Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 268
1.
J Med Chem ; 65(3): 2434-2457, 2022 02 10.
Article En | MEDLINE | ID: mdl-35043615

A series of 2-phenylthiazole analogues were designed and synthesized as potential histone deacetylase 6 (HDAC6) inhibitors based on compound 12c (an HDAC6/tubulin dual inhibitor discovered by us recently) and CAY10603 (a known HDAC6 inhibitor). Among them, compound XP5 was the most potent HDAC6 inhibitor with an IC50 of 31 nM and excellent HDAC6 selectivity (SI = 338 for HDAC6 over HDAC3). XP5 also displayed high antiproliferative activity against various cancer cell lines including the HDACi-resistant YCC3/7 gastric cancer cells (IC50 = 0.16-2.31 µM), better than CAY10603. Further, XP5 (50 mg/kg) exhibited significant antitumor efficacy in a melanoma tumor model with a tumor growth inhibition (TGI) of 63% without apparent toxicity. Moreover, XP5 efficiently enhanced the in vivo antitumor immune response when combined with a small-molecule PD-L1 inhibitor, as demonstrated by the increased tumor-infiltrating lymphocytes and reduced PD-L1 expression levels. Taken together, the above results suggest that XP5 is a promising HDAC6 inhibitor deserving further investigation.


Antineoplastic Agents/therapeutic use , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Immunity/drug effects , Melanoma/drug therapy , Thiazoles/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , G2 Phase Cell Cycle Checkpoints/drug effects , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/toxicity , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/therapeutic use , Hydroxamic Acids/toxicity , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Male , Melanoma/therapy , Mice , Molecular Structure , Rats, Sprague-Dawley , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/pharmacokinetics , Thiazoles/toxicity
2.
Invest Ophthalmol Vis Sci ; 62(14): 8, 2021 11 01.
Article En | MEDLINE | ID: mdl-34757417

Purpose: Current melphalan-based regimens for intravitreal chemotherapy for retinoblastoma vitreous seeds are effective but toxic to the retina. Thus, alternative agents are needed. Based on the known biology of histone deacetylases (HDACs) in the retinoblastoma pathway, we systematically studied whether the HDAC inhibitor belinostat is a viable, molecularly targeted alternative agent for intravitreal delivery that might provide comparable efficacy, without toxicity. Methods: In vivo pharmacokinetic experiments in rabbits and in vitro cytotoxicity experiments were performed to determine the 90% inhibitory concentration (IC90). Functional toxicity by electroretinography and structural toxicity by optical coherence tomography (OCT), OCT angiography, and histopathology were evaluated in rabbits following three injections of belinostat 350 µg (2× IC90) or 700 µg (4× IC90), compared with melphalan 12.5 µg (rabbit equivalent of the human dose). The relative efficacy of intravitreal belinostat versus melphalan to treat WERI-Rb1 human cell xenografts in rabbit eyes was directly quantified. RNA sequencing was used to assess belinostat-induced changes in RB cell gene expression. Results: The maximum nontoxic dose of belinostat was 350 µg, which caused no reductions in electroretinography parameters, retinal microvascular loss on OCT angiography, or retinal degeneration. Melphalan caused severe retinal structural and functional toxicity. Belinostat 350 µg (equivalent to 700 µg in the larger human eye) was equally effective at eradicating vitreous seeds in the rabbit xenograft model compared with melphalan (95.5% reduction for belinostat, P < 0.001; 89.4% reduction for melphalan, P < 0.001; belinostat vs. melphalan, P = 0.10). Even 700 µg belinostat (equivalent to 1400 µg in humans) caused only minimal toxicity. Widespread changes in gene expression resulted. Conclusions: Molecularly targeted inhibition of HDACs with intravitreal belinostat was equally effective as standard-of-care melphalan but without retinal toxicity. Belinostat may therefore be an attractive agent to pursue clinically for intravitreal treatment of retinoblastoma.


Disease Models, Animal , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Neoplasm Seeding , Retina/drug effects , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy , Sulfonamides/therapeutic use , Animals , Annexin A5 , Antineoplastic Agents, Alkylating/therapeutic use , Electroretinography , Fluorescein Angiography , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/toxicity , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/toxicity , Intravitreal Injections , Maximum Tolerated Dose , Melphalan/therapeutic use , Rabbits , Retina/physiology , Retinal Neoplasms/diagnosis , Retinal Neoplasms/physiopathology , Retinoblastoma/diagnosis , Retinoblastoma/physiopathology , Retrospective Studies , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity , Tomography, Optical Coherence , Vitreous Body/metabolism , Xenograft Model Antitumor Assays
3.
J Med Chem ; 64(12): 8486-8509, 2021 06 24.
Article En | MEDLINE | ID: mdl-34101461

