Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Biomater Adv ; 139: 213038, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35908474

ABSTRACT

The repurposing of old drugs can reduce the cost of drug development and speed up the availability of drugs for clinical use. Disulfiram (DSF) is an approved drug for alcohol abuse. In recent years, it has been established that DSF exerts an antitumor effect via targeted inhibition of ALDH1+ cancer stem cells (CSCs). However, due to its metal ion dependence, easy hydrolysis and low availability, the clinical application of DSF is limited. Previous studies have also shown that Zn2+ can inhibit CSCs. Accordingly, we developed a novel metal organic framework (IRMOF3)-Zn2+, and DSF was incorporated in the IRMOF3. Folic acid (FA) was subsequently loaded on the surface yielding IRMOF3 (IRMOF3-DSF-FA) for targeted therapy of tumors. The nanoscale IRMOF3-DSF-FA exhibited a high loading capacity, good biocompatibility and strong cell uptake capacity, which could provide metal ions, target tumor tissues and inhibit ALDH1+ CSCs. In vivo experiments showed that IRMOF3-DSF-FA could significantly inhibit the growth of CSCs and tumors, with no significant vital organ damage during treatment. Accordingly, IRMOF3-DSF-FA has great prospects for application as a DSF carrier, opening new horizons for targeted therapy of oral cancer.


Subject(s)
Antineoplastic Agents , Mouth Neoplasms , Nanoparticles , Neoplastic Stem Cells , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Disulfiram/pharmacology , Folic Acid/pharmacology , Humans , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Nanoparticles/metabolism , Nanoparticles/therapeutic use , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Organometallic Compounds/pharmacology , Retinal Dehydrogenase/antagonists & inhibitors , Zinc/pharmacology
2.
Molecules ; 27(2)2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35056791

ABSTRACT

Aldehyde dehydrogenase-1a1 (ALDH1a1), the enzyme responsible for the oxidation of retinal into retinoic acid, represents a key therapeutic target for the treatment of debilitating disorders such as cancer, obesity, and inflammation. Drugs that can inhibit ALDH1a1 include disulfiram, an FDA-approved drug to treat chronic alcoholism. Disulfiram, by carbamylation of the catalytic cysteines, irreversibly inhibits ALDH1a1 and ALDH2. The latter is the isozyme responsible for important physiological processes such as the second stage of alcohol metabolism. Given the fact that ALDH1a1 has a larger substrate tunnel than that in ALDH2, replacing disulfiram ethyl groups with larger motifs will yield selective ALDH1a1 inhibitors. We report herein the synthesis of new inhibitors of ALDH1a1 where (hetero)aromatic rings were introduced into the structure of disulfiram. Most of the developed compounds retained the anti-ALDH1a1 activity of disulfiram; however, they were completely devoid of inhibitory activity against ALDH2.


Subject(s)
Acetaldehyde Dehydrogenase Inhibitors/chemistry , Acetaldehyde Dehydrogenase Inhibitors/pharmacology , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Disulfiram/chemistry , Disulfiram/pharmacology , Retinal Dehydrogenase/antagonists & inhibitors , Acetaldehyde Dehydrogenase Inhibitors/chemical synthesis , Acetaldehyde Dehydrogenase Inhibitors/metabolism , Aldehyde Dehydrogenase 1 Family/chemistry , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase, Mitochondrial/antagonists & inhibitors , Aldehyde Dehydrogenase, Mitochondrial/chemistry , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Disulfiram/analogs & derivatives , Disulfiram/chemical synthesis , Humans , Molecular Docking Simulation , Recombinant Proteins/metabolism , Retinal Dehydrogenase/chemistry , Retinal Dehydrogenase/metabolism
3.
Int J Mol Sci ; 22(21)2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34769507

