Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(11): 7374-7386, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37216489

ABSTRACT

Leishmaniasis, a neglected tropical disease caused by Leishmania species parasites, annually affects over 1 million individuals worldwide. Treatment options for leishmaniasis are limited due to high cost, severe adverse effects, poor efficacy, difficulty of use, and emerging drug resistance to all approved therapies. We discovered 2,4,5-trisubstituted benzamides (4) that possess potent antileishmanial activity but poor aqueous solubility. Herein, we disclose our optimization of the physicochemical and metabolic properties of 2,4,5-trisubstituted benzamide that retains potency. Extensive structure-activity and structure-property relationship studies allowed selection of early leads with suitable potency, microsomal stability, and improved solubility for progression. Early lead 79 exhibited an 80% oral bioavailability and potently blocked proliferation of Leishmania in murine models. These benzamide early leads are suitable for development as orally available antileishmanial drugs.


Subject(s)
Antiprotozoal Agents , Leishmania , Leishmaniasis , Humans , Animals , Mice , Leishmaniasis/drug therapy , Leishmaniasis/chemically induced , Leishmaniasis/parasitology , Antiprotozoal Agents/chemistry , Benzamides/pharmacology , Benzamides/therapeutic use
2.
EBioMedicine ; 80: 104065, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35598441

ABSTRACT

BACKGROUND: SJ733, a newly developed inhibitor of P. falciparum ATP4, has a favorable safety profile and rapid antiparasitic effect but insufficient duration to deliver a single-dose cure of malaria. We investigated the safety, tolerability, and pharmacokinetics of a multidose SJ733 regimen and a single-dose pharmacoboost approach using cobicistat to inhibit CYP3A4, thereby increasing exposure. METHODS: Two multidose unboosted cohorts (n = 9) (SJ733, 300 mg and 600 mg daily for 3 days) followed by three single-dose boosted cohorts combining SJ733 (n = 18) (75-, 300-, or 600-mg single dose) with cobicistat (150-mg single dose) as a pharmacokinetic booster were evaluated in healthy volunteers (ClinicalTrials.gov: NCT02661373). FINDINGS: All participants tolerated SJ733 well, with no serious adverse events (AEs), dose-limiting toxicity, or clinically significant electrocardiogram or laboratory test findings. All reported AEs were Grade 1, clinically insignificant, and considered unlikely or unrelated to SJ733. Compared to unboosted cohorts, the SJ733/cobicistat-boosted cohorts showed a median increase in area under the curve and maximum concentration of 3·9 × and 2·6 ×, respectively, and a median decrease in the ratio of the major CYP3A-produced metabolite SJ506 to parent drug of 4·6 × . Incorporating these data in a model of parasite dynamics indicated that a 3-day regimen of SJ733/cobicistat (600 mg/150 mg daily) relative to a single 600-mg dose ± cobicistat would increase parasite clearance from 106 to 1012 parasites/µL. INTERPRETATION: The multidose and pharmacoboosted approaches to delivering SJ733 were well-tolerated and significantly increased drug exposure and prediction of cure. This study supports the further development of SJ733 and demonstrates an innovative pharmacoboost approach for an antimalarial. FUNDING: Global Health Innovative Technology Fund, Medicines for Malaria Venture, National Institutes of Health, and American Lebanese Syrian Associated Charities.


Subject(s)
Antimalarials , Folic Acid Antagonists , Malaria, Falciparum , Malaria , Antimalarials/adverse effects , Cobicistat/therapeutic use , Heterocyclic Compounds, 4 or More Rings , Humans , Isoquinolines , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Plasmodium falciparum
3.
J Med Chem ; 64(16): 12152-12162, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34355566

ABSTRACT

Leishmaniasis, a disease caused by protozoa of the Leishmania species, afflicts roughly 12 million individuals worldwide. Most existing drugs for leishmaniasis are toxic, expensive, difficult to administer, and subject to drug resistance. We report a new class of antileishmanial leads, the 3-arylquinolines, that potently block proliferation of the intramacrophage amastigote form of Leishmania parasites with good selectivity relative to the host macrophages. Early lead 34 was rapidly acting and possessed good potency against L. mexicana (EC50 = 120 nM), 30-fold selectivity for the parasite relative to the macrophage (EC50 = 3.7 µM), and also blocked proliferation of Leishmania donovani parasites resistant to antimonial drugs. Finally, another early lead, 27, which exhibited reasonable in vivo tolerability, impaired disease progression during the dosing period in a murine model of cutaneous leishmaniasis. These results suggest that the arylquinolines provide a fruitful departure point for the development of new antileishmanial drugs.