Epigenetic targeting has emerged as an efficacious therapy for hematological cancers. The rare and incurable T-cell prolymphocytic leukemia (T-PLL) is known for its aggressive clinical course. Current epigenetic agents such as histone deacetylase (HDAC) inhibitors are increasingly used for targeted therapy. Through a structure-activity relationship (SAR) study, we developed an HDAC6 inhibitor KT-531, which exhibited higher potency in T-PLL compared to other hematological cancers. KT-531 displayed strong HDAC6 inhibitory potency and selectivity, on-target biological activity, and a safe therapeutic window in nontransformed cell lines. In primary T-PLL patient cells, where HDAC6 was found to be overexpressed, KT-531 exhibited strong biological responses, and safety in healthy donor samples. Notably, combination studies in T-PLL patient samples demonstrated KT-531 synergizes with approved cancer drugs, bendamustine, idasanutlin, and venetoclax. Our work suggests HDAC inhibition in T-PLL could afford sufficient therapeutic windows to achieve durable remission either as stand-alone or in combination with targeted drugs.


Antineoplastic Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Leukemia, Prolymphocytic, T-Cell/drug therapy , Sulfonamides/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Bendamustine Hydrochloride/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Drug Synergism , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacokinetics , Male , Mice , Molecular Docking Simulation , Molecular Structure , Pyrrolidines/pharmacology , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , para-Aminobenzoates/pharmacology
4.
J Med Chem ; 64(4): 2254-2271, 2021 02 25.
Article En | MEDLINE | ID: mdl-33541085

Previously, we identified the clinical anticancer drug candidate quisinostat as a novel and potent antimalarial lead compound. To further enhance the antimalarial effect and improve safety, 31 novel spirocyclic hydroxamic acid derivatives were synthesized based on the structure of quisinostat, and their antimalarial activities and cytotoxicity were evaluated. Among them, compound 11 displayed broad potency in vitro against several multiresistant malarial parasites, especially two artemisinin-resistant clinical isolates. Moreover, 11 could eliminate both liver and erythrocytic parasites in vivo, kill all morphological erythrocytic parasites with specific potency against schizonts, and show acceptable metabolic stability and pharmacokinetic properties. Western blot analysis, PfHDAC gene knockdown, and enzymatic inhibition experiments collectively confirmed that PfHDAC1 was the target of 11. In summary, 11 is a structurally novel PfHDAC1 inhibitor with the potential to prevent and cure malaria, overcome multidrug resistance, and provide a prospective prototype for antimalarial drug research.


Antimalarials/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Animals , Antimalarials/chemical synthesis , Antimalarials/metabolism , Antimalarials/pharmacokinetics , Drug Design , Drug Repositioning , Drug Resistance, Microbial/drug effects , Drug Resistance, Multiple/drug effects , Drug Stability , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacokinetics , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacokinetics , Mice , Microsomes, Liver/metabolism , Molecular Structure , Plasmodium falciparum/enzymology , Structure-Activity Relationship
5.
Daru ; 29(1): 73-84, 2021 Jun.
Article En | MEDLINE | ID: mdl-33537864

PURPOSE: To predict potential inhibitors of alpha-enolase to reduce plasminogen binding of Streptococcus pneumoniae (S. pneumoniae) that may lead as an orally active drug. S. pneumoniae remains dominant in causing invasive diseases. Fibrinolytic pathway is a critical factor of S. pneumoniae to invade and progression of disease in the host body. Besides the low mass on the cell surface, alpha-enolase possesses significant plasminogen binding among all exposed proteins. METHODS: In-silico based drug designing approach was implemented for evaluating potential inhibitors against alpha-enolase based on their binding affinities, energy score and pharmacokinetics. Lipinski's rule of five (LRo5) and Egan's (Brain Or IntestinaL EstimateD) BOILED-Egg methods were executed to predict the best ligand for biological systems. RESULTS: Molecular docking analysis revealed, Sodium (1,5-dihydroxy-2-oxopyrrolidin-3-yl)-hydroxy-dioxidophosphanium (SF-2312) as a promising inhibitor that fabricates finest attractive charges and conventional hydrogen bonds with S. pneumoniae alpha-enolase. Moreover, the pharmacokinetics of SF-2312 predict it as a therapeutic inhibitor for clinical trials. Like SF-2312, phosphono-acetohydroxamate (PhAH) also constructed adequate interactions at the active site of alpha-enolase, but it predicted less favourable than SF-2312 based on binding affinity. CONCLUSION: Briefly, SF-2312 and PhAH ligands could inhibit the role of alpha-enolase to restrain plasminogen binding, invasion and progression of S. pneumoniae. As per our investigation and analysis, SF-2312 is the most potent naturally existing inhibitor of S. pneumoniae alpha-enolase in current time.


Phosphopyruvate Hydratase/chemistry , Streptococcus pneumoniae/enzymology , Administration, Oral , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacokinetics , Molecular Docking Simulation , Organophosphonates/chemistry , Organophosphonates/pharmacokinetics , Phosphonoacetic Acid/analogs & derivatives , Phosphonoacetic Acid/chemistry , Phosphonoacetic Acid/pharmacokinetics , Phosphopyruvate Hydratase/antagonists & inhibitors , Phosphopyruvate Hydratase/metabolism , Pneumococcal Infections/drug therapy , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacokinetics
6.
J Med Chem ; 64(5): 2691-2704, 2021 03 11.
Article En | MEDLINE | ID: mdl-33576627

Histone deacetylase 6 (HDAC6) is involved in multiple regulatory processes, ranging from cellular stress to intracellular transport. Inhibition of aberrant HDAC6 activity in several cancers and neurological diseases has been shown to be efficacious in both preclinical and clinical studies. While selective HDAC6 targeting has been pursued as an alternative to pan-HDAC drugs, identifying truly selective molecular templates has not been trivial. Herein, we report a structure-activity relationship study yielding TO-317, which potently binds HDAC6 catalytic domain 2 (Ki = 0.7 nM) and inhibits the enzyme function (IC50 = 2 nM). TO-317 exhibits 158-fold selectivity for HDAC6 over other HDAC isozymes by binding the catalytic Zn2+ and, uniquely, making a never seen before direct hydrogen bond with the Zn2+ coordinating residue, His614. This novel structural motif targeting the second-sphere His614 interaction, observed in a 1.84 Å resolution crystal structure with drHDAC6 from zebrafish, can provide new pharmacophores for identifying enthalpically driven, high-affinity, HDAC6-selective inhibitors.


Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Sulfonamides/pharmacology , Animals , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacokinetics , Male , Mice, Inbred BALB C , Molecular Docking Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
7.
Acta Pharmacol Sin ; 42(7): 1124-1138, 2021 Jul.
Article En | MEDLINE | ID: mdl-32811965

Intestinal fibrosis is a common complication of inflammatory bowel disease (IBD) and is defined as an excessive accumulation of scar tissue in the intestinal wall. Intestinal fibrosis occurs in both forms of IBD: ulcerative colitis and Crohn's disease. Small-molecule inhibitors targeting hypoxia-inducing factor (HIF) prolyl-hydroxylases are promising for the development of novel antifibrotic therapies in IBD. Herein, we evaluated the therapeutic efficacy of hydroxamate of betulinic acid (BHA), a hypoxia mimetic derivative of betulinic acid, against IBD in vitro and in vivo. We showed that BAH (5-20 µM) dose-dependently enhanced collagen gel contraction and activated the HIF pathway in NIH-3T3 fibroblasts; BAH treatment also prevented the loss of trans-epithelial electrical resistance induced by proinflammatory cytokines in Caco-2 cells. In two different murine models (TNBS- and DSS-induced IBD) that cause colon fibrosis, oral administration of BAH (20, 50 mg/kg·d, for 17 days) prevented colon inflammation and fibrosis, as detected using immunohistochemistry and qPCR assays. BAH-treated animals showed a significant reduction of fibrotic markers (Tnc, Col1a2, Col3a1, Timp-1, α-SMA) and inflammatory markers (F4/80+, CD3+, Il-1ß, Ccl3) in colon tissue, as well as an improvement in epithelial barrier integrity and wound healing. BHA displayed promising oral bioavailability, no significant activity against a panel of 68 potential pharmacological targets and was devoid of genotoxicity and cardiotoxicity. Taken together, our results provide evidence that oral administration of BAH can alleviate colon inflammation and colitis-associated fibrosis, identifying the enhancement of colon barrier integrity as a possible mechanism of action, and providing a solid rationale for additional clinical studies.


Anti-Inflammatory Agents/therapeutic use , Fibrosis/prevention & control , Hydroxamic Acids/therapeutic use , Inflammation/prevention & control , Inflammatory Bowel Diseases/complications , Pentacyclic Triterpenes/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacokinetics , Caco-2 Cells , Colon/drug effects , Colon/pathology , Dextran Sulfate , Fibrosis/etiology , Fibrosis/pathology , Gastrointestinal Agents/pharmacokinetics , Gastrointestinal Agents/therapeutic use , Gene Expression/drug effects , Humans , Hydroxamic Acids/pharmacokinetics , Inflammation/etiology , Inflammation/pathology , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NIH 3T3 Cells , Pentacyclic Triterpenes/pharmacokinetics , Trinitrobenzenesulfonic Acid , Betulinic Acid
8.
Sci Rep ; 10(1): 21314, 2020 12 04.
Article En | MEDLINE | ID: mdl-33277582

An intra-hippocampus injection of kainic acid serves as a model of status epilepticus and the subsequent development of temporal lobe epilepsy. Matrix metalloproteinase-9 (MMP-9) is an enzyme that controls remodeling of the extracellular milieu under physiological and pathological conditions. In response to brain insult, MMP-9 contributes to pathological synaptic plasticity that may play a role in the progression of an epileptic condition. Marimastat is a metalloproteinase inhibitor that was tested in clinical trials of cancer. The present study assessed whether marimastat can impair the development of epilepsy. The inhibitory efficacy of marimastat was initially tested in neuronal cultures in vitro. As a marker substrate, we used nectin-3. Next, we investigated the blood-brain barrier penetration of marimastat using mass spectrometry and evaluated the therapeutic potential of marimastat against seizure outcomes. We found that marimastat inhibited the cleavage of nectin-3 in hippocampal neuronal cell cultures. Marimastat penetrated the blood-brain barrier and exerted an inhibitory effect on metalloproteinase activity in the brain. Finally, marimastat decreased some seizure parameters, such as seizure score and number, but did not directly affect status epilepticus. The long-term effects of marimastat were evident up to 6 weeks after kainic acid administration, in which marimastat still inhibited seizure duration.


Hydroxamic Acids/therapeutic use , Matrix Metalloproteinase Inhibitors/therapeutic use , Status Epilepticus/drug therapy , Animals , Blood-Brain Barrier/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Hydroxamic Acids/pharmacokinetics , Kainic Acid , Male , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacokinetics , Mice, Inbred C57BL , Nectins/metabolism
9.
Int J Mol Sci ; 21(13)2020 Jul 03.
Article En | MEDLINE | ID: mdl-32635347

Hemoglobin, heme and iron are implicated in the progression of atherosclerosis. Therefore, we investigated whether the hydrophobic fungal iron chelator siderophore, desferricoprogen (DFC) inhibits atherosclerosis. DFC reduced atherosclerotic plaque formation in ApoE-/- mice on an atherogenic diet. It lowered the plasma level of oxidized LDL (oxLDL) and inhibited lipid peroxidation in aortic roots. The elevated collagen/elastin content and enhanced expression of adhesion molecule VCAM-1 were decreased. DFC diminished oxidation of Low-density Lipoprotein (LDL) and plaque lipids catalyzed by heme or hemoglobin. Formation of foam cells, uptake of oxLDL by macrophages, upregulation of CD36 and increased expression of TNF-α were reduced by DFC in macrophages. TNF-triggered endothelial cell activation (vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecules (ICAMs), E-selectin) and increased adhesion of monocytes to endothelium were attenuated. The increased endothelial permeability and intracellular gap formation provoked by TNF-α was also prevented by DFC. DFC acted as a cytoprotectant in endothelial cells and macrophages challenged with a lethal dose of oxLDL and lowered the expression of stress-responsive heme oxygenase-1 as sublethal dose was employed. Saturation of desferrisiderophore with iron led to the loss of the beneficial effects. We demonstrated that DFC accumulated within the atheromas of the aorta in ApoE-/- mice. DFC represents a novel therapeutic approach to control the progression of atherosclerosis.


Diketopiperazines/pharmacology , Hydroxamic Acids/pharmacology , Plaque, Atherosclerotic/prevention & control , Siderophores/pharmacology , Animals , Aorta/diagnostic imaging , Aorta/drug effects , Aorta/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Diet, Atherogenic , Diketopiperazines/pharmacokinetics , Disease Models, Animal , Disease Progression , Foam Cells/drug effects , Foam Cells/pathology , Heme/metabolism , Hydroxamic Acids/pharmacokinetics , Lipid Peroxidation/drug effects , Lipoproteins, LDL/metabolism , Macrophage Activation/drug effects , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Neurospora crassa/chemistry , Oxidative Stress/drug effects , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Positron-Emission Tomography , Siderophores/pharmacokinetics
10.
Eur J Med Chem ; 201: 112411, 2020 Sep 01.
Article En | MEDLINE | ID: mdl-32615502

Dysregulated Histone Deacetylase (HDAC) activity across multiple human pathologies have highlighted this family of epigenetic enzymes as critical druggable targets, amenable to small molecule intervention. While efficacious, current approaches using non-selective HDAC inhibitors (HDACi) have been shown to cause a range of undesirable clinical toxicities. To circumvent this, recent efforts have focused on the design of highly selective HDACi as a novel therapeutic strategy. Beyond roles in regulating transcription, the unique HDAC6 (with two catalytic domains) regulates the deacetylation of α-tubulin; promoting growth factor-controlled cell motility, cell division, and metastatic hallmarks. Recent studies have linked aberrant HDAC6 function in various hematological cancers including acute myeloid leukaemia and multiple myeloma. Herein, we report the discovery, in vitro characterization, and biological evaluation of PTG-0861 (JG-265), a novel HDAC6-selective inhibitor with strong isozyme-selectivity (∼36× ) and low nanomolar potency (IC50 = 5.92 nM) against HDAC6. This selectivity profile was rationalized via in silico docking studies and also observed in cellulo through cellular target engagement. Moreover, PTG-0861 achieved relevant potency against several blood cancer cell lines (e.g. MV4-11, MM1S), whilst showing limited cytotoxicity against non-malignant cells (e.g. NHF, HUVEC) and CD-1 mice. In examining compound stability and cellular permeability, PTG-0861 revealed a promising in vitro pharmacokinetic (PK) profile. Altogether, in this study we identified a novel and potent HDAC6-selective inhibitor (∼4× more selective than current clinical standards - citarinostat, ricolinostat), which achieves cellular target engagement, efficacy in hematological cancer cells with a promising safety profile and in vitro PK.


Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Benzamides/chemical synthesis , Benzamides/metabolism , Benzamides/pharmacokinetics , Catalytic Domain , Cell Line, Tumor , Histone Deacetylase 6/chemistry , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacokinetics , Male , Mice , Molecular Docking Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship
11.
Biochim Biophys Acta Gen Subj ; 1864(10): 129670, 2020 10.
Article En | MEDLINE | ID: mdl-32565293

BACKGROUND: Microorganisms produce siderophores in order to scavenge iron from the environment and this study focuses on the characterization of the two siderophores secreted by Bacillus megaterium. The general biological properties and pharmacokinetics following oral application of these compounds are reported. METHODS: Under optimized culture conditions, the siderophores were harvested, purified by chromatography and identified using LC-MS and NMR. Two dihydroxamate siderophores were isolated, schizokinen (MW = 420) and schizokinen imide (MW = 402). RESULTS: Both compounds demonstrate strong antioxidant activity and were found to be relatively nontoxic to both human hepatocellular carcinoma (Huh7) and peripheral blood mononuclear cells. The siderophores possess a strong affinity for iron(III) and decrease the levels of the labile iron pool (LIP) in iron-loaded cells in a concentration-dependent manner. Schizokinen, was detected as both the free siderophore and the iron complex in the plasma and urine of rats after oral gavage. CONCLUSIONS: However, the bioavailability was low and thus schizokinen, like deferoxamine, has no potential as an orally active iron chelator for the treatment of systemic iron overload. GENERAL SIGNIFICANCE: By virtue of the high affinity of schizokinen for tribasic metals, this siderophore does have considerable potential for the chelation of gallium(III) and the development of clinical diagnostic reagents.


Antioxidants/chemistry , Antioxidants/pharmacology , Bacillus megaterium/chemistry , Siderophores/chemistry , Siderophores/pharmacology , Animals , Antioxidants/pharmacokinetics , Cell Line, Tumor , Cells, Cultured , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/pharmacology , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacokinetics , Iron Chelating Agents/pharmacology , Male , Rats, Sprague-Dawley , Siderophores/pharmacokinetics
12.
Mol Pharm ; 17(7): 2599-2611, 2020 07 06.
Article En | MEDLINE | ID: mdl-32379457

Parenteral chemotherapy is usually administered intravenously, although patient preference and health economics suggest the subcutaneous (sc) route could be an attractive alternative. However, due to the low aqueous solubility of hydrophobic drugs and injection volume limitations, the total amount of drug that can be administered in a single sc injection is frequently insufficient. We have developed hyaluronidase coated nanoparticles (NPs) that efficiently encapsulate such drugs, thus addressing both issues and allowing sufficient amounts of hydrophobic drug to be administered and absorbed effectively. CUDC-101, a poorly water-soluble multitargeted anticancer drug that simultaneously inhibits the receptor tyrosine kinases (RTKs) EGFR and HER2, as well as histone deacetylase (HDAC), was encapsulated in polymeric Molecular Envelope Technology (MET) NPs. The role of polymer chemistry, formulation parameters, and coating with hyaluronidase (HYD) on MET-CUDC-101 NP formulations was examined and optimized to yield high drug loading and colloidal stability, and, after freeze-drying, stable storage at room temperature for up to 90 days. The pharmacokinetic studies in healthy rats showed that plasma AUC0-24h after sc administration correlates tightly with formulation physical chemistry, specifically in vitro colloidal stability. Compared to uncoated NPs, the HYD-coating doubled the drug plasma exposure. In a murine A431 xenograft model, the coated HYD-MET-CUDC-101 NPs at a dose equivalent to 90 mg kg-1 CUDC-101 increased the survival time from 15 days (control animals treated with hyaluronidase alone) to 43 days. Polymer MET nanoparticles coated with hyaluronidase enabled the subcutaneous delivery of a hydrophobic drug with favorable therapeutic outcomes.


Antineoplastic Agents/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Drug Delivery Systems/methods , Histone Deacetylase Inhibitors/pharmacology , Hyaluronoglucosaminidase/chemistry , Hydroxamic Acids/administration & dosage , Nanoparticles/chemistry , Polymers/chemistry , Quinazolines/administration & dosage , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chitosan/analogs & derivatives , Chitosan/chemistry , Drug Carriers/chemistry , Female , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/blood , Histone Deacetylases , Histones/metabolism , Hydrophobic and Hydrophilic Interactions , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacokinetics , Mice , Mice, Nude , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Particle Size , Quinazolines/blood , Quinazolines/pharmacokinetics , Rats , Solubility , Xenograft Model Antitumor Assays
13.
Invest New Drugs ; 38(4): 977-989, 2020 08.
Article En | MEDLINE | ID: mdl-31493129

Cancer treatment is challenging, mainly due to high levels of drug toxicity and the resistance of tumours to chemotherapy. Hydroxamic acid derivatives have recently aroused attention due to their potential to treat malignancies. In the present study, we sought to investigate the anticancer effects of a new series of synthetic acetohydroxamates. The in vitro cytotoxic and antiproliferative effects of 11 synthetic acetohydroxamates were evaluated against the melanoma cell line A375. Apoptosis, cell cycle, and autophagy assays were employed to elucidate the cell death pathways induced by the compounds. The in vivo pharmacokinetic profiles of the most promising compounds were determined in CD-1 mice, while the in vivo antitumour efficacies were evaluated using the A375 melanoma xenograft model in nude mice. MTT assays revealed that all compounds presented concentration-dependent cytotoxicity against the A375 cell line. AKS 61 produced the most favourable antiproliferative activity according to the sulphorhodamine B and clonogenic assays. AKS 61 treatment resulted in decreased mitochondrial membrane potential and increased apoptosis and autophagy in the A375 cell line. However, AKS 61 failed to prevent in vivo tumour growth in a melanoma xenograft, whereas compound AKS 7 was able to inhibit tumour growth when administered orally. These in vivo findings may be explained by a more favourable pharmacokinetic profile presented by AKS 7 when compared to AKS 61. Taken together, these results suggest that acetohydroxamates have potential anticancer effects and will guide future optimisation of these molecules to allow for further non-clinical development.


Antineoplastic Agents/therapeutic use , Hydroxamic Acids/therapeutic use , Melanoma/drug therapy , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/pharmacology , Male , Melanoma/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Tumor Burden/drug effects
14.
J Labelled Comp Radiopharm ; 63(2): 85-95, 2020 02.
Article En | MEDLINE | ID: mdl-31881107

Histone deacetylase 6 (HDAC6) is a unique member of the HDAC family because of its characteristics, namely, its cytoplasmic localization and ubiquitin binding. HDAC6 has been implicated in cancer metastasis and neurodegeneration. In the present study, we performed radiosynthesis and biological evaluation of a fluorine-18-labeled ligand [18 F]3, which is an analog of the HDAC6-selective inhibitor tubastatin A, for positron emission tomography (PET) imaging. [18 F]3 was synthesized by a two-step reaction composed of 18 F-fluorination and formation of a hydroxamic acid group. IC50 values of 3 against HDAC1 and HDAC6 activities were 996 nM and 33.1 nM, respectively. A biodistribution study in mice demonstrated low brain uptake of [18 F]3. Furthermore, bone radioactivity was stable at around 2% ID/g after injection, suggesting high tolerance to defluorination. Regarding metabolic stability, 70% of the compound was observed as the unchanged form at 30 minutes post injection in mouse plasma. A small animal PET study in mice showed that pretreatment with cyclosporine A had no effect on initial brain uptake of [18 F]3, suggesting low brain uptake of [18 F]3 was not caused by the P-glycoprotein-mediated efflux. While PET imaging using [18 F]3 has a limitation with respect to neurodegenerative diseases, further studies evaluating its utility for certain cancers are worth evaluating.


Fluorine Radioisotopes , Histone Deacetylase 6/metabolism , Hydroxamic Acids/chemistry , Hydroxamic Acids/chemical synthesis , Indoles/chemistry , Indoles/chemical synthesis , Positron-Emission Tomography , Animals , Cell Line, Tumor , Hydroxamic Acids/pharmacokinetics , Indoles/pharmacokinetics , Mice , Tissue Distribution
15.
Mol Imaging Biol ; 22(3): 569-577, 2020 06.
Article En | MEDLINE | ID: mdl-31290052

PURPOSE: With the emerging knowledge about the impact of epigenetic alterations on behavior and brain disorders, the ability to measure epigenetic alterations in brain tissue in vivo has become critically important. We present the first in vivo/in vitro cross-validation of the novel positron emission tomography (PET) radioligand [11C]Martinostat in the pig brain with regard to its ability to measure histone deacetylase 1-3 (HDAC1-3) levels in vivo. PROCEDURES: Nine female Danish landrace pigs underwent 121-min dynamic PET scans with [11C]Martinostat. We quantified [11C]Martinostat uptake using both a simple ratio method and kinetic models with arterial input function. By the end of the scan, the animals were euthanized and the brains were extracted. We measured HDAC1-3 protein levels in frontal cortex, cerebellum vermis, and hippocampus and compared the protein levels and regional outcome values to the [11C]Martinostat PET quantification. RESULTS: [11C]Martinostat distributed widely across brain regions, with the highest uptake in the cerebellum vermis and the lowest in the olfactory bulbs. Based on the Akaike information criterion, the quantification was most reliably performed by Ichise MA1 kinetic modeling, but since the radioligand displayed very slow kinetics, we also calculated standard uptake value (SUV) ratios which correlated well with VT. The western blots revealed higher brain tissue protein levels of HDAC1/2 compared to HDAC3, and HDAC1 and HDAC2 levels were highly correlated in all three investigated brain regions. The in vivo SUV ratio measure correlated well with the in vitro HDAC1-3 levels, whereas no correlation was found between VT values and HDAC levels. CONCLUSIONS: We found good correlation between in vivo measured SUV ratios and in vitro measures of HDAC 1-3 proteins, supporting that [11C]Martinostat provides a good in vivo measure of the cerebral HDAC1-3 protein levels.


Adamantane/analogs & derivatives , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes/pharmacokinetics , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacokinetics , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Adamantane/pharmacokinetics , Animals , Brain/enzymology , Carbon Radioisotopes/chemistry , Female , Humans , Models, Animal , Radiopharmaceuticals/chemistry , Swine , Tissue Distribution
16.
J Med Chem ; 63(1): 66-87, 2020 01 09.
Article En | MEDLINE | ID: mdl-31804826

UDP-3-O-((R)-3-hydroxymyristoyl)-N-glucosamine deacetylase (LpxC) is as an attractive target for the discovery and development of novel antibacterial drugs to address the critical medical need created by multidrug resistant Gram-negative bacteria. By using a scaffold hopping approach on a known family of methylsulfone hydroxamate LpxC inhibitors, several hit series eliciting potent antibacterial activities against Enterobacteriaceae and Pseudomonas aeruginosa were identified. Subsequent hit-to-lead optimization, using cocrystal structures of inhibitors bound to Pseudomonas aeruginosa LpxC as guides, resulted in the discovery of multiple chemical series based on (i) isoindolin-1-ones, (ii) 4,5-dihydro-6H-thieno[2,3-c]pyrrol-6-ones, and (iii) 1,2-dihydro-3H-pyrrolo[1,2-c]imidazole-3-ones. Synthetic methods, antibacterial activities and relative binding affinities, as well as physicochemical properties that allowed compound prioritization are presented. Finally, in vivo properties of lead molecules which belong to the most promising pyrrolo-imidazolone series, such as 18d, are discussed.


Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Escherichia coli Infections/drug therapy , Gram-Negative Bacteria/drug effects , Hydroxamic Acids/therapeutic use , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacokinetics , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Escherichia coli/drug effects , Female , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacokinetics , Klebsiella pneumoniae/drug effects , Mice, Inbred ICR , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , Pyrroles/chemical synthesis , Pyrroles/pharmacokinetics , Pyrroles/therapeutic use
17.
J Med Chem ; 63(1): 88-102, 2020 01 09.
Article En | MEDLINE | ID: mdl-31804829

LpxC inhibitors were optimized starting from lead compounds with limited efficacy and solubility and with the goal to provide new options for the treatment of serious infections caused by Gram-negative pathogens in hospital settings. To enable the development of an aqueous formulation for intravenous administration of the drug at high dose, improvements in both solubility and antibacterial activity in vivo were prioritized early on. This lead optimization program resulted in the discovery of compounds such as 13 and 30, which exhibited high solubility and potent efficacy against Gram-negative pathogens in animal infection models.


Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Escherichia coli Infections/drug therapy , Hydroxamic Acids/therapeutic use , Administration, Intravenous , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacokinetics , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Gram-Negative Bacteria/drug effects , Hepatocytes/metabolism , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacokinetics , Mice , Microbial Sensitivity Tests , Molecular Structure , Rats , Solubility
18.
Neurochem Res ; 44(11): 2460-2469, 2019 Nov.
Article En | MEDLINE | ID: mdl-31571096

Chemotherapy-related cognitive impairment (CRCI) is a potential long-term side effect during cancer treatment. There are currently no effective treatments for CRCI. Reduction or inhibition of histone deacetylase 6 (HDAC6) has been considered a possible therapeutic strategy for cognitive deficits. HDAC6 inhibition recently has been shown to reverse chemotherapy-induced peripheral neuropathy effectively. In the present study, we examined the effect of HDAC6 inhibitor ACY-1215 (Ricolinostat) on cisplatin-induced brain damage and cognitive deficits in mice. Our results showed that ACY-1215 ameliorated behavioral deficits and dendritic spine loss and increased synaptic density in cisplatin-treated mice. Mechanistically, HDAC6 inhibitor ACY-1215 enhanced α-tubulin acetylation in the hippocampus of cisplatin-treated mice. Furthermore, ACY-1215 recovered cisplatin-induced impaired mitochondrial transport and mitochondrial dysfunction in the hippocampus. Our results suggest that inhibition of HDAC6 improves established cisplatin-induced cognitive deficits by the restoration of mitochondrial and synaptic impairments. These results offer prospective approaches for CRCI, especially because ACY1215 currently serves as an add-on cancer therapy during clinical trials.


Cognitive Dysfunction/prevention & control , Hydroxamic Acids/therapeutic use , Neuroprotective Agents/therapeutic use , Pyrimidines/therapeutic use , Animals , Cisplatin , Cognitive Dysfunction/chemically induced , Dendritic Spines/drug effects , Hippocampus/drug effects , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/pharmacokinetics , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Neuroprotective Agents/pharmacokinetics , Pyrimidines/pharmacokinetics
19.
Cancer Chemother Pharmacol ; 84(6): 1201-1208, 2019 12.
Article En | MEDLINE | ID: mdl-31522242

PURPOSE: The reported maximum tolerated dose (MTD) of single-agent belinostat is 1000 mg/m2 given days 1-5, every 21 days. Pre-clinical evidence suggests histone deacetylase inhibitors enhance retinoic acid signaling in a variety of solid tumors. We conducted a phase I study of belinostat combined with 50-100 mg/m2/day 13-cis-retinoic acid (13-cRA) in patients with advanced solid tumors. METHODS: Belinostat was administered days 1-5 and 13-cRA days 1-14, every 21 days. Dose-limiting toxicity (DLT) was defined as cycle 1 hematologic toxicity grade ≥ 3 not resolving to grade ≤ 1 within 1 week or non-hematologic toxicity grade ≥ 3 (except controlled nausea and vomiting and transient liver function abnormalities) attributable to belinostat. RESULTS: Among 51 patients, two DLTs were observed: grade 3 hypersensitivity with dizziness and hypoxia at 1700 mg/m2/day belinostat with 100 mg/m2/day 13-cRA, and grade 3 allergic reaction at 2000 mg/m2/day belinostat with 100 mg/m2/day 13-cRA. The MTD was not reached. Pharmacokinetics of belinostat may be non-linear at high doses. Ten patients had stable disease, including one with neuroendocrine pancreatic cancer for 56 cycles, one with breast cancer for 12 cycles, and one with lung cancer for 8 cycles. Partial responses included a patient with keratinizing squamous cell carcinoma of the tonsils, and a patient with lung cancer. CONCLUSIONS: The combination of belinostat 2000 mg/m2 days 1-5 and 13-cRA 100 mg/m2 days 1-14, every 21 days, was well-tolerated and an MTD was not reached despite doubling the established single-agent MTD of belinostat.


Antineoplastic Combined Chemotherapy Protocols/toxicity , Hydroxamic Acids/toxicity , Isotretinoin/toxicity , Maximum Tolerated Dose , Neoplasms/drug therapy , Sulfonamides/toxicity , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacokinetics , Infusions, Intravenous , Isotretinoin/administration & dosage , Isotretinoin/pharmacokinetics , Male , Middle Aged , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics
20.
ACS Chem Neurosci ; 10(8): 3769-3777, 2019 08 21.
Article En | MEDLINE | ID: mdl-31330099

Histone deacetylases (HDACs) are enzymes involved in the epigenetic control of gene expression. A handful of HDAC inhibitors have been approved for the treatment of cancer, and HDAC inhibition has also been proposed as a novel therapeutic strategy for neurodegenerative disorders. These disorders include progranulin (PGRN)-deficient forms of frontotemporal dementia caused by mutations in the GRN gene that lead to haploinsufficiency. Hydroxamic-acid-based inhibitors of HDACs 1-3, reported to have fast-on/fast-off binding kinetics, induce increased expression of PGRN in human neuronal models, while the benzamide class of slow-binding HDAC inhibitors does not produce this effect. These observations indicate that the kinetics of HDAC inhibitor binding can be tuned for optimal induction of human PGRN expression in neurons. Here, we further expand on these findings using human cortical-like, glutamatergic neurons. We provide evidence that two prototypical, potent hydroxamic acid HDAC inhibitors that induce PGRN (panobinostat and trichostatin A) exhibit an initial fast-binding step followed by a second, slower step, referred to as mechanism B of slow binding, rather than simpler fast-on/fast-off binding kinetics. In addition, we show that trapoxin A, a macrocyclic, epoxyketone-containing class I HDAC inhibitor, exhibits slow binding with high, picomolar potency and also induces PGRN expression in human neurons. Finally, we demonstrate induction of PGRN expression by fast-on/fast-off, highly potent, macrocyclic HDAC inhibitors with ethyl ketone or ethyl ester Zn2+ binding groups. Taken together, these data expand our understanding of HDAC1-3 inhibitor binding kinetics, and further delineate the specific combinations of structural and kinetic features of HDAC inhibitors that are optimal for upregulating PGRN expression in human neurons and thus may have translational relevance in neurodegenerative disease.


Histone Deacetylase Inhibitors/pharmacokinetics , Hydroxamic Acids/pharmacokinetics , Neurons/drug effects , Panobinostat/pharmacokinetics , Progranulins/metabolism , Frontotemporal Dementia/metabolism , Gene Expression/drug effects , Glutamic Acid/metabolism , Histone Deacetylases/metabolism , Humans , Neural Stem Cells , Neurons/metabolism , Peptides/pharmacokinetics
...