ABSTRACT

Activating molecule in Beclin-1-regulated autophagy (AMBRA1), a negative regulator of tumorigenesis, is a substrate receptor of the ubiquitin conjugation system. ALDH1B1, an aldehyde dehydrogenase, is a cancer stem cell (CSC) marker that is required for carcinogenesis via upregulation of the ß-catenin pathway. Although accumulating evidence suggests a role for ubiquitination in the regulation of CSC markers, the ubiquitination-mediated regulation of ALDH1B1 has not been unraveled. While proteome analysis has suggested that AMBRA1 and ALDH1B1 can interact, their interaction has not been validated. Here, we show that AMBRA1 is a negative regulator of ALDH1B1. The expression of ALDH1B1-regulated genes, including PTEN, CTNNB1 (ß-catenin), and CSC-related ß-catenin target genes, is inversely regulated by AMBRA1, suggesting a negative regulatory role of AMBRA1 in the expression of ALDH1B1-regulated genes. We found that the K27- and K33-linked ubiquitination of ALDH1B1 is mediated via the cooperation of AMBRA1 with other E3 ligases, such as TRAF6. Importantly, ubiquitination site mapping revealed that K506, K511, and K515 are important for the K27-linked ubiquitination of ALDH1B1, while K33-linked ubiquitination occurs at K506. A ubiquitination-defective mutant of ALDH1B1 increased the self-association ability of ALDH1B1, suggesting a negative correlation between the ubiquitination and self-association of ALDH1B1. Together, our findings indicate that ALDH1B1 is negatively regulated by AMBRA1-mediated noncanonical ubiquitination.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Neoplastic Stem Cells/pathology , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin-Protein Ligases/metabolism , beta Catenin/metabolism , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Aldehyde Dehydrogenase, Mitochondrial/antagonists & inhibitors , Cell Line , Cell Line, Tumor , Humans , Neoplastic Stem Cells/metabolism , Ubiquitination , Wnt Signaling Pathway
4.
Bioorg Med Chem ; 46: 116352, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34403955

ABSTRACT

Aldehyde dehydrogenase 1A1 (ALDH1A1) plays vital physiological and toxicological functions in many areas, such as CNS, inflammation, metabolic disorders, and cancers. Overexpression of ALDH1A1 has been disclosed to play an important role in obesity, diabetes and other diseases, indicating the potential need for the identification and development of small molecule ALDH1A1 inhibitors. Herein, a series of benzimidazole derivatives was designed, synthesized and evaluated. Among them, compounds 21, 27, 29, 61 and 65 exhibited excellent inhibitory activity against ALDH1A1 with IC50 values in the low micromolar range and high selectivity over ALDH1A2, ALDH1A3, ALDH2 and ALDH3A1. Moreover, an in vitro study demonstrated that all five compounds effectively improved glucose consumption in HepG2 cells, of which, 61 and 65 at 10 µM produced nearly equal glucose consumption with positive control Metformin (Met) at 1 mM. Furthermore, 61 and 65 showed desirable metabolic stability in human liver microsomes. All these results suggest that 61 and 65 are suitable for further studies.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Benzimidazoles/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Retinal Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase 1 Family/metabolism , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Retinal Dehydrogenase/metabolism , Structure-Activity Relationship
5.
Int J Obes (Lond) ; 45(7): 1542-1552, 2021 07.
Article in English | MEDLINE | ID: mdl-33934107

ABSTRACT

BACKGROUND: Retinoic acid (RA) controls diverse physiological functions including weight regulation and energy metabolism. It has been reported that mice lacking ALDH1A1, one of the aldehyde dehydrogenases (ALDH) that synthesize RA, are healthy and resistant to weight gain, raising the possibility that inhibiting this enzyme might treat obesity. We previously demonstrated that treatment with a pan-ALDH1A enzyme inhibitor, WIN18446, suppressed weight gain in mice fed a high-fat diet (HFD), but caused increased hepatic lipidosis and reversible male infertility. METHODS: A series of piperazine compounds that inhibited ALDH1A1 were identified and their inhibitory activity was characterized in vitro using purified recombinant enzymes and cell-based assay systems. One potent compound, FSI-TN42 (N42) was examined for its oral bioavailability and pharmacodynamic effects. In addition, its effect on weight gain was investigated by daily oral administration to C57BL/6 male mice receiving a HFD, and compared with mice receiving WIN18446 or vehicle alone (n = 6/group, 200 mg compound/kg body weight) for 5 weeks. Body weights were measured weekly, and a glucose tolerance test was performed after 4 weeks of treatment. Tissues were collected to determine changes in adipose weight, hepatic lipidosis, retinoid metabolism, and expression of genes associated with RA and lipid metabolism. RESULTS: N42 irreversibly binds and inhibits ALDH1A1 in vitro with a low nM IC50 and 800-fold specificity for ALDH1A1 compared to ALDH1A2. Daily oral administration of N42 significantly suppressed weight gain (P < 0.05) and reduced visceral adiposity (p < 0.05) in mice fed a HFD without the hepatic lipidosis observed with WIN18446 treatment. CONCLUSIONS: We developed a potent and specific inhibitor of ALDH1A1 that suppressed weight gain in mice fed a HFD. These findings demonstrate that inhibition of ALDH1A1 is a feasible target for drug development to treat and/or prevent obesity.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Obesity/metabolism , Piperazines/pharmacology , Retinal Dehydrogenase/antagonists & inhibitors , Weight Gain/drug effects , Adipose Tissue/drug effects , Administration, Oral , Animals , Diet, High-Fat , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Piperazines/administration & dosage , Piperazines/chemistry
6.
Bioorg Med Chem Lett ; 40: 127958, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33744437

ABSTRACT

Disulfiram is an FDA-approved drug used to treat chronic alcoholism. This drug works by blocking the second step of ethanol metabolism by inhibiting aldehyde dehydrogenase-2 (ALDH2), the enzyme responsible for acetaldehyde oxidation into acetic acid. This leads to the accumulation of acetaldehyde in the blood following alcohol ingestion and to highly unpleasant symptoms known as acetaldehyde syndrome. Disulfiram also inhibits ALDH1a1, another member of the aldehyde dehydrogenases that catalyzes the oxidation of retinal into retinoic acid. ALDH1a1 represents a key therapeutic target for the treatment of important diseases such as cancer and obesity. The substrate tunnel is larger in ALDH1a1 than in ALDH2; therefore. Thus, replacing disulfiram ethyl groups with larger groups will yield selective ALDH1a1 inhibitors. In this work, we successfully synthesized derivative 2b, in which two ethyl groups were replaced by two para fluorobenzyl groups. The 2b derivative showed a comparable activity to disulfiram against ALDH1a1; however, it was completely devoid of inhibitory activity against ALDH2.


Subject(s)
Acetaldehyde Dehydrogenase Inhibitors/chemistry , Alcohol Deterrents/chemistry , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Disulfiram/analogs & derivatives , Retinal Dehydrogenase/antagonists & inhibitors , Acetaldehyde Dehydrogenase Inhibitors/chemical synthesis , Alcohol Deterrents/chemical synthesis , Aldehyde Dehydrogenase 1 Family/chemistry , Aldehyde Dehydrogenase, Mitochondrial/antagonists & inhibitors , Aldehyde Dehydrogenase, Mitochondrial/chemistry , Disulfiram/chemical synthesis , Enzyme Assays , Humans , Retinal Dehydrogenase/chemistry , Substrate Specificity
7.
Theranostics ; 11(8): 3540-3551, 2021.
Article in English | MEDLINE | ID: mdl-33664846

ABSTRACT

Rationale: Aldehyde dehydrogenase (ALDH) enzymes are often upregulated in cancer cells and associated with therapeutic resistance. ALDH enzymes protect cells by metabolizing toxic aldehydes which can induce DNA double stand breaks (DSB). We recently identified a novel ALDH1A family inhibitor (ALDHi), 673A. We hypothesized that 673A, via inhibition of ALDH1A family members, could induce intracellular accumulation of genotoxic aldehydes to cause DSB and that ALDHi could synergize with inhibitors of the ATM and ATR, proteins which direct DSB repair. Methods: We used immunofluorescence to directly assess levels of the aldehyde 4-hydroxynonenal and comet assays to evaluate DSB. Western blot was used to evaluate activation of the DNA damage response pathways. Cell counts were performed in the presence of 673A and additional aldehydes or aldehyde scavengers. ALDH inhibition results were confirmed using ALDH1A3 CRISPR knockout. Synergy between 673A and ATM or ATR inhibitors was evaluated using the Chou-Talalay method and confirmed in vivo using cell line xenograft tumor studies. Results: The ALDHi 673A cellular accumulation of toxic aldehydes which induce DNA double strand breaks. This is exacerbated by addition of exogenous aldehydes such as vitamin-A (retinaldehyde) and ameliorated by aldehyde scavengers such as metformin and hydralazine. Importantly, ALDH1A3 knockout cells demonstrated increased sensitivity to ATM/ATR inhibitors. And, ALDHi synergized with inhibitors of ATM and ATR, master regulators of the DSB DNA damage response, both in vitro and in vivo. This synergy was evident in homologous recombination (HR) proficient cell lines. Conclusions: ALDHi can be used to induce DNA DSB in cancer cells and synergize with inhibitors the ATM/ATR pathway. Our data suggest a novel therapeutic approach to target HR proficient ovarian cancer cells.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , DNA Damage , Enzyme Inhibitors/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Aldehyde Oxidoreductases/deficiency , Aldehyde Oxidoreductases/genetics , Aldehydes/metabolism , Aldehydes/toxicity , Animals , Cell Line, Tumor , DNA Breaks, Double-Stranded , Drug Synergism , Enzyme Inhibitors/administration & dosage , Female , Gene Knockout Techniques , Humans , Mice , Precision Medicine , Protein Kinase Inhibitors/administration & dosage , Xenograft Model Antitumor Assays
8.
Eur J Med Chem ; 209: 112940, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33328099

ABSTRACT

ALDH1A1, one important member of 19 ALDHs, can metabolize reactive aldehydes to their corresponding carboxylic acid derivatives and play important physiological and toxicological roles in many areas, including CNS, metabolic disorders, and cancers. Overexpression of ALDH1A1 correlates with poor prognosis and tumor aggressiveness, is associated with drug resistance in traditional chemotherapy for cancer treatment and contributes to obesity, diabetes, and inflammation. So, inhibition of ALDH1A1 may offer new therapeutic options for patients with cancer, obesity, diabetes, and inflammation. Up to now, many ALDH1A1 inhibitors with different scaffolds have been identified and developed as useful chemical tools for better understanding of the functions of ALDH1A1 in physiologic and pathophysiologic conditions. In this review, the advances in the discovery and development of selective ALDH1A1 inhibitors are summarized, and opportunities and challenges associated with this field are also discussed.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Retinal Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Humans
9.
Nutrients ; 12(10)2020 Sep 24.
Article in English | MEDLINE | ID: mdl-32987910

ABSTRACT

Many inflammatory bowel disease (IBD) patients require surgical intervention due to limited pharmacological treatment options. Antibodies targeting α4ß7, a gut-homing integrin, are one of the most promising IBD treatments. As retinoic acid (RA) regulates expression of gut-homing proteins including α4ß7 integrin, we tested if ALDH1A enzymes in the RA synthesis pathway could be targeted for IBD treatment using a potent inhibitor, WIN 18,446. Age- and sex-matched Smad3-/- mice were fed a diet with and without WIN 18,446 for 3 weeks before triggering inflammation with Helicobacter bilis infection. Colitis was evaluated by histopathology one week following the IBD trigger, and T cell subsets were evaluated before and after the IBD trigger. WIN 18,446 treatment significantly reduced IBD severity in Smad3-/- mice and reduced expression of α4ß7 integrin on multiple activated CD4+ T cell subsets. This change was associated with increased ratios of induced regulatory T cells to Th17 cells during the inflammatory response in the draining lymph nodes. These studies indicate that RA reduction via ALDH1A enzyme inhibition is a potential new target for IBD treatment. Further studies are needed to examine its effects on other types of immune cells, to evaluate the efficacy window for this target, and to determine its efficacy in other animal models of IBD.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Colitis/drug therapy , Helicobacter/metabolism , Integrin alpha4/genetics , Lymphocyte Activation/drug effects , Retinal Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Animals , Colitis/etiology , Colitis/microbiology , Diamines/pharmacology , Disease Models, Animal , Female , Helicobacter Infections/complications , Helicobacter Infections/drug therapy , Integrin alpha4/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinal Dehydrogenase/antagonists & inhibitors
10.
Bioorg Chem ; 101: 103971, 2020 08.
Article in English | MEDLINE | ID: mdl-32480173

ABSTRACT

LDH1A1, one of 19 NAD(P)+-dependent aldehyde dehydrogenases, participates in multiple metabolic pathways and has been indicated to play an important role in obesity and diabetes. In this study, a series of 1,3-dimethylpyrimidine-2,4-diones were designed, synthesized and evaluated as novel selective aldehyde dehydrogenase 1A1 inhibitors. Among them, compounds 46, 50, 53, 56 and 57 exhibited excellent inhibitory activity against ALDH1A1 with IC50 values in the low nanomolar range and high selectivity over ALDH1A2, ALDH1A3, ALDH2 and ALDH3A1. Furthermore, in vitro study demonstrated that compound 57 effectively improved glucose consumption in HepG2 cells compared to compound 1 (CM026).


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Glucose/metabolism , Retinal Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors , Humans , Molecular Structure , Structure-Activity Relationship
11.
Drug Des Devel Ther ; 14: 1995-2019, 2020.
Article in English | MEDLINE | ID: mdl-32546966

ABSTRACT

INTRODUCTION: Targeted multimodal approaches need to be strategically developed to control tumour growth and prevent metastatic burden successfully. Breast cancer presents a unique clinical problem because of the variety of cellular subtypes that arise. The tumour stage and cellular subtypes often dictate the appropriate clinical treatment regimen. Also, the development of chemoresistance is a common clinical challenge with breast cancer. Higher doses and additional drug agents can produce additional adverse effects leading to a more aggressive malignancy. Acetylsalicylic acid (ASA), metformin (Met), and oseltamivir phosphate (OP) were investigated for their efficacy to sensitize MDA-MB-231 triple-negative breast cancer and its tamoxifen (Tmx) resistant variant (MDA-MB-231-TmxR) together in combination with Tmx treatment. METHODS: Microscopic imaging, the formation of 3D multicellular tumour spheroids, immunocytochemistry, flow cytometry, Annexin V Assay, Caspase 3/7 Apoptosis Assay, tube formation assay and analysis, and WST-1 cell viability assay evaluated the formation of MCTS, morphologic changes, cell viability, apoptosis activity and the expression levels of ALDH1A1, CD44 and CD24 on the cell surface, MDA-MB231 triple-negative breast cancer, tamoxifen (Tmx) resistant variant (MDA-MB-231-TmxR). RESULTS: The results using a triple combination of ASA, Met and OP on MDA-MB-231 and MDA-MB-231-TmxR cells and their matrix-free 3D multicellular tumour spheroids (MCTS) formed by using the cyclic Arg-Gly-Asp-D-Phe-Lys peptide modified with 4-carboxybutyl-triphenylphosphonium bromide (cyclo-RGDfK(TPP)) peptide method demonstrate a consistent and significant decrease in cell and tumour spheroid viability and volume with increased apoptotic activity, and increased sensitivity to Tmx therapy. Tmx treatment of MDA-MB-231 cells in combination with ASA, Met and OP markedly reduced the CD44/CD24 ratio by 6.5-fold compared to the untreated control group. Tmx treatment of MDA-MB-231-TmxR cells in combination with ASA, Met and OP markedly reduced the ALDH1A1 by 134-fold compared to the same treatment for the parental cell line. Also, the triple combination treatment of ASA, Met, and OP inhibited vasculogenic endothelial cell tube formation and induced endothelial cell apoptosis. CONCLUSION: For the first time, the findings demonstrate that repurposing ASA, Met, and OP provides a novel and promising targeted multimodal approach in the treatment of triple-negative breast cancer and its chemoresistant variant.


Subject(s)
Antineoplastic Agents/pharmacology , Aspirin/pharmacology , Breast Neoplasms/drug therapy , Metformin/pharmacology , Oseltamivir/pharmacology , Spheroids, Cellular/drug effects , Triple Negative Breast Neoplasms/drug therapy , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Aldehyde Dehydrogenase 1 Family/metabolism , Apoptosis/drug effects , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , CD24 Antigen/antagonists & inhibitors , CD24 Antigen/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Humans , Hyaluronan Receptors/antagonists & inhibitors , Hyaluronan Receptors/metabolism , Retinal Dehydrogenase/antagonists & inhibitors , Retinal Dehydrogenase/metabolism , Tamoxifen/pharmacology , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured
12.
Life Sci ; 253: 117733, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32360127

ABSTRACT

AIMS: Aldehyde dehydrogenase-1 (ALDH-1) is considered a signature of breast cancer stem cells and is linked to poor outcomes in breast cancer patients. This study aimed at investigating the effect of vitamin D3 on enhancing the tumor responsiveness to different conventional chemotherapeutic agents, viz., cisplatin, methotrexate, and doxorubicin. MAIN METHODS: In vitro and in vivo experiments were performed using combinations of vitamin D3 and chemotherapeutic agents. Cell cycle analysis and apoptosis assays were performed. Moreover, ALDH-1 expression levels were estimated in cancer cell lines and solid tumors. For solid tumors, tumor volume and histopathological necrotic indices were estimated. Leukocyte presence was also evaluated in tumors using leukocyte common antigen (LCA). KEY FINDINGS: Results showed a synergistic interaction between vitamin D3 and each of the chemotherapeutic agents on breast cancer cell lines as well as cell cycle arrest at G2/M phase. A decrease in ALDH-1 levels was reported in both breast cancer cells and in tumor tissues. Reductions in tumor volume were also observed in the groups which received the combination therapy. An influence on necrosis rather than apoptosis was also reported, as evidenced by necrotic indices and Bcl-2 expression in tumor sections, respectively. Increased local leukocytes in tumors was also evident, as indicated by increased expression of leukocyte common antigen (LCA). SIGNIFICANCE: Overall, the present study shows that vitamin D3 has an impact on resistance to different chemotherapeutic agents which could be due to the inhibition of ALDH-1, suggesting its use as an adjuvant therapy in cancer patients receiving chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Carcinoma, Ehrlich Tumor/drug therapy , Cholecalciferol/pharmacology , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/pathology , Carcinoma, Ehrlich Tumor/pathology , Cell Line, Tumor , Cholecalciferol/administration & dosage , Cisplatin/administration & dosage , Cisplatin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Humans , Leukocyte Common Antigens , Methotrexate/administration & dosage , Methotrexate/pharmacology , Mice , Neoplastic Stem Cells/enzymology
13.
Theranostics ; 10(7): 3083-3098, 2020.
Article in English | MEDLINE | ID: mdl-32194856

ABSTRACT

Over a decade of research has confirmed the critical role of cancer stem-like cells (CSCs) in tumor initiation, chemoresistance, and metastasis. Increasingly, CSC hierarchies have begun to be defined with some recurring themes. This includes evidence that these hierarchies are 'flexible,' with both cell state transitions and dedifferentiation events possible. These findings pose therapeutic hurdles and opportunities. Here, we review cancer stem cell hierarchies and their interactions with the tumor microenvironment. We also discuss the current therapeutic approaches designed to target CSC hierarchies and initial clinical trial results for CSC targeting agents. While cancer stem cell targeted therapies are still in their infancy, we are beginning to see encouraging results that suggest a positive outlook for CSC-targeting approaches.


Subject(s)
Neoplastic Stem Cells/classification , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Animals , Antigens, CD/drug effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Brain Neoplasms/pathology , Breast Neoplasms/pathology , Cell Dedifferentiation , Cell Lineage , Clinical Trials as Topic , Epigenesis, Genetic , Female , Humans , Metformin/pharmacology , Metformin/therapeutic use , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Tumor Microenvironment
14.
Oncogene ; 39(13): 2756-2771, 2020 03.
Article in English | MEDLINE | ID: mdl-32015486

ABSTRACT

Aldehyde dehydrogenases (ALDHs) are multifunctional enzymes that oxidize diverse endogenous and exogenous aldehydes. We conducted a meta-analysis based on The Cancer Genome Atlas and Gene Expression Omnibus data and detected genetic alterations in ALDH1A1, ALDH1A3, or ALDH3A1, 86% of which were gene amplification or mRNA upregulation, in 31% of nonsmall cell lung cancers (NSCLCs). The expression of these isoenzymes impacted chemoresistance and shortened survival times in patients. We hypothesized that these enzymes provide an oxidative advantage for the persistence of NSCLC. To test this hypothesis, we used genetic and pharmacological approaches with DIMATE, an irreversible inhibitor of ALDH1/3. DIMATE showed cytotoxicity in 73% of NSCLC cell lines tested and demonstrated antitumor activity in orthotopic xenografts via hydroxynonenal-protein adduct accumulation, GSTO1-mediated depletion of glutathione and increased H2O2. Consistent with this result, ALDH1/3 disruption synergized with ROS-inducing agents or glutathione synthesis inhibitors to trigger cell death. In lung cancer xenografts with high to moderate cisplatin resistance, combination treatment with DIMATE promoted strong synergistic responses with tumor regression. These results indicate that NSCLCs with increased expression of ALDH1A1, ALDH1A3, or ALDH3A1 may be targeted by strategies involving inhibitors of these isoenzymes as monotherapy or in combination with chemotherapy to overcome patient-specific drug resistance.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Oxidoreductases/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Retinal Dehydrogenase/antagonists & inhibitors , Aged , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Alkynes/pharmacology , Alkynes/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Amplification , Glutathione/metabolism , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Reactive Oxygen Species/metabolism , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Sulfhydryl Compounds/pharmacology , Sulfhydryl Compounds/therapeutic use , Up-Regulation , Xenograft Model Antitumor Assays
15.
Mol Cancer Ther ; 19(1): 199-210, 2020 01.
Article in English | MEDLINE | ID: mdl-31534014

ABSTRACT

Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) are approved to treat recurrent ovarian cancer with BRCA1 or BRCA2 mutations, and as maintenance therapy for recurrent platinum-sensitive ovarian cancer (BRCA wild-type or mutated) after treatment with platinum. However, the acquired resistance against PARPi remains a clinical hurdle. Here, we demonstrated that PARP inhibitor (olaparib)-resistant epithelial ovarian cancer (EOC) cells exhibited an elevated aldehyde dehydrogenase (ALDH) activity, mainly contributed by increased expression of ALDH1A1 due to olaparib-induced expression of BRD4, a member of bromodomain and extraterminal (BET) family protein. We also revealed that ALDH1A1 enhanced microhomology-mediated end joining (MMEJ) activity in EOC cells with inactivated BRCA2, a key protein that promotes homologous recombination (HR) by using an intrachromosomal MMEJ reporter. Moreover, NCT-501, an ALDH1A1-selective inhibitor, can synergize with olaparib in killing EOC cells carrying BRCA2 mutation in both in vitro cell culture and the in vivo xenograft animal model. Given that MMEJ activity has been reported to be responsible for PARPi resistance in HR-deficient cells, we conclude that ALDH1A1 contributes to the resistance to PARP inhibitors via enhancing MMEJ in BRCA2-/- ovarian cancer cells. Our findings provide a novel mechanism underlying PARPi resistance in BRCA2-mutated EOC cells and suggest that inhibition of ALDH1A1 could be exploited for preventing and overcoming PARPi resistance in EOC patients carrying BRCA2 mutation.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , DNA Repair , Nuclear Proteins/metabolism , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Retinal Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Aldehyde Dehydrogenase 1 Family/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA End-Joining Repair , Drug Resistance, Neoplasm , Drug Synergism , Female , Humans , Mice , Mice, Nude , Mutation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phthalazines/administration & dosage , Phthalazines/pharmacology , Piperazines/administration & dosage , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Retinal Dehydrogenase/antagonists & inhibitors , Retinal Dehydrogenase/genetics , Theophylline/administration & dosage , Theophylline/pharmacology , Transcription Factors/metabolism , Transfection , Xenograft Model Antitumor Assays
16.
Int J Cancer ; 146(4): 1099-1113, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31187490

ABSTRACT

Acquired chemoresistance is a critical issue for advanced bladder cancer patients during long-term treatment. Recent studies reveal that a fraction of tumor cells with enhanced tumor-initiating potential, or cancer stem-like cells (CSCs), may particularly contribute to acquired chemoresistance and recurrence. Thus, CSC characterization will be the first step towards understanding the mechanisms underlying advanced disease. Here we generated long-term patient-derived cancer cells (PDCs) from bladder cancer patient specimens in spheroid culture, which is favorable for CSC enrichment. Pathological features of bladder cancer PDCs and PDC-dependent patient-derived xenografts (PDXs) were basically similar to those of their corresponding patients' specimens. Notably, CSC marker aldehyde dehydrogenase 1A1 (ALDH1A1), a critical enzyme that synthesizes retinoic acid (RA), was abundantly expressed in PDCs. ALDH1A1 inhibitors and shRNAs repressed both PDC proliferation and spheroid formation, whereas all-trans RA could rescue ALDH1A1 shRNA-suppressed spheroid formation. ALDH inhibitor also reduced the in vivo growth of PDC-derived xenografts. ALDH1A1 knockdown study showed that tubulin beta III (TUBB3) was one of the downregulated genes in PDCs. We identified functional RA response elements in TUBB3 promoter, whose transcriptional activities were substantially activated by RA. Clinical survival database reveals that TUBB3 expression may associate with poor prognosis in bladder cancer patients. Moreover, TUBB3 knockdown was sufficient to suppress PDC proliferation and spheroid formation. Taken together, our results indicate that ALDH1A1 and its putative downstream target TUBB3 are overexpressed in bladder cancer, and those molecules could be applied to alternative diagnostic and therapeutic options for advanced disease.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Retinal Dehydrogenase/metabolism , Tubulin/biosynthesis , Urinary Bladder Neoplasms/metabolism , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Aldehyde Dehydrogenase 1 Family/genetics , Animals , Cell Line, Tumor , Disease Progression , Down-Regulation , HEK293 Cells , Heterografts , Humans , Male , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Retinal Dehydrogenase/antagonists & inhibitors , Retinal Dehydrogenase/genetics , Retinoic Acid Receptor alpha , Signal Transduction , Spheroids, Cellular , Tretinoin , Tubulin/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
17.
Eur J Med Chem ; 187: 111923, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31816557

ABSTRACT

Overexpression of aldehyde dehydrogenase 1A1 (ALDH1A1) is associated with the occurrence and development of obesity and insulin resistance. Herein, a series of coumarin-based ALDH1A1 inhibitors were designed, synthesized and evaluated. Among them, compounds 10, 14 and 26 exhibited potent inhibitory activity against ALDH1A1 and high selectivity over ALDH1A2, ALDH1A3, ALDH2 and ALDH3A1. Optimized compound 10 showed markedly improved pharmacokinetic characters and ADME profiles comparing to the lead compound 1. In vitro study demonstrated that 10 alleviated palmitic acid-induced impairment of glucose consumption in HepG2 cells.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Retinal Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase 1 Family/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hep G2 Cells , Humans , Molecular Structure , Retinal Dehydrogenase/metabolism , Structure-Activity Relationship
18.
Mol Cancer Ther ; 19(2): 447-459, 2020 02.
Article in English | MEDLINE | ID: mdl-31754071