Subject(s)
Leishmaniasis, Cutaneous/drug therapy , Quinolines/therapeutic use , Trypanocidal Agents/therapeutic use , Animals , Female , Leishmania/drug effects , Mice, Inbred BALB C , Microsomes, Liver/metabolism , Molecular Structure , Quinolines/chemical synthesis , Quinolines/metabolism , Quinolines/pharmacokinetics , Structure-Activity Relationship , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/metabolism , Trypanocidal Agents/pharmacokinetics
4.
Bioorg Med Chem Lett ; 47: 128216, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34157390

ABSTRACT

Malaria remains one of the deadliest infectious diseases worldwide and continues to infect hundreds of millions of individuals each year. Here we report the discovery and derivatization of a series of 2,6-dibenzylidenecyclohexanones targeting the chloroquine-sensitive 3D7 strain of Plasmodium falciparum . While the initial lead compound displayed significant toxicity in a human cell proliferation assay, we were able to identify a derivative with no detectable toxicity and sub-micromolar potency.


Subject(s)
Antimalarials/pharmacology , Chloroquine/pharmacology , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cell Proliferation/drug effects , Chloroquine/chemical synthesis , Chloroquine/chemistry , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Humans , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship
5.
J Med Chem ; 64(9): 5850-5862, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33945681

ABSTRACT

The cullin-RING ubiquitin ligases (CRLs) are ubiquitin E3 enzymes that play a key role in controlling proteasomal degradation and are activated by neddylation. We previously reported inhibitors that target CRL activation by disrupting the interaction of defective in cullin neddylation 1 (DCN1), a CRL neddylation co-E3, and UBE2M, a neddylation E2. Our first-generation inhibitors possessed poor oral bioavailability and fairly rapid clearance that hindered the study of acute inhibition of DCN-controlled CRL activity in vivo. Herein, we report studies to improve the pharmacokinetic performance of the pyrazolo-pyridone inhibitors. The current best inhibitor, 40, inhibits the interaction of DCN1 and UBE2M, blocks NEDD8 transfer in biochemical assays, thermally stabilizes cellular DCN1, and inhibits anchorage-independent growth in a DCN1 amplified squamous cell carcinoma cell line. Additionally, we demonstrate that a single oral 50 mg/kg dose sustains plasma exposures above the biochemical IC90 for 24 h in mice.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Pyrazoles/chemistry , Pyridines/chemistry , Ubiquitin-Conjugating Enzymes/metabolism , Administration, Oral , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Design , Drug Stability , Half-Life , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mice , Molecular Dynamics Simulation , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyridines/metabolism , Pyridines/pharmacology , Structure-Activity Relationship , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors
6.
Malar J ; 20(1): 107, 2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33608015

