol/chemistry , Doxorubicin/pharmacology , Vorinostat/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Delivery Systems/methods , Humans , Indoles/chemistry , MCF-7 Cells , Nanotubes , Polyethyleneimine/chemistry , Polymers/chemistry , Porosity , Vorinostat/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL