Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Chem Pharm Bull (Tokyo) ; 72(7): 630-637, 2024.
Article in English | MEDLINE | ID: mdl-38945939

ABSTRACT

Alzheimer's disease (AD) is the leading cause of senile dementia, and the rapid increase in the frequency of AD cases has been attributed to population aging. However, current drugs have difficulty adequately suppressing symptoms and there is still a medical need for symptomatic agents. On the other hand, it has recently become clear that epigenetic dysfunctions are deeply involved in the development of cognitive impairments. Therefore, epigenetics-related proteins have attracted much attention as drug targets for AD. Early-developed epigenetic inhibitors were inappropriate for AD treatment because of their limited potential for oral administration, blood-brain barrier penetration, high target selectivity, and sufficient dose-limiting toxicity which are essential properties for small molecule drugs targeting chronic neurodegenerative diseases such as AD. In recent years, drug discovery studies have been actively performed to overcome such problems and several novel inhibitors targeting the epigenetics-related proteins are of interest as promising AD therapeutic agents. Here, we review the small molecule inhibitors of histone deacetylase (HDAC), lysine-specific demethylase 1 (LSD1) or bromodomains and extra-terminal domain (BET) protein, that enable memory function improvement in AD model mice.


Subject(s)
Alzheimer Disease , Epigenesis, Genetic , Histone Deacetylase Inhibitors , Histone Demethylases , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Animals , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Histone Deacetylases/metabolism
2.
Chem Pharm Bull (Tokyo) ; 72(7): 638-647, 2024.
Article in English | MEDLINE | ID: mdl-38945940

ABSTRACT

Lysine demethylase 5 (KDM5) proteins are involved in various neurological disorders, including Alzheimer's disease, and KDM5 inhibition is expected to be a therapeutic strategy for these diseases. However, the pharmacological effects of conventional KDM5 inhibitors are insufficient, as they only target the catalytic functionality of KDM5. To identify compounds that exhibit more potent pharmacological activity, we focused on proteolysis targeting chimeras (PROTACs), which degrade target proteins and thus inhibit their entire functionality. We designed and synthesized novel KDM5 PROTAC candidates based on previously identified KDM5 inhibitors. The results of cellular assays revealed that two compounds, 20b and 23b, exhibited significant neurite outgrowth-promoting activity through the degradation of KDM5A in neuroblastoma neuro 2a cells. These results suggest that KDM5 PROTACs are promising drug candidates for the treatment of neurological disorders.


Subject(s)
Neuronal Outgrowth , Proteolysis , Proteolysis/drug effects , Humans , Neuronal Outgrowth/drug effects , Structure-Activity Relationship , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Cell Line, Tumor , Molecular Structure , Retinoblastoma-Binding Protein 2/metabolism , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Animals , Mice , Dose-Response Relationship, Drug , Proteolysis Targeting Chimera
3.
Bioorg Med Chem Lett ; 105: 129759, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38636717

ABSTRACT

Histone H2A mono-ubiquitination plays important roles in epigenetic gene expression and is also involved in tumorigenesis. Small molecules controlling H2A ubiquitination are of interest as potential chemical tools and anticancer drugs. To identify novel small molecule inhibitors of H2A ubiquitination, we synthesized and evaluated several compounds designed based on PRT4165 (1), which is a reported histone ubiquitin ligase RING1A inhibitor. We found that compound 11b strongly inhibited the viability and reduced histone H2A mono-ubiquitination in human osteosarcoma U2OS cells. Therefore, compound 11b is a promising lead compound for the development of H2A histone ubiquitination-inhibiting small molecules.


Subject(s)
Histones , Small Molecule Libraries , Ubiquitination , Humans , Histones/metabolism , Ubiquitination/drug effects , Cell Line, Tumor , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Molecular Structure , Cell Survival/drug effects , Dose-Response Relationship, Drug
4.
Bioorg Med Chem ; 100: 117632, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38340642

ABSTRACT

Small molecule-based selective cancer cell-targeting can be a desirable anticancer therapeutic strategy. Aiming to discover such small molecules, we previously developed phenylcyclopropylamine (PCPA)-drug conjugates (PDCs) that selectively release anticancer agents in cancer cells where lysine-specific demethylase 1 (LSD1) is overexpressed. In this work, we designed PCPA-entinostat conjugates for selective cancer cell targeting. PCPA-entinostat conjugate 12 with a 4-oxybenzyl group linker released entinostat in the presence of LSD1 in in vitro assays and selectively inhibited the growth of cancer cells in preference to normal cells, suggesting the potential of PCPA-entinostat conjugates as novel anticancer drug delivery small molecules.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzamides , Histone Demethylases , Neoplasms/drug therapy , Pyridines , Cyclopropanes/chemistry
5.
Bioorg Med Chem ; 98: 117579, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38168630

ABSTRACT

Lysine demethylase 5 (KDM5) subfamily proteins are important in epigenetic gene regulation. They are involved in the growth and drug resistance of cancer cells. Therefore, KDM5s are potential cancer therapeutic targets, and their inhibitors hold promise as anti-cancer drugs. Several KDM5 inhibitors, including KDM5-C49 (2a), have exhibited potent KDM5-inhibitory activities in in vitro enzyme assays. However, they do not show enough cellular activity despite being converted to their prodrugs. We hypothesized that their poor lipophilicity should prevent them from sufficiently penetrating the cell membrane, and introducing more lipophilic groups should improve cellular activities. In this study, we investigated 2a and KDM5-C70 (3a), a prodrug of 2a, and attempted to improve its cellular activity by replacing the N,N-dimethyl amino group of 3a with more lipophilic groups. N-Butyl, N-methyl amino compound 2e exhibited potent and selective KDM5-inhibitory activity equal to that of 2a. Furthermore, the cell membrane permeability of 3e, an ethyl ester prodrug of 2e, was six times higher than that of 3a in a parallel artificial membrane permeation assay. In addition, western blot analysis indicated that treating human lung cancer A549 cells with 3e increased histone methylation levels more strongly than that with 3a. Thus, we identified compound 3e as a more cell-active KDM5 inhibitor that has sufficient cell membrane permeability.


Subject(s)
Antineoplastic Agents , Neoplasms , Prodrugs , Humans , Lysine , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Neoplasms/metabolism , Prodrugs/pharmacology
6.
Chem Pharm Bull (Tokyo) ; 72(2): 155-160, 2024.
Article in English | MEDLINE | ID: mdl-38296557

ABSTRACT

Peptides have recently garnered attention as middle-molecular-weight drugs with the characteristics of small molecules and macromolecules. Lysine-specific demethylase 1 (LSD1) is a potential therapeutic target for lung cancer, neuroblastoma, and leukemia, and some peptide-based LSD1 inhibitors designed based on the N-terminus of SNAIL1, a member of the SNAIL/SCRATCH family of transcription factors, have been reported. The N-terminus of SNAIL1 peptide acts as a cap of the catalytic site of LSD1, inhibiting interactions with LSD1. However, the structure-activity relationship (SAR) of these inhibitors is not yet fully understood. Therefore, in the present study, we aimed to uncover the SAR and to identify novel SNAIL1 peptide-based LSD1 inhibitors. We synthesized peptide inhibitor candidates based on truncating the N-terminus of SNAIL1 or substituting its amino acid residues. In the truncation study, we found that SNAIL1 1-16 (2), which was composed of 16 residues, strongly inhibited LSD1. Furthermore, we investigated the SAR at residues-3 and -5 from the N-terminus and found that peptides 2j and 2k, in which leucine 5 of the parent peptide is substituted with unnatural amino acids, cyclohexylalanine and norleucine, respectively, strongly inhibited LSD1. This result suggests that the hydrophobic interaction between the inhibitor peptides and LSD1 affects the LSD1-inhibitory activity. We believe that this SAR information provides a basis for the development of more potent LSD1 inhibitors.


Subject(s)
Enzyme Inhibitors , Lysine , Lysine/chemistry , Enzyme Inhibitors/chemistry , Peptides/pharmacology , Peptides/chemistry , Structure-Activity Relationship , Amino Acids , Histone Demethylases
7.
Chem Pharm Bull (Tokyo) ; 72(2): 173-178, 2024.
Article in English | MEDLINE | ID: mdl-38296560

ABSTRACT

Histone deacetylase 8 (HDAC8) is a zinc-dependent HDAC that catalyzes the deacetylation of nonhistone proteins. It is involved in cancer development and HDAC8 inhibitors are promising candidates as anticancer agents. However, most reported HDAC8 inhibitors contain a hydroxamic acid moiety, which often causes mutagenicity. Therefore, we used machine learning for drug screening and attempted to identify non-hydroxamic acids as HDAC8 inhibitors. In this study, we established a prediction model based on the random forest (RF) algorithm for screening HDAC8 inhibitors because it exhibited the best predictive accuracy in the training dataset, including data generated by the synthetic minority over-sampling technique (SMOTE). Using the trained RF-SMOTE model, we screened the Osaka University library for compounds and selected 50 virtual hits. However, the 50 hits in the first screening did not show HDAC8-inhibitory activity. In the second screening, using the RF-SMOTE model, which was established by retraining the dataset including 50 inactive compounds, we identified non-hydroxamic acid 12 as an HDAC8 inhibitor with an IC50 of 842 nM. Interestingly, its IC50 values for HDAC1 and HDAC3-inhibitory activity were 38 and 12 µM, respectively, showing that compound 12 has high HDAC8 selectivity. Using machine learning, we expanded the chemical space for HDAC8 inhibitors and identified non-hydroxamic acid 12 as a novel HDAC8 selective inhibitor.


Subject(s)
Antineoplastic Agents , Histone Deacetylase Inhibitors , Humans , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry , Drug Evaluation, Preclinical , Histone Deacetylases/metabolism , Antineoplastic Agents/pharmacology , Hydroxamic Acids/pharmacology , Hydroxamic Acids/chemistry , Machine Learning , Repressor Proteins
8.
J Med Chem ; 66(22): 15171-15188, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37847303

ABSTRACT

Histone deacetylase 1 and 2 (HDAC1/2) inhibitors are potentially useful as tools for probing the biological functions of the isoforms and as therapeutic agents for cancer and neurodegenerative disorders. To discover potent and selective inhibitors, we screened a focused library synthesized by using click chemistry and obtained KPZ560 as an HDAC1/2-selective inhibitor. Kinetic binding analysis revealed that KPZ560 inhibits HDAC2 through a two-step slow-binding mechanism. In cellular assays, KPZ560 induced a dose- and time-dependent increase of histone acetylation and showed potent breast cancer cell growth-inhibitory activity. In addition, gene expression analyses suggested that the two-step slow-binding inhibition by KPZ560 regulated the expression of genes associated with cell proliferation and DNA damage. KPZ560 also induced neurite outgrowth of Neuro-2a cells and an increase in the spine density of granule neuron dendrites of mice. The unique two-step slow-binding character of o-aminoanilides such as KPZ560 makes them interesting candidates as therapeutic agents.


Subject(s)
Histone Deacetylase 1 , Histone Deacetylases , Mice , Animals , Histone Deacetylases/metabolism , Click Chemistry , Histone Deacetylase Inhibitors/pharmacology , Neurons/metabolism , Histone Deacetylase 2
9.
ACS Med Chem Lett ; 13(10): 1568-1573, 2022 Oct 13.
Article in English | MEDLINE | ID: mdl-36262394

ABSTRACT

Anticancer drug delivery by small molecules offers a number of advantages over conventional macromolecular drug delivery systems. We previously developed phenylcyclopropylamine (PCPA)-drug conjugates (PDCs) as small-molecule-based drug delivery vehicles for targeting lysine-specific demethylase 1 (LSD1)-overexpressing cancers. In this study, we applied this PDC strategy to the HDAC-inhibitory anticancer agent vorinostat. Among three synthesized PCPA or arylcyclopropylamine (ACPA)-vorinostat conjugates 1, 9, and 32, conjugate 32 with a 4-oxybenzyl linker showed sufficient stability in buffer solutions, potent LSD1 inhibition, efficient LSD1-dependent vorinostat release, and potent and selective antiproliferative activity toward LSD1-expressing human breast cancer and small-cell lung cancer cell lines. These results indicate that the conjugate selectively releases vorinostat in cancer cells. A similar strategy may be applicable to other anticancer drugs.

10.
Curr Opin Chem Biol ; 67: 102130, 2022 04.
Article in English | MEDLINE | ID: mdl-35240380

ABSTRACT

For many years, drug discovery studies in the field of epigenetics have focused mainly on specific enzymes such as histone deacetylases (HDACs). However, recently there has been increasing interest in small molecules targeting the multiprotein enzyme/transcription factor complexes that play key roles in the epigenetic control of gene expression. Aberrant function of these complexes often has pathological consequences. Here, we review small molecules that modulate the function of three well-known epigenetic complexes, namely, polycomb repressive complex 2 (PRC2), PRC1, and corepressor of RE1-silencing transcription factor (CoREST) complex, focusing on recent drug discovery studies targeting these epigenetic complexes.


Subject(s)
Gene Expression Regulation , Polycomb Repressive Complex 2 , Epigenesis, Genetic , Epigenomics , Histone Deacetylases/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
11.
Chem Commun (Camb) ; 58(29): 4635-4638, 2022 Apr 07.
Article in English | MEDLINE | ID: mdl-35311871

ABSTRACT

We developed a first-in-class proteolysis targeting chimera (PROTAC) for selective degradation of histone deacetylase 8 (HDAC8). The PROTAC induced degradation of HDAC8 without affecting the levels of other HDACs in cellular assays, and inhibited the growth of T-cell leukemia Jurkat cells more potently than a conventional HDAC8 inhibitor.


Subject(s)
Drug Discovery , Histone Deacetylase Inhibitors , Histone Deacetylases , Proteolysis , Repressor Proteins , Drug Discovery/methods , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Jurkat Cells , Repressor Proteins/metabolism
12.
J Med Chem ; 64(21): 15810-15824, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34727689

ABSTRACT

Fat mass obesity-associated protein (FTO) is a DNA/RNA demethylase involved in the epigenetic regulation of various genes and is considered a therapeutic target for obesity, cancer, and neurological disorders. Here, we aimed to design novel FTO-selective inhibitors by merging fragments of previously reported FTO inhibitors. Among the synthesized analogues, compound 11b, which merges key fragments of Hz (3) and MA (4), inhibited FTO selectively over alkylation repair homologue 5 (ALKBH5), another DNA/RNA demethylase. Treatment of acute monocytic leukemia NOMO-1 cells with a prodrug of 11b decreased the viability of acute monocytic leukemia cells, increased the level of the FTO substrate N6-methyladenosine in mRNA, and induced upregulation of MYC and downregulation of RARA, which are FTO target genes. Thus, Hz (3)/MA (4) hybrid analogues represent an entry into a new class of FTO-selective inhibitors.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Adenosine/analogs & derivatives , Adenosine/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Drug Design , Humans , Substrate Specificity , Up-Regulation/drug effects
13.
Chembiochem ; 22(22): 3158-3163, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34224197

ABSTRACT

Pharmacological inhibition of histone deacetylase 6 (HDAC6) is an effective therapeutic strategy for cancer and immunological diseases. Most of the previously reported HDAC6 inhibitors have a hydroxamate group as a zinc binding group (ZBG), which coordinates to the catalytic zinc ion of HDAC6. The hydroxamate group is liable to metabolically generate mutagenetic hydroxylamine; therefore, non-hydroxamate HDAC6 inhibitors would be advantageous. In this study, to identify novel non-hydroxamate HDAC6-selective inhibitors, screening of a chemical library and the subsequent structural optimization were performed, which led to the identification of HDAC6-selective inhibitors with 3,3,3-trifluorolactic amide (TFLAM) as a novel ZBG. The identified inhibitor showed potent and selective HDAC6-inhibitory activity in cells and induced regulatory T (Treg) cell differentiation.


Subject(s)
Amides/pharmacology , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Lactates/pharmacology , Zinc/pharmacology , Amides/chemistry , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemistry , Humans , Lactates/chemistry , Molecular Docking Simulation , Molecular Structure , Zinc/chemistry
14.
Chem Soc Rev ; 50(2): 1305-1353, 2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33290462

ABSTRACT

Antibody-drug conjugates (ADCs) harness the highly specific targeting capabilities of an antibody to deliver a cytotoxic payload to specific cell types. They have garnered widespread interest in drug discovery, particularly in oncology, as discrimination between healthy and malignant tissues or cells can be achieved. Nine ADCs have received approval from the US Food and Drug Administration and more than 80 others are currently undergoing clinical investigations for a range of solid tumours and haematological malignancies. Extensive research over the past decade has highlighted the critical nature of the linkage strategy adopted to attach the payload to the antibody. Whilst early generation ADCs were primarily synthesised as heterogeneous mixtures, these were found to have sub-optimal pharmacokinetics, stability, tolerability and/or efficacy. Efforts have now shifted towards generating homogeneous constructs with precise drug loading and predetermined, controlled sites of attachment. Homogeneous ADCs have repeatedly demonstrated superior overall pharmacological profiles compared to their heterogeneous counterparts. A wide range of methods have been developed in the pursuit of homogeneity, comprising chemical or enzymatic methods or a combination thereof to afford precise modification of specific amino acid or sugar residues. In this review, we discuss advances in chemical and enzymatic methods for site-specific antibody modification that result in the generation of homogeneous ADCs.


Subject(s)
Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Immunoconjugates/chemistry , Humans , Molecular Structure
15.
Nat Commun ; 11(1): 4935, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004797

ABSTRACT

Gramicidin A (1) is a peptide antibiotic that disrupts the transmembrane ion concentration gradient by forming an ion channel in a lipid bilayer. Although long used clinically, it is limited to topical application because of its strong hemolytic activity and mammalian cytotoxicity, likely arising from the common ion transport mechanism. Here we report an integrated high-throughput strategy for discovering analogues of 1 with altered biological activity profiles. The 4096 analogue structures are designed to maintain the charge-neutral, hydrophobic, and channel forming properties of 1. Synthesis of the analogues, tandem mass spectrometry sequencing, and 3 microscale screenings enable us to identify 10 representative analogues. Re-synthesis and detailed functional evaluations find that all 10 analogues share a similar ion channel function, but have different cytotoxic, hemolytic, and antibacterial activities. Our large-scale structure-activity relationship studies reveal the feasibility of developing analogues of 1 that selectively induce toxicity toward target organisms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Discovery/methods , Gramicidin/analogs & derivatives , High-Throughput Screening Assays/methods , Animals , Anti-Bacterial Agents/chemistry , Cell Line, Tumor , Chemistry, Pharmaceutical , Erythrocytes , Feasibility Studies , Gram-Positive Bacteria/drug effects , Gramicidin/chemistry , Gramicidin/pharmacology , Hemolysis/drug effects , Inhibitory Concentration 50 , Mice , Microbial Sensitivity Tests , Molecular Structure , Rabbits , Structure-Activity Relationship , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...