ABSTRACT

BACKGROUND: The ongoing global malaria eradication campaign requires development of potent, safe, and cost-effective drugs lacking cross-resistance with existing chemotherapies. One critical step in drug development is selecting a suitable clinical candidate from late leads. The process used to select the clinical candidate SJ733 from two potent dihydroisoquinolone (DHIQ) late leads, SJ733 and SJ311, based on their physicochemical, pharmacokinetic (PK), and toxicity profiles is described. METHODS: The compounds were tested to define their physicochemical properties including kinetic and thermodynamic solubility, partition coefficient, permeability, ionization constant, and binding to plasma proteins. Metabolic stability was assessed in both microsomes and hepatocytes derived from mice, rats, dogs, and humans. Cytochrome P450 inhibition was assessed using recombinant human cytochrome enzymes. The pharmacokinetic profiles of single intravenous or oral doses were investigated in mice, rats, and dogs. RESULTS: Although both compounds displayed similar physicochemical properties, SJ733 was more permeable but metabolically less stable than SJ311 in vitro. Single dose PK studies of SJ733 in mice, rats, and dogs demonstrated appreciable oral bioavailability (60-100%), whereas SJ311 had lower oral bioavailability (mice 23%, rats 40%) and higher renal clearance (10-30 fold higher than SJ733 in rats and dogs), suggesting less favorable exposure in humans. SJ311 also displayed a narrower range of dose-proportional exposure, with plasma exposure flattening at doses above 200 mg/kg. CONCLUSION: SJ733 was chosen as the candidate based on a more favorable dose proportionality of exposure and stronger expectation of the ability to justify a strong therapeutic index to regulators.


Subject(s)
Antimalarials/pharmacology , Isoquinolines/pharmacology , Animals , Antimalarials/pharmacokinetics , Antimalarials/toxicity , Biological Availability , Dogs , Hepatocytes/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/toxicity , Humans , Isoquinolines/pharmacokinetics , Isoquinolines/toxicity , Mice , Microsomes, Liver/drug effects , Rats
7.
Bioorg Med Chem Lett ; 35: 127818, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33513390

ABSTRACT

A virtual screen was performed to identify anti-malarial compounds targeting Plasmodium falciparum heat shock 90 protein by applying a series of drug-like and commercial availability filters to compounds in the ZINC database, resulting in a virtual library of more than 13 million candidates. The goal of the virtual screen was to identify novel compounds which could serve as a starting point for the development of antimalarials with a mode of action different from anything currently used in the clinic. The screen targeted the ATP binding pocket of the highly conserved Plasmodium heat shock 90 protein, as this protein is critical to the survival of the parasite and has several significant structural differences from the human homolog. The top twelve compounds from the virtual screen were tested in vitro, with all twelve showing no antiproliferative activity against the human fibroblast cell line and three compounds exhibiting single digit or better micromolar antiproliferative activity against the chloroquine-sensitive P. falciparum 3D7 strain.


Subject(s)
Antimalarials/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Molecular Docking Simulation , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Fibroblasts/drug effects , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/metabolism , Structure-Activity Relationship
8.
Bioorg Med Chem ; 28(22): 115758, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33007559

ABSTRACT

The 4-(heteroarylthio)thieno[2,3-d]pyrimidine (TTP) series of antimalarials, represented by 1 and 17, potently inhibit proliferation of the 3D7 strain of P. falciparum (EC50 70-100 nM), but suffer from oxidative metabolism. The 1,1-cyclopropylidene isosteres 6 and 16 were designed to obviate this drawback. They were prepared by a short route that features a combined Peterson methylenation / cyclopropanation transformation of, e. g., ketone 7. Isosteres 6 and 16 possess significantly attenuated antimalarial potency relative to parents 1 and 17. This outcome can be rationalized based on the increased out-of-plane steric demands of the latter two. In support of this hypothesis, the relatively flat ketone 7 retains some of the potency of 1, even though it appears to be a comparatively inferior mimic with respect to electronics and bond lengths and angles. We also demonstrate crystallographically and computationally an apparent increase in the strength of the intramolecular sulfur hole interaction of 1 upon protonation.


Subject(s)
Antimalarials/pharmacology , Cyclopropanes/pharmacology , Plasmodium falciparum/drug effects , Pyrimidines/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cells, Cultured , Crystallography, X-Ray , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Density Functional Theory , Dose-Response Relationship, Drug , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
9.
J Med Chem ; 63(20): 11902-11919, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32945666

ABSTRACT

Malaria remains one of the most deadly infectious diseases, causing hundreds of thousands of deaths each year, primarily in young children and pregnant mothers. Here, we report the discovery and derivatization of a series of pyrazolo[3,4-b]pyridines targeting Plasmodium falciparum, the deadliest species of the malaria parasite. Hit compounds in this series display sub-micromolar in vitro activity against the intraerythrocytic stage of the parasite as well as little to no toxicity against the human fibroblast BJ and liver HepG2 cell lines. In addition, our hit compounds show good activity against the liver stage of the parasite but little activity against the gametocyte stage. Parasitological profiles, including rate of killing, docking, and molecular dynamics studies, suggest that our compounds may target the Qo binding site of cytochrome bc1.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cell Line , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 30(21): 127502, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32822760

ABSTRACT

A series of tetrahydro-ß-carboline derivatives of a lead compound known to target the heat shock 90 protein of Plasmodium falciparum were synthesized and assayed for both potency against the parasite and toxicity against a human cell line. Using a rationalized structure based design strategy, a new lead compound with a potency two orders of magnitude greater than the original lead compound was found. Additional modeling of this new lead compound suggests multiple avenues to further increase potency against this target, potentially paving the path for a therapeutic with a mode of action different than any current clinical treatment.


Subject(s)
Adenosine Triphosphate/chemistry , Antimalarials/pharmacology , Carbolines/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Binding Sites/drug effects , Carbolines/chemical synthesis , Carbolines/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Plasmodium falciparum/chemistry , Plasmodium falciparum/cytology , Structure-Activity Relationship
11.
J Med Chem ; 62(18): 8429-8442, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31465221

ABSTRACT

Chemical control of cullin neddylation is attracting increased attention based largely on the successes of the NEDD8-activating enzyme (E1) inhibitor pevonedistat. Recently reported chemical probes enable selective and time-dependent inhibition of downstream members of the neddylation trienzymatic cascade including the co-E3, DCN1. In this work, we report the optimization of a novel class of small molecule inhibitors of the DCN1-UBE2M interaction. Rational X-ray co-structure enabled optimization afforded a 25-fold improvement in potency relative to the initial screening hit. The potency gains are largely attributed to additional hydrophobic interactions mimicking the N-terminal acetyl group that drives binding of UBE2M to DCN1. The compounds inhibit the protein-protein interaction, block NEDD8 transfer in biochemical assays, engage DCN1 in cells, and selectively reduce the steady-state neddylation of Cul1 and Cul3 in two squamous carcinoma cell lines harboring DCN1 amplification.


Subject(s)
Cullin Proteins/chemistry , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , NEDD8 Protein/chemistry , Pyrazoles/chemistry , Pyridones/chemistry , Amides/chemistry , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Cell Proliferation , Cyclopentanes/pharmacology , Drug Design , Fibroblasts/metabolism , Glycine/chemistry , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Protein Domains , Protein Interaction Mapping , Pyrimidines/pharmacology , Reactive Oxygen Species/chemistry , Structure-Activity Relationship , Ubiquitin-Conjugating Enzymes/chemistry
12.
J Med Chem ; 61(7): 2694-2706, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29547693

ABSTRACT

We previously reported the discovery, validation, and structure-activity relationships of a series of piperidinyl ureas that potently inhibit the DCN1-UBE2M interaction. We demonstrated that compound 7 inhibits both the DCN1-UBE2M protein-protein interaction and DCN1-mediated cullin neddylation in biochemical assays and reduces levels of steady-state cullin neddylation in a squamous carcinoma cell line harboring DCN1 amplification. Although compound 7 exhibits good solubility and permeability, it is rapidly metabolized in microsomal models (CLint = 170 mL/min/kg). This work lays out the discovery of an orally bioavailable analogue, NAcM-OPT (67). Compound 67 retains the favorable biochemical and cellular activity of compound 7 but is significantly more stable both in vitro and in vivo. Compound 67 is orally bioavailable, well tolerated in mice, and currently used to study the effects of acute pharmacologic inhibition of the DCN1-UBE2M interaction on the NEDD8/CUL pathway.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cullin Proteins/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Biological Availability , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Drug Discovery , Drug Screening Assays, Antitumor , Female , Humans , Intracellular Signaling Peptides and Proteins , Lung Neoplasms/drug therapy , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , NEDD8 Protein/antagonists & inhibitors , NEDD8 Protein/drug effects , Proteins , Proto-Oncogene Proteins/metabolism , Structure-Activity Relationship , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Urea/analogs & derivatives , Urea/chemistry
13.
J Med Chem ; 61(7): 2680-2693, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29547696

ABSTRACT

We previously discovered and validated a class of piperidinyl ureas that regulate defective in cullin neddylation 1 (DCN1)-dependent neddylation of cullins. Here, we report preliminary structure-activity relationship studies aimed at advancing our high-throughput screen hit into a tractable tool compound for dissecting the effects of acute DCN1-UBE2M inhibition on the NEDD8/cullin pathway. Structure-enabled optimization led to a 100-fold increase in biochemical potency and modestly increased solubility and permeability as compared to our initial hit. The optimized compounds inhibit the DCN1-UBE2M protein-protein interaction in our TR-FRET binding assay and inhibit cullin neddylation in our pulse-chase NEDD8 transfer assay. The optimized compounds bind to DCN1 and selectively reduce steady-state levels of neddylated CUL1 and CUL3 in a squamous cell carcinoma cell line. Ultimately, we anticipate that these studies will identify early lead compounds for clinical development for the treatment of lung squamous cell carcinomas and other cancers.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cullin Proteins/antagonists & inhibitors , NEDD8 Protein/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Crystallography, X-Ray , Drug Discovery , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Intracellular Signaling Peptides and Proteins , Lung Neoplasms/drug therapy , Models, Molecular , Molecular Conformation , NEDD8 Protein/metabolism , Protein Binding , Proteins , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors
14.
PLoS Negl Trop Dis ; 11(12): e0006157, 2017 12.
Article in English | MEDLINE | ID: mdl-29287089

ABSTRACT

Leishmaniasis is a parasitic infection that afflicts approximately 12 million people worldwide. There are several limitations to the approved drug therapies for leishmaniasis, including moderate to severe toxicity, growing drug resistance, and the need for extended dosing. Moreover, miltefosine is currently the only orally available drug therapy for this infection. We addressed the pressing need for new therapies by pursuing a two-step phenotypic screen to discover novel, potent, and orally bioavailable antileishmanials. First, we conducted a high-throughput screen (HTS) of roughly 600,000 small molecules for growth inhibition against the promastigote form of the parasite life cycle using the nucleic acid binding dye SYBR Green I. This screen identified approximately 2,700 compounds that inhibited growth by over 65% at a single point concentration of 10 µM. We next used this 2700 compound focused library to identify compounds that were highly potent against the disease-causing intra-macrophage amastigote form and exhibited limited toxicity toward the host macrophages. This two-step screening strategy uncovered nine unique chemical scaffolds within our collection, including two previously described antileishmanials. We further profiled two of the novel compounds for in vitro absorption, distribution, metabolism, excretion, and in vivo pharmacokinetics. Both compounds proved orally bioavailable, affording plasma exposures above the half-maximal effective concentration (EC50) concentration for at least 12 hours. Both compounds were efficacious when administered orally in a murine model of cutaneous leishmaniasis. One of the two compounds exerted potent activity against trypanosomes, which are kinetoplastid parasites related to Leishmania species. Therefore, this compound could help control multiple parasitic diseases. The promising pharmacokinetic profile and significant in vivo efficacy observed from our HTS hits highlight the utility of our two-step phenotypic screening strategy and strongly suggest that medicinal chemistry optimization of these newly identified scaffolds will lead to promising candidates for an orally available anti-parasitic drug.


Subject(s)
Antiprotozoal Agents/pharmacokinetics , Drug Evaluation, Preclinical/methods , Leishmania mexicana/drug effects , Leishmaniasis, Cutaneous/drug therapy , Administration, Oral , Animals , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/adverse effects , Antiprotozoal Agents/chemistry , Cell Line , Chemistry, Pharmaceutical , Drug Discovery , Female , Humans , Leishmania mexicana/growth & development , Leishmaniasis, Cutaneous/parasitology , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Phenotype
15.
Nat Chem Biol ; 13(8): 850-857, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28581483

ABSTRACT

N-terminal acetylation is an abundant modification influencing protein functions. Because ∼80% of mammalian cytosolic proteins are N-terminally acetylated, this modification is potentially an untapped target for chemical control of their functions. Structural studies have revealed that, like lysine acetylation, N-terminal acetylation converts a positively charged amine into a hydrophobic handle that mediates protein interactions; hence, this modification may be a druggable target. We report the development of chemical probes targeting the N-terminal acetylation-dependent interaction between an E2 conjugating enzyme (UBE2M or UBC12) and DCN1 (DCUN1D1), a subunit of a multiprotein E3 ligase for the ubiquitin-like protein NEDD8. The inhibitors are highly selective with respect to other protein acetyl-amide-binding sites, inhibit NEDD8 ligation in vitro and in cells, and suppress anchorage-independent growth of a cell line with DCN1 amplification. Overall, our data demonstrate that N-terminal acetyl-dependent protein interactions are druggable targets and provide insights into targeting multiprotein E2-E3 ligases.


Subject(s)
Enzyme Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Acetylation/drug effects , Binding Sites , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , NEDD8 Protein , Small Molecule Libraries/chemistry , Structure-Activity Relationship
16.
J Med Chem ; 60(7): 2651-2653, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28296396

ABSTRACT

The majority of frontline therapies for the treatment of malaria are combination drugs containing artemisinin (or its semisynthetic analogs), known as artemisinin combination therapies (ACTs). While generally efficacious, ACTs and the first generation fully synthetic ozonide, arterolane (OZ277, 1), suffer from rapid clearance requiring 3-day dosing regimens. Extensive structure-activity studies led to the discovery of a second-generation ozonide, artefenomel (OZ439, 2), which has overcome this limitation, maintaining the rapid onset of action and potent activity of the artemisinin derivatives while exhibiting greatly improved pharmacokinetics, low projected cost of goods, prophylactic activity, and the potential for a single dose cure.


Subject(s)
Adamantane/analogs & derivatives , Antimalarials/chemistry , Antimalarials/pharmacology , Drug Discovery , Malaria/drug therapy , Peroxides/chemistry , Peroxides/pharmacology , Plasmodium/drug effects , Adamantane/chemistry , Adamantane/pharmacology , Adamantane/therapeutic use , Animals , Antimalarials/therapeutic use , Humans , Peroxides/therapeutic use
17.
Bioorg Med Chem ; 21(18): 5673-8, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23932448

ABSTRACT

The bacterial cell division protein FtsZ polymerizes in a GTP-dependent manner to form a Z-ring that marks the plane of division. As a validated antimicrobial target, considerable efforts have been devoted to identify small molecule FtsZ inhibitors. We recently discovered the chrysophaentins, a novel suite of marine natural products that inhibit FtsZ activity in vitro. These natural products along with a synthetic hemi-chrysophaentin exhibit strong antimicrobial activity toward a broad spectrum of Gram-positive pathogens. To define their mechanisms of FtsZ inhibition and determine their in vivo effects in live bacteria, we used GTPase assays and fluorescence anisotropy to show that hemi-chrysophaentin competitively inhibits FtsZ activity. Furthermore, we developed a model system using a permeable Escherichia coli strain, envA1, together with an inducible FtsZ-yellow fluorescent protein construct to show by fluorescence microscopy that both chrysophaentin A and hemi-chrysophaentin disrupt Z-rings in live bacteria. We tested the E. coli system further by reproducing phenotypes observed for zantrins Z1 and Z3, and demonstrate that the alkaloid berberine, a reported FtsZ inhibitor, exhibits auto-fluorescence, making it incompatible with systems that employ GFP or YFP tagged FtsZ. These studies describe unique examples of nonnucleotide, competitive FtsZ inhibitors that disrupt FtsZ in vivo, together with a model system that should be useful for in vivo testing of FtsZ inhibitor leads that have been identified through in vitro screens but are unable to penetrate the Gram-negative outer membrane.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Benzyl Compounds/chemistry , Cytoskeletal Proteins/antagonists & inhibitors , Ethers, Cyclic/chemistry , Amidohydrolases/genetics , Amidohydrolases/metabolism , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzyl Compounds/chemical synthesis , Benzyl Compounds/isolation & purification , Benzyl Compounds/pharmacology , Berberine/chemistry , Berberine/pharmacology , Cytoskeletal Proteins/metabolism , Escherichia coli/drug effects , Escherichia coli/metabolism , Ethers, Cyclic/isolation & purification , Ethers, Cyclic/pharmacology , GTP Phosphohydrolases/metabolism , Gram-Positive Bacteria/drug effects , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
18.
Mar Drugs ; 10(5): 1103-1125, 2012 May.
Article in English | MEDLINE | ID: mdl-22822360

ABSTRACT

Drug-resistant Staphylococcus aureus is a continuing public health concern, both in the hospital and community settings. Antibacterial compounds that possess novel structural scaffolds and are effective against multiple S. aureus strains, including current drug-resistant ones, are needed. Previously, we have described the chrysophaentins, a family of bisdiarylbutene macrocycles from the chrysophyte alga Chrysophaeum taylori that inhibit the growth of S. aureus and methicillin-resistant S. aureus (MRSA). In this study we have analyzed the geographic variability of chrysophaentin production in C. taylori located at different sites on the island of St. John, U.S. Virgin Islands, and identified two new linear chrysophaentin analogs, E2 and E3. In addition, we have expanded the structure activity relationship through synthesis of fragments comprising conserved portions of the chrysophaentins, and determined the antimicrobial activity of natural chrysophaentins and their synthetic analogs against five diverse S. aureus strains. We find that the chrysophaentins show similar activity against all S. aureus strains, regardless of their drug sensitivity profiles. The synthetic chrysophaentin fragments indeed mimic the natural compounds in their spectrum of antibacterial activity, and therefore represent logical starting points for future medicinal chemistry studies of the natural products and their analogs.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Methicillin-Resistant Staphylococcus aureus/chemistry , Anti-Bacterial Agents/chemical synthesis , Chrysophyta/chemistry , Geography , Macrocyclic Compounds/chemical synthesis , Microbial Sensitivity Tests/methods , United States Virgin Islands
19.
Tetrahedron ; 66(31): 5852-5862, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20798897

ABSTRACT

Functionalized bicyclo[3.3.1]non-3-en-2-ones are obtained from commercially available phenols by a hypervalent iodine oxidation, enone epoxidation, epoxide thiolysis, and intramolecular aldol reaction sequence. Reaction optimization studies identified room temperature as well as microwave-mediated procedures, providing moderate to good yields (57%-88%) in the thiophenol-mediated epoxide opening and intramolecular aldol reaction. In addition, the isolation of a key intermediate and in situ NMR studies supported the mechanistic hypothesis. The bicyclic ring products occupy novel chemical space according to ChemGPS and Chemaxon chemical diversity and cheminformatics analyses.

20.
PLoS One ; 5(6): e11378, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20614028

ABSTRACT

The botulinum neurotoxin serotype A light chain (BoNT/A LC) protease is the catalytic component responsible for the neuroparalysis that is characteristic of the disease state botulism. Three related peptide-like molecules (PLMs) were designed using previous information from co-crystal structures, synthesized, and assayed for in vitro inhibition against BoNT/A LC. Our results indicate these PLMS are competitive inhibitors of the BoNT/A LC protease and their K(i) values are in the nM-range. A co-crystal structure for one of these inhibitors was determined and reveals that the PLM, in accord with the goals of our design strategy, simultaneously involves both ionic interactions via its P1 residue and hydrophobic contacts by means of an aromatic group in the P2' position. The PLM adopts a helical conformation similar to previously determined co-crystal structures of PLMs, although there are also major differences to these other structures such as contacts with specific BoNT/A LC residues. Our structure further demonstrates the remarkable plasticity of the substrate binding cleft of the BoNT/A LC protease and provides a paradigm for iterative structure-based design and development of BoNT/A LC inhibitors.


Subject(s)
Botulinum Toxins, Type A/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Peptides/pharmacology , Catalysis , Crystallization , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Models, Molecular , Peptides/chemistry , Protein Conformation , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...