ABSTRACT

The aldehyde dehydrogenases (ALDH) are a major family of detoxifying enzymes that contribute to cancer progression and therapy resistance. ALDH overexpression is associated with a poor prognosis in many cancer types. The use of multi-ALDH isoform or isoform-specific ALDH inhibitors as anticancer agents is currently hindered by the lack of viable candidates. Most multi-ALDH isoform inhibitors lack bioavailability and are nonspecific or toxic, whereas most isoform-specific inhibitors are not effective as monotherapy due to the overlapping functions of ALDH family members. The present study details the development of a novel, potent, multi-isoform ALDH inhibitor, called KS100. The rationale for drug development was that inhibition of multiple ALDH isoforms might be more efficacious for cancer compared with isoform-specific inhibition. Enzymatic IC50s of KS100 were 207, 1,410, and 240 nmol/L toward ALDH1A1, 2, and 3A1, respectively. Toxicity of KS100 was mitigated by development of a nanoliposomal formulation, called NanoKS100. NanoKS100 had a loading efficiency of approximately 69% and was stable long-term. NanoKS100 was 5-fold more selective for killing melanoma cells compared with normal human fibroblasts. NanoKS100 administered intravenously at a submaximal dose (3-fold lower) was effective at inhibiting xenografted melanoma tumor growth by approximately 65% without organ-related toxicity. Mechanistically, inhibition by KS100 significantly reduced total cellular ALDH activity to increase reactive oxygen species generation, lipid peroxidation, and accumulation of toxic aldehydes leading to apoptosis and autophagy. Collectively, these data suggest the successful preclinical development of a nontoxic, bioavailable, nanoliposomal formulation containing a novel multi-ALDH isoform inhibitor effective in the treatment of cancer.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Melanoma/drug therapy , Animals , Cell Line, Tumor , Humans , Mice , Transfection
19.
Acta Biochim Pol ; 66(4): 627-632, 2019 Dec 28.
Article in English | MEDLINE | ID: mdl-31883320

ABSTRACT

The aim of the study presented here was an attempt to answer the question posed in the title: Is the mechanism of nitroglycerin tolerance associated with aldehyde dehydrogenase (ALDH) activity? Here, we investigated the effect of administration (separately or jointly) of lipoic acid (LA), nitroglycerin (GTN), and disulfiram (DSF; an irreversible in vivo inhibitor of all ALDH isozymes (including ALDH2)), on the development of tolerance to GTN. We also assessed the total activity of ALDH in the rat liver homogenates. Our data revealed that not only DSF and GTN inhibited the total ALDH activity in the rat liver, but LA also proved to be an inhibitor of this enzyme. At the same time, the obtained results demonstrated that the GTN tolerance did not develop in GTN, DSF and LA jointly treated rats, but did develop in GTN and DSF jointly treated rats. This means that the ability of LA to prevent GTN tolerance is not associated with the total ALDH activity in the rat liver. In this context, the fact that animals jointly receiving GTN and DSF developed tolerance to GTN, and in animals that in addition to GTN and DSF also received LA such tolerance did not develop, is - in our opinion - a sufficient premise to conclude that the nitrate tolerance certainly is not caused by a decrease in the activity of any of the ALDH isoenzymes present in the rat liver, including ALDH2. However, many questions still await an answer, including the basic one: What is the mechanism of tolerance to nitroglycerin?


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/antagonists & inhibitors , Disulfiram/pharmacology , Drug Tolerance/genetics , Nitroglycerin/pharmacology , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Animals , Humans , Oxidation-Reduction/drug effects , Rats , Thioctic Acid/pharmacology
20.
Cell Rep ; 26(11): 3061-3075.e6, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30865894

ABSTRACT

Ovarian cancer is typified by the development of chemotherapy resistance. Chemotherapy resistance is associated with high aldehyde dehydrogenase (ALDH) enzymatic activity, increased cancer "stemness," and expression of the stem cell marker CD133. As such, ALDH activity has been proposed as a therapeutic target. Although it remains controversial which of the 19 ALDH family members drive chemotherapy resistance, ALDH1A family members have been primarily linked with chemotherapy resistant and stemness. We identified two ALDH1A family selective inhibitors (ALDH1Ai). ALDH1Ai preferentially kills CD133+ ovarian cancer stem-like cells (CSCs). ALDH1Ai induce necroptotic CSC death, mediated, in part, by the induction of mitochondrial uncoupling proteins and reduction in oxidative phosphorylation. ALDH1Ai is highly synergistic with chemotherapy, reducing tumor initiation capacity and increasing tumor eradication in vivo. These studies link ALDH1A with necroptosis and confirm the family as a critical therapeutic target to overcome chemotherapy resistance and improve patient outcomes.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Necroptosis , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Retinal Dehydrogenase/antagonists & inhibitors , AC133 Antigen/genetics , AC133 Antigen/metabolism , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Mice , Neoplastic Stem Cells/drug effects , Oxidative Phosphorylation , Retinal